1
|
Grunwell JR, Fitzpatrick AM. Asthma Phenotypes and Biomarkers. Respir Care 2025. [PMID: 40013975 DOI: 10.1089/respcare.12352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Asthma experienced by both adults and children is a phenotypically heterogeneous condition. Severe asthma, characterized by ongoing symptoms and airway inflammation despite high doses of inhaled and/or systemic corticosteroids, is the focus of research efforts to understand this underlying heterogeneity. Clinical phenotypes in both adult and pediatric asthma have been determined using supervised definition-driven classification and unsupervised data-driven clustering methods. Efforts to understand the underlying inflammatory patterns of severe asthma have led to the seminal discovery of type 2-high versus type 2-low phenotypes and to the development of biologics targeted at type 2-high inflammation to reduce the rates of severe asthma exacerbations. Type 2-high asthma is characterized by upregulation of T helper 2 immune pathways including interleukin (IL)-4, IL-5, and IL-13 along with eosinophilic airway inflammation, sometimes allergic sensitization, and responsiveness to treatment with corticosteroids. Type 2-low asthma is poorly responsive to corticosteroids and is not as well characterized as type 2-high asthma. Type 2-low asthma is limited by being defined as the absence of type 2-high inflammatory markers. Choosing a biologic for the treatment of severe asthma involves the evaluation of a panel of biomarkers such as blood eosinophils, total and specific immunoglobulin E/allergic sensitization, and fractional exhaled nitric oxide. In this review, we focus on the underlying pathobiology of adult and pediatric asthma, discuss the different phenotype-based treatment options for adult and pediatric type 2-high with or without allergic asthma and type 2-low asthma, and describe a clinical phenotyping approach to patients to guide out-patient therapy. Finally, we end with a discussion of whether pediatric asthma exacerbations necessitating admission to an ICU constitute their own high-risk phenotype and/or whether it is a part of other previously defined high-risk subgroups such as difficult-to-control asthma, exacerbation-prone asthma, and severe treatment-resistant asthma.
Collapse
Affiliation(s)
- Jocelyn R Grunwell
- Dr. Grunwell is affiliated with Division of Critical Care Medicine, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| | - Anne M Fitzpatrick
- Dr. Fitzpatrick is affiliated with Division of Pulmonary, Allergy/Immunology, Cystic Fibrosis, and Sleep Medicine, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
2
|
Pasha MA, Hopp RJ, Habib N, Tang DD. Biomarkers in asthma, potential for therapeutic intervention. J Asthma 2024; 61:1376-1391. [PMID: 38805392 DOI: 10.1080/02770903.2024.2361783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/09/2024] [Accepted: 05/26/2024] [Indexed: 05/30/2024]
Abstract
Asthma is a heterogeneous disease characterized by multiple phenotypes with varying risk factors and therapeutic responses. This Commentary describes research on biomarkers for T2-"high" and T2-"low" inflammation, a hallmark of the disease. Patients with asthma who exhibit an increase in airway T2 inflammation are classified as having T2-high asthma. In this endotype, Type 2 cytokines interleukins (IL)-4, IL-5, and IL-13, plus other inflammatory mediators, lead to increased eosinophilic inflammation and elevated fractional exhaled nitric oxide (FeNO). In contrast, T2-low asthma has no clear definition. Biomarkers are considered valuable tools as they can help identify various phenotypes and endotypes, as well as treatment response to standard treatment or potential therapeutic targets, particularly for biologics. As our knowledge of phenotypes and endotypes expands, biologics are increasingly integrated into treatment strategies for severe asthma. These treatments block specific inflammatory pathways or single mediators. While single or composite biomarkers may help to identify subsets of patients who might benefit from these treatments, only a few inflammatory biomarkers have been validated for clinical application. One example is sputum eosinophilia, a particularly useful biomarker, as it may suggest corticosteroid responsiveness or reflect non-compliance to inhaled corticosteroids. As knowledge develops, a meaningful goal would be to provide individualized care to patients with asthma.
Collapse
Affiliation(s)
- M Asghar Pasha
- Department of Medicine, Division of Allergy and Immunology, Albany Medical College, Albany, NY, USA
| | - Russell J Hopp
- Department of Pediatrics, University of NE Medical Center and Children's Hospital and Medical Center, Omaha, NE, USA
| | - Nazia Habib
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Dale D Tang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
3
|
Xie C, Yang J, Gul A, Li Y, Zhang R, Yalikun M, Lv X, Lin Y, Luo Q, Gao H. Immunologic aspects of asthma: from molecular mechanisms to disease pathophysiology and clinical translation. Front Immunol 2024; 15:1478624. [PMID: 39439788 PMCID: PMC11494396 DOI: 10.3389/fimmu.2024.1478624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
In the present review, we focused on recent translational and clinical discoveries in asthma immunology, facilitating phenotyping and stratified or personalized interventions for patients with this condition. The immune processes behind chronic inflammation in asthma exhibit marked heterogeneity, with diverse phenotypes defining discernible features and endotypes illuminating the underlying molecular mechanisms. In particular, two primary endotypes of asthma have been identified: "type 2-high," characterized by increased eosinophil levels in the airways and sputum of patients, and "type 2-low," distinguished by increased neutrophils or a pauci-granulocytic profile. Our review encompasses significant advances in both innate and adaptive immunities, with emphasis on the key cellular and molecular mediators, and delves into innovative biological and targeted therapies for all the asthma endotypes. Recognizing that the immunopathology of asthma is dynamic and continuous, exhibiting spatial and temporal variabilities, is the central theme of this review. This complexity is underscored through the innumerable interactions involved, rather than being driven by a single predominant factor. Integrated efforts to improve our understanding of the pathophysiological characteristics of asthma indicate a trend toward an approach based on disease biology, encompassing the combined examination of the clinical, cellular, and molecular dimensions of the disease to more accurately correlate clinical traits with specific disease mechanisms.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Jingyan Yang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Aman Gul
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
- Department of Respiratory Medicine, Uyghur Medicines Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China
- College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Yifan Li
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Maimaititusun Yalikun
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiaotong Lv
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhan Lin
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qingli Luo
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Huijuan Gao
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
4
|
Rupani H, Busse WW, Howarth PH, Bardin PG, Adcock IM, Konno S, Jackson DJ. Therapeutic relevance of eosinophilic inflammation and airway viral interactions in severe asthma. Allergy 2024; 79:2589-2604. [PMID: 39087443 DOI: 10.1111/all.16242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/21/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024]
Abstract
The role of eosinophils in airway inflammation and asthma pathogenesis is well established, with raised eosinophil counts in blood and sputum associated with increased disease severity and risk of asthma exacerbation. Conversely, there is also preliminary evidence suggesting antiviral properties of eosinophils in the airways. These dual roles for eosinophils are particularly pertinent as respiratory virus infections contribute to asthma exacerbations. Biologic therapies targeting key molecules implicated in eosinophil-associated pathologies have been approved in patients with severe asthma and, therefore, the effects of depleting eosinophils in a clinical setting are of considerable interest. This review discusses the pathological and antiviral roles of eosinophils in asthma and exacerbations. We also highlight the significant reduction in asthma exacerbations seen with biologic therapies, even at the height of the respiratory virus season. Furthermore, we discuss the implications of these findings in relation to the role of eosinophils in inflammation and antiviral responses to respiratory virus infection in asthma.
Collapse
Affiliation(s)
- Hitasha Rupani
- Department of Respiratory Medicine, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
| | - William W Busse
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Peter H Howarth
- Global Medical, Global Specialty and Primary Care, GSK, Brentford, Middlesex, UK
| | - Philip G Bardin
- Monash Lung Sleep Allergy and Immunology, Monash University and Medical Centre and Hudson Institute, Melbourne, Victoria, Australia
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - David J Jackson
- Guy's Severe Asthma Centre, Guy's and St Thomas' Hospitals, London, UK
- School of Immunology and Microbial Sciences, King's College London, London, UK
| |
Collapse
|
5
|
Teague WG, Griffiths CD, Boyd K, Kellams SC, Lawrence M, Offerle TL, Heymann P, Brand W, Greenwell A, Middleton J, Wavell K, Payne J, Spano M, Etter E, Wall B, Borish L. A novel syndrome of silent rhinovirus-associated bronchoalveolitis in children with recurrent wheeze. J Allergy Clin Immunol 2024; 154:571-579.e6. [PMID: 38761997 DOI: 10.1016/j.jaci.2024.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 04/01/2024] [Accepted: 04/19/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Rhinovirus (RV) infections trigger wheeze episodes in children. Thus, understanding of the lung inflammatory response to RV in children with wheeze is important. OBJECTIVES This study sought to examine the associations of RV on bronchoalveolar lavage (BAL) granulocyte patterns and biomarkers of inflammation with age in children with treatment-refractory, recurrent wheeze (n = 616). METHODS Children underwent BAL to examine viral nucleic acid sequences, bacterial cultures, granulocyte counts, and phlebotomy for both general and type-2 inflammatory markers. RESULTS Despite the absence of cold symptoms, RV was the most common pathogen detected (30%), and when present, was accompanied by BAL granulocytosis in 75% of children. Compared to children with no BAL pathogens (n = 341), those with RV alone (n = 127) had greater (P < .05) isolated neutrophilia (43% vs 16%), mixed eosinophils and neutrophils (26% vs 11%), and less pauci-granulocytic (27% vs 61%) BAL. Children with RV alone furthermore had biomarkers of active infection with higher total blood neutrophils and serum C-reactive protein, but no differences in blood eosinophils or total IgE. With advancing age, the log odds of BAL RV alone were lower, 0.82 (5th-95th percentile CI: 0.76-0.88; P < .001), but higher, 1.58 (5th-95th percentile CI: 1.01-2.51; P = .04), with high-dose daily corticosteroid treatment. CONCLUSIONS Children with severe recurrent wheeze often (22%) have a silent syndrome of lung RV infection with granulocytic bronchoalveolitis and elevated systemic markers of inflammation. The syndrome is less prevalent by school age and is not informed by markers of type-2 inflammation. The investigators speculate that dysregulated mucosal innate antiviral immunity is a responsible mechanism.
Collapse
Affiliation(s)
- W Gerald Teague
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va; Beirne Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, Va; Division of Respiratory Medicine, Allergy, Immunology, and Sleep, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va.
| | - Cameron D Griffiths
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Va
| | - Kelly Boyd
- Division of Asthma, Allergy, and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Va
| | - Stella C Kellams
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va
| | - Monica Lawrence
- Division of Asthma, Allergy, and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Va
| | - Thomas L Offerle
- Division of Asthma, Allergy, and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Va
| | - Peter Heymann
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va; Division of Respiratory Medicine, Allergy, Immunology, and Sleep, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va
| | - William Brand
- Pediatric Otolaryngology, Department of Otolaryngology-Head and Neck Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Ariana Greenwell
- Pediatric Otolaryngology, Department of Otolaryngology-Head and Neck Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Jeremy Middleton
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va
| | - Kristin Wavell
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va; Division of Respiratory Medicine, Allergy, Immunology, and Sleep, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va
| | - Jacqueline Payne
- Division of Respiratory Medicine, Allergy, Immunology, and Sleep, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va
| | - Marthajoy Spano
- Beirne Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, Va; Division of Asthma, Allergy, and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Va
| | - Elaine Etter
- Division of Asthma, Allergy, and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Va
| | - Brittany Wall
- Division of Respiratory Medicine, Allergy, Immunology, and Sleep, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va
| | - Larry Borish
- Beirne Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, Va; Division of Asthma, Allergy, and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Va; Department of Microbiology, University of Virginia School of Medicine, Charlottesville, Va
| |
Collapse
|
6
|
Gay ACA, Banchero M, Carpaij O, Kole TM, Apperloo L, van Gosliga D, Fajar PA, Koppelman GH, Bont L, Hendriks RW, van den Berge M, Nawijn MC. Airway epithelial cell response to RSV is mostly impaired in goblet and multiciliated cells in asthma. Thorax 2024; 79:811-821. [PMID: 38373824 PMCID: PMC11347251 DOI: 10.1136/thorax-2023-220230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 11/27/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND In patients with asthma, respiratory syncytial virus (RSV) infections can cause disease exacerbation by infecting the epithelial layer of the airways, inducing subsequent immune response. The type I interferon antiviral response of epithelial cells upon RSV infection is found to be reduced in asthma in most-but not all-studies. Moreover, the molecular mechanisms causing the differences in the asthmatic bronchial epithelium in response to viral infection are poorly understood. METHODS Here, we investigated the transcriptional response to RSV infection of primary bronchial epithelial cells (pBECs) from patients with asthma (n=8) and healthy donors (n=8). The pBECs obtained from bronchial brushes were differentiated in air-liquid interface conditions and infected with RSV. After 3 days, cells were processed for single-cell RNA sequencing. RESULTS A strong antiviral response to RSV was observed for all cell types, for all samples (p<1e-48). Most (1045) differentially regulated genes following RSV infection were found in cells transitioning to secretory cells. Goblet cells from patients with asthma showed lower expression of genes involved in the interferon response (false discovery rate <0.05), including OASL, ICAM1 and TNFAIP3. In multiciliated cells, an impairment of the signalling pathways involved in the response to RSV in asthma was observed. CONCLUSION Our results highlight that the response to RSV infection of the bronchial epithelium in asthma and healthy airways was largely similar. However, in asthma, the response of goblet and multiciliated cells is impaired, highlighting the need for studying airway epithelial cells at high resolution in the context of asthma exacerbation.
Collapse
Affiliation(s)
- Aurore C A Gay
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Martin Banchero
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Orestes Carpaij
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tessa M Kole
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Leonie Apperloo
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Djoke van Gosliga
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Putri Ayu Fajar
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Gerard H Koppelman
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Louis Bont
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Division of Infectious Diseases, Department of Pediatrics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maarten van den Berge
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Martijn C Nawijn
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
7
|
Dong J, Su D, Zhao B, Han J, Tu M, Zhang K, Wang F, An Y. Potential Protective Factors for Allergic Rhinitis Patients Infected with COVID-19. Curr Issues Mol Biol 2024; 46:6633-6645. [PMID: 39057037 PMCID: PMC11275266 DOI: 10.3390/cimb46070395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
At the beginning of the 2019 coronavirus disease (COVID-19) pandemic, airway allergic diseases such as asthma and allergic rhinitis (AR) were considered as risk factors for COVID-19, as they would aggravate symptoms. With further research, more and more literature has shown that airway allergic disease may not be a high-risk factor, but may be a protective factor for COVID-19 infection, which is closely related to its low-level expression of the ACE2 receptor and the complex cytokines network as underlying molecular regulatory mechanisms. In addition, steroid hormones and age factors could not be ignored. In this review, we have summarized some current evidence on the relationship between COVID-19 and allergic rhinitis to highlight the underlying mechanisms of COVID-19 infection and provide novel insights for its prevention and treatment. The key findings show that allergic rhinitis and its related molecular mechanisms may have a protective effect against COVID-19 infection.
Collapse
Affiliation(s)
- Jiaoyue Dong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Dingyuan Su
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Kaifeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Fengling Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| |
Collapse
|
8
|
Sverrild A, Cerps S, Nieto-Fontarigo JJ, Ramu S, Hvidtfeldt M, Menzel M, Kearley J, Griffiths JM, Parnes JR, Porsbjerg C, Uller L. Tezepelumab decreases airway epithelial IL-33 and T2-inflammation in response to viral stimulation in patients with asthma. Allergy 2024; 79:656-666. [PMID: 37846599 DOI: 10.1111/all.15918] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/04/2023] [Accepted: 09/21/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Respiratory virus infections are main triggers of asthma exacerbations. Tezepelumab, an anti-TSLP mAb, reduces exacerbations in patients with asthma, but the effect of blocking TSLP on host epithelial resistance and tolerance to virus infection is not known. AIM To examine effects of blocking TSLP in patients with asthma on host resistance (IFNβ, IFNλ, and viral load) and on the airway epithelial inflammatory response to viral challenge. METHODS Bronchoalveolar lavage fluid (BALF, n = 39) and bronchial epithelial cells (BECs) were obtained from patients with uncontrolled asthma before and after 12 weeks of tezepelumab treatment (n = 13) or placebo (n = 13). BECs were cultured in vitro and exposed to the viral infection mimic poly(I:C) or infected by rhinovirus (RV). Alarmins, T2- and pro-inflammatory cytokines, IFNβ IFNλ, and viral load were analyzed by RT-qPCR and multiplex ELISA before and after stimulation. RESULTS IL-33 expression in unstimulated BECs and IL-33 protein levels in BALF were reduced after 12 weeks of tezepelumab. Further, IL-33 gene and protein levels decreased in BECs challenged with poly(I:C) after tezepelumab whereas TSLP gene expression remained unaffected. Poly(I:C)-induced IL-4, IL-13, and IL-17A release from BECs was also reduced with tezepelumab whereas IFNβ and IFNλ expression and viral load were unchanged. CONCLUSION Blocking TSLP with tezepelumab in vivo in asthma reduced the airway epithelial inflammatory response including IL-33 and T2 cytokines to viral challenge without affecting anti-viral host resistance. Our results suggest that blocking TSLP stabilizes the bronchial epithelial immune response to respiratory viruses.
Collapse
Affiliation(s)
- A Sverrild
- Department of Respiratory Medicine, University Hospital Bispebjerg, Copenhagen, Denmark
| | - S Cerps
- Department of Experimental Medicine, Lund University, Lund, Sweden
| | - J J Nieto-Fontarigo
- Department of Experimental Medicine, Lund University, Lund, Sweden
- BioLympho Research group, Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Translational Research in Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - S Ramu
- Department of Experimental Medicine, Lund University, Lund, Sweden
| | - M Hvidtfeldt
- Department of Respiratory Medicine, University Hospital Bispebjerg, Copenhagen, Denmark
| | - M Menzel
- Department of Experimental Medicine, Lund University, Lund, Sweden
| | - J Kearley
- Bioscience, Research & Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - J M Griffiths
- Translational Science and Experimental Medicine, Research & Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - J R Parnes
- Translational Medicine, Amgen, Thousand Oaks, California, USA
| | - C Porsbjerg
- Department of Respiratory Medicine, University Hospital Bispebjerg, Copenhagen, Denmark
| | - L Uller
- Department of Experimental Medicine, Lund University, Lund, Sweden
| |
Collapse
|
9
|
Papadopoulos NG, Apostolidou E, Miligkos M, Xepapadaki P. Bacteria and viruses and their role in the preschool wheeze to asthma transition. Pediatr Allergy Immunol 2024; 35:e14098. [PMID: 38445451 DOI: 10.1111/pai.14098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 03/07/2024]
Abstract
Wheezing is the cardinal symptom of asthma; its presence early in life, mostly caused by viral infections, is a major risk factor for the establishment of persistent or recurrent disease. Early-life wheezing and asthma exacerbations are triggered by common respiratory viruses, mainly rhinoviruses (RV), and to a lesser extent, respiratory syncytial virus, parainfluenza, human metapneumovirus, coronaviruses, adenoviruses, influenza, and bocavirus. The excess presence of bacteria, several of which are part of the microbiome, has also been identified in association with wheezing and acute asthma exacerbations, including haemophilus influenza, streptococcus pneumoniae, moraxella catarrhalis, mycoplasma pneumoniae, and chlamydophila pneumonia. While it is not clear when asthma starts, its characteristics develop over time. Airway remodeling already appears between the ages of 1 and 3 years of age even prior to the presence of atopic inflammation or an asthma diagnosis. The role of genetic defect or variations hampering the airway epithelium in response to environmental stimuli and severe disease morbidity are now considered as major determinants for early structural changes. Repeated viral infections can induce and perpetuate airway hyperresponsiveness. Allergic sensitization, that often precedes infection-induced wheezing, shifts inflammation toward type-2, while common respiratory infections themselves promote type-2 inflammation. Nevertheless, most children who wheeze with viral infections during infancy and during preschool years do not develop persistent asthma. Multiple factors, including illness severity, viral etiology, allergic sensitization, and the exposome, are associated with disease persistence. Here, we summarize current knowledge and developments in infection epidemiology of asthma in children, describing the known impact of each individual agent and mechanisms of transition from recurrent wheeze to asthma.
Collapse
Affiliation(s)
- Nikolaos G Papadopoulos
- Allergy Department, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
| | | | - Michael Miligkos
- Allergy Department, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Paraskevi Xepapadaki
- Allergy Department, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Tian K, Dangarh P, Zhang H, Hines CL, Bush A, Pybus HJ, Harker JA, Lloyd CM, Tanaka RJ, Saglani S. Role of epithelial barrier function in inducing type 2 immunity following early-life viral infection. Clin Exp Allergy 2024; 54:109-119. [PMID: 38011856 DOI: 10.1111/cea.14425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 10/26/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Preschool wheeze attacks triggered by recurrent viral infections, including respiratory syncytial virus (RSV), are associated with an increased risk of childhood asthma. However, mechanisms that lead to asthma following early-life viral wheezing remain uncertain. METHODS To investigate a causal relationship between early-life RSV infections and onset of type 2 immunity, we developed a neonatal murine model of recurrent RSV infection, in vivo and in silico, and evaluated the dynamical changes of altered airway barrier function and downstream immune responses, including eosinophilia, mucus secretion and type 2 immunity. RESULTS RSV infection of neonatal BALB/c mice at 5 and 15 days of age induced robust airway eosinophilia, increased pulmonary CD4+ IL-13+ and CD4+ IL-5+ cells, elevated levels of IL-13 and IL-5 and increased airway mucus at 20 days of age. Increased bronchoalveolar lavage albumin levels, suggesting epithelial barrier damage, were present and persisted following the second RSV infection. Computational in silico simulations demonstrated that recurrent RSV infection resulted in severe damage of the airway barrier (epithelium), triggering the onset of type 2 immunity. The in silico results also demonstrated that recurrent infection is not always necessary for the development of type 2 immunity, which could also be triggered with single infection of high viral load or when the epithelial barrier repair is compromised. CONCLUSIONS The neonatal murine model demonstrated that recurrent RSV infection in early life alters airway barrier function and promotes type 2 immunity. A causal relationship between airway barrier function and type 2 immunity was suggested using in silico model simulations.
Collapse
Affiliation(s)
- Kunyuan Tian
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Prakrati Dangarh
- Department of Bioengineering, Imperial College London, London, UK
| | - Haina Zhang
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Andrew Bush
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Respiratory Paediatrics, Royal Brompton Hospital, London, UK
| | - Hannah J Pybus
- Department of Bioengineering, Imperial College London, London, UK
| | - James A Harker
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Reiko J Tanaka
- Department of Bioengineering, Imperial College London, London, UK
| | - Sejal Saglani
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Respiratory Paediatrics, Royal Brompton Hospital, London, UK
| |
Collapse
|
11
|
Assaf S, Stenberg H, Jesenak M, Tarasevych SP, Hanania NA, Diamant Z. Asthma in the era of COVID-19. Respir Med 2023; 218:107373. [PMID: 37567514 DOI: 10.1016/j.rmed.2023.107373] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023]
Abstract
Since its global invasion in 2019, COVID-19 has affected several aspects of patients' lives and posed a significant impact on the health care system. Several patient populations were identified to be at high risk of contracting SARS-CoV-2 infection and/or developing severe COVID-19-related sequelae. Conversely, anyone who has contracted SARS-CoV-2 is at risk to experience symptoms and signs consistent with post-COVID manifestations. Patients with asthma were initially thought to be at increased risk and severity for SARS-CoV-2 infection. However, accumulating evidence demonstrates that asthma endotypes/phenotypes and comorbidities influence the risk stratification in this population. Furthermore, initial concerns about the potentially increased risk of poor outcomes with asthma treatments such as inhaled corticosteroids and biologics have not been substantiated. In this review, we provide an update on COVID-19 and asthma, including risk of susceptibility, clinical manifestations and course in this population as well as discuss recommendations for management.
Collapse
Affiliation(s)
- Sara Assaf
- Section of Pulmonary and Critical Care Medicine, University of New Mexico, Albuquerque, NM, USA.
| | - Henning Stenberg
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Milos Jesenak
- Departments of Pulmonology and Phthisiology and Paediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Department of Clinical Immunology and Allergology, University Teaching Hospital in Martin, Martin, Slovak Republic
| | | | - Nicola A Hanania
- Section of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Zuzana Diamant
- Dept of Microbiology Immunology & Transplantation, KU Leuven, Catholic University of Leuven, Belgium; Dept 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; Dept Clin Pharm & Pharmacol, Univ Groningen, Univ Med Ctr Groningen, Groningen, Netherlands.
| |
Collapse
|
12
|
Medeleanu MV, Qian YC, Moraes TJ, Subbarao P. Early-immune development in asthma: A review of the literature. Cell Immunol 2023; 393-394:104770. [PMID: 37837916 DOI: 10.1016/j.cellimm.2023.104770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/16/2023]
Abstract
This review presents a comprehensive examination of the various factors contributing to the immunopathogenesis of asthma from the prenatal to preschool period. We focus on the contributions of genetic and environmental components as well as the role of the nasal and gut microbiome on immune development. Predisposing genetic factors, including inherited genes associated with increased susceptibility to asthma, are discussed alongside environmental factors such as respiratory viruses and pollutant exposure, which can trigger or exacerbate asthma symptoms. Furthermore, the intricate interplay between the nasal and gut microbiome and the immune system is explored, emphasizing their influence on allergic immune development and response to environmental stimuli. This body of literature underscores the necessity of a comprehensive approach to comprehend and manage asthma, as it emphasizes the interactions of multiple factors in immune development and disease progression.
Collapse
Affiliation(s)
- Maria V Medeleanu
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Canada; Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada
| | - Yu Chen Qian
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Canada; Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada
| | - Theo J Moraes
- Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada; Laboratory Medicine and Pathology, Temerty Faculty of Medicine, University of Toronto, Canada; Department of Paediatrics, Temerty Faculty of Medicine, University of Toronto, Canada; Division of Respiratory Medicine, Hospital for Sick Children, Canada
| | - Padmaja Subbarao
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Canada; Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Canada; Department of Paediatrics, Temerty Faculty of Medicine, University of Toronto, Canada; Division of Respiratory Medicine, Hospital for Sick Children, Canada; Epidemiology Division, Dalla Lana School of Public Health, University of Toronto, Canada.
| |
Collapse
|
13
|
Lloyd CM, Saglani S. Early-life respiratory infections and developmental immunity determine lifelong lung health. Nat Immunol 2023; 24:1234-1243. [PMID: 37414905 DOI: 10.1038/s41590-023-01550-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023]
Abstract
Respiratory infections are common in infants and young children. However, the immune system develops and matures as the child grows, thus the effects of infection during this time of dynamic change may have long-term consequences. The infant immune system develops in conjunction with the seeding of the microbiome at the respiratory mucosal surface, at a time that the lungs themselves are maturing. We are now recognizing that any disturbance of this developmental trajectory can have implications for lifelong lung health. Here, we outline our current understanding of the molecular mechanisms underlying relationships between immune and structural cells in the lung with the local microorganisms. We highlight the importance of gaining greater clarity as to what constitutes a healthy respiratory ecosystem and how environmental exposures influencing this network will aid efforts to mitigate harmful effects and restore lung immune health.
Collapse
Affiliation(s)
- Clare M Lloyd
- National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK.
| | - Sejal Saglani
- National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, UK.
| |
Collapse
|
14
|
Veerati PC, Reid AT, Nichol KS, Wark PAB, Knight DA, Bartlett NW, Grainge CL. Mechanical forces suppress antiviral innate immune responses from asthmatic airway epithelial cells following rhinovirus infection. Am J Physiol Lung Cell Mol Physiol 2023; 325:L206-L214. [PMID: 37280545 PMCID: PMC10396277 DOI: 10.1152/ajplung.00074.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2023] [Accepted: 05/25/2023] [Indexed: 06/08/2023] Open
Abstract
Bronchoconstriction is the main physiological event in asthma, which leads to worsened clinical symptoms and generates mechanical stress within the airways. Virus infection is the primary cause of exacerbations in people with asthma, however, the impact that bronchoconstriction itself on host antiviral responses and viral replication is currently not well understood. Here we demonstrate how mechanical forces generated during bronchoconstriction may suppress antiviral responses at the airway epithelium without any difference in viral replication. Primary bronchial epithelial cells from donors with asthma were differentiated at the air-liquid interface. Differentiated cells were apically compressed (30 cmH2O) for 10 min every hour for 4 days to mimic bronchoconstriction. Two asthma disease models were developed with the application of compression, either before ("poor asthma control model," n = 7) or following ("exacerbation model," n = 4) rhinovirus (RV) infection. Samples were collected at 0, 24, 48, 72, and 96 h postinfection (hpi). Viral RNA, interferon (IFN)-β, IFN-λ, and host defense antiviral peptide gene expressions were measured along with IFN-β, IFN-λ, TGF-β2, interleukin-6 (IL-6), and IL-8 protein expression. Apical compression significantly suppressed RV-induced IFN-β protein from 48 hpi and IFN-λ from 72 hpi in the poor asthma control model. There was a nonsignificant reduction of both IFN-β and IFN-λ proteins from 48 hpi in the exacerbation model. Despite reductions in antiviral proteins, there was no significant change in viral replication in either model. Compressive stress mimicking bronchoconstriction inhibits antiviral innate immune responses from asthmatic airway epithelial cells when applied before RV infection.NEW & NOTEWORTHY Bronchoconstriction is the main physiological event in asthma, which leads to worsened clinical symptoms and generates mechanical stress within the airways. Virus infection is the primary cause of exacerbations in people with asthma, however, the impact of bronchoconstriction on host antiviral responses and viral replication is unknown. We developed two disease models, in vitro, and found suppressed IFN response from cells following the application of compression and RV-A1 infection. This explains why people with asthma have deficient IFN response.
Collapse
Affiliation(s)
- Punnam Chander Veerati
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Asthma and Breathing Research Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Andrew T Reid
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Asthma and Breathing Research Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Kristy S Nichol
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Peter A B Wark
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
- Research and Academic Affairs, Providence Health Care Research Institute, Vancouver, British Columbia, Canada
| | - Nathan W Bartlett
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Christopher L Grainge
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Asthma and Breathing Research Program, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
15
|
Wang C, Du Z, Li R, Luo Y, Zhu C, Ding N, Lei A. Interferons as negative regulators of ILC2s in allergic lung inflammation and respiratory viral infections. J Mol Med (Berl) 2023; 101:947-959. [PMID: 37414870 DOI: 10.1007/s00109-023-02345-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s), characterized by a lack of antigen receptors, have been regarded as an important component of type 2 pulmonary immunity. Analogous to Th2 cells, ILC2s are capable of releasing type 2 cytokines and amphiregulin, thus playing an essential role in a variety of diseases, such as allergic diseases and virus-induced respiratory diseases. Interferons (IFNs), an important family of cytokines with potent antiviral effects, can be triggered by microbial products, microbial exposure, and pathogen infections. Interestingly, the past few years have witnessed encouraging progress in revealing the important role of IFNs and IFN-producing cells in modulating ILC2 responses in allergic lung inflammation and respiratory viral infections. This review underscores recent progress in understanding the role of IFNs and IFN-producing cells in shaping ILC2 responses and discusses disease phenotypes, mechanisms, and therapeutic targets in the context of allergic lung inflammation and infections with viruses, including influenza virus, rhinovirus (RV), respiratory syncytial virus (RSV), and severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Cui Wang
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Zhaoxiang Du
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Ranhui Li
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Ying Luo
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Cuiming Zhu
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Nan Ding
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China
| | - Aihua Lei
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China.
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, China.
| |
Collapse
|
16
|
Urbani F, Cometa M, Martelli C, Santoli F, Rana R, Ursitti A, Bonato M, Baraldo S, Contoli M, Papi A. Update on virus-induced asthma exacerbations. Expert Rev Clin Immunol 2023; 19:1259-1272. [PMID: 37470413 DOI: 10.1080/1744666x.2023.2239504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/01/2023] [Accepted: 07/18/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Viral infections are common triggers for asthma exacerbation. Subjects with asthma are more susceptible to viral infections and develop more severe or long-lasting lower respiratory tract symptoms than healthy individuals owing to impaired immune responses. Of the many viruses associated with asthma exacerbation, rhinovirus (RV) is the most frequently identified virus in both adults and children. AREAS COVERED We reviewed epidemiological and clinical links and mechanistic studies on virus-associated asthma exacerbations. We included sections on severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), the latest evidence of coronavirus disease 2019 (COVID-19) in asthma patients, and past and future searches for therapeutic and prevention targets. EXPERT OPINION Early treatment or prevention of viral infections might significantly reduce the rate of asthma exacerbation, which is one of the key points of disease management. Although it is hypothetically possible nowadays to interfere with every step of the infectious cycle of respiratory tract viruses, vaccination development has provided some of the most encouraging results. Future research should proceed toward the development of a wider spectrum of vaccines to achieve a better quality of life for patients with asthma and to reduce the economic burden on the healthcare system.
Collapse
Affiliation(s)
- Francesca Urbani
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Marianna Cometa
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Chiara Martelli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Federica Santoli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Roberto Rana
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Antonio Ursitti
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Matteo Bonato
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Simonetta Baraldo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Marco Contoli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Alberto Papi
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| |
Collapse
|
17
|
Radzikowska U, Eljaszewicz A, Tan G, Stocker N, Heider A, Westermann P, Steiner S, Dreher A, Wawrzyniak P, Rückert B, Rodriguez-Coira J, Zhakparov D, Huang M, Jakiela B, Sanak M, Moniuszko M, O'Mahony L, Jutel M, Kebadze T, Jackson JD, Edwards RM, Thiel V, Johnston LS, Akdis AC, Sokolowska M. Rhinovirus-induced epithelial RIG-I inflammasome suppresses antiviral immunity and promotes inflammation in asthma and COVID-19. Nat Commun 2023; 14:2329. [PMID: 37087523 PMCID: PMC10122208 DOI: 10.1038/s41467-023-37470-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/16/2023] [Indexed: 04/24/2023] Open
Abstract
Rhinoviruses and allergens, such as house dust mite are major agents responsible for asthma exacerbations. The influence of pre-existing airway inflammation on the infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is largely unknown. We analyse mechanisms of response to viral infection in experimental in vivo rhinovirus infection in healthy controls and patients with asthma, and in in vitro experiments with house dust mite, rhinovirus and SARS-CoV-2 in human primary airway epithelium. Here, we show that rhinovirus infection in patients with asthma leads to an excessive RIG-I inflammasome activation, which diminishes its accessibility for type I/III interferon responses, leading to their early functional impairment, delayed resolution, prolonged viral clearance and unresolved inflammation in vitro and in vivo. Pre-exposure to house dust mite augments this phenomenon by inflammasome priming and auxiliary inhibition of early type I/III interferon responses. Prior infection with rhinovirus followed by SARS-CoV-2 infection augments RIG-I inflammasome activation and epithelial inflammation. Timely inhibition of the epithelial RIG-I inflammasome may lead to more efficient viral clearance and lower the burden of rhinovirus and SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Herman-Burchard-Strasse 1, 7265, Davos Wolfgang, Switzerland
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Waszyngtona 13 Str., 15-269, Bialystok, Poland
| | - Andrzej Eljaszewicz
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Herman-Burchard-Strasse 1, 7265, Davos Wolfgang, Switzerland
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Waszyngtona 13 Str., 15-269, Bialystok, Poland
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Nino Stocker
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
| | - Anja Heider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
| | - Patrick Westermann
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
| | - Silvio Steiner
- Institute of Virology and Immunology (IVI), Laenggassstrasse 122, 3012, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 122, 3012, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Mittelstrasse 43, 3012, Bern, Switzerland
| | - Anita Dreher
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Herman-Burchard-Strasse 1, 7265, Davos Wolfgang, Switzerland
| | - Paulina Wawrzyniak
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Herman-Burchard-Strasse 1, 7265, Davos Wolfgang, Switzerland
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Beate Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
| | - Juan Rodriguez-Coira
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
- IMMA, Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities Madrid, C. de Julian Romea 23, 28003, Madrid, Spain
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities Madrid, Urb. Monteprincipe 28925, Alcorcon, Madrid, Spain
| | - Damir Zhakparov
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
| | - Mengting Huang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
| | - Bogdan Jakiela
- Department of Internal Medicine, Jagiellonian University Medical College, M. Skawinska 8 Str., 31-066, Krakow, Poland
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, M. Skawinska 8 Str., 31-066, Krakow, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Waszyngtona 13 Str., 15-269, Bialystok, Poland
- Department of Allergology and Internal Medicine, Medical University of Bialystok, M. Sklodowskiej-Curie 24A Str., 15-276, Bialystok, Poland
| | - Liam O'Mahony
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
- Department of Medicine and School of Microbiology, APC Microbiome Ireland, University College Cork, College Rd, T12 E138, Cork, Ireland
| | - Marek Jutel
- Department of Clinical Immunology, Wroclaw Medical University, wyb. Lidwika Pasteura 1 Str, 50-367, Wroclaw, Poland
- ALL-MED Medical Research Institute, Gen. Jozefa Hallera 95 Str., 53-201, Wroclaw, Poland
| | - Tatiana Kebadze
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, SW3 6LY, UK
- Department of Infectious Diseases, Imperial College London, School of Medicine, St Mary's Hospital, Praed Street, London, W21NY, UK
| | - J David Jackson
- Guy's Severe Asthma Centre, School of Immunology & Microbial Sciences, King's College London, Strand, London, WC2R 2LS, UK
- Guy's & St Thomas' NHS Trust, St Thomas' Hospital, Westminster Bridge Rd, London, SE1 7EH, UK
| | - R Michael Edwards
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, SW3 6LY, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, Norfolk Place, London, W2 1PG, UK
| | - Volker Thiel
- Institute of Virology and Immunology (IVI), Laenggassstrasse 122, 3012, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases, University of Bern, Hallerstrasse 6, 3012, Bern, Switzerland
| | - L Sebastian Johnston
- National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, SW3 6LY, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, Norfolk Place, London, W2 1PG, UK
- Imperial College Healthcare HNS Trust, The Bays, S Wharf Rd, London, W2 1NY, UK
| | - A Cezmi Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Herman-Burchard-Strasse 1, 7265, Davos Wolfgang, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard-Strasse 9, 7265, Davos Wolfgang, Switzerland.
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Herman-Burchard-Strasse 1, 7265, Davos Wolfgang, Switzerland.
| |
Collapse
|
18
|
Pathinayake PS, Awatade NT, Wark PAB. Type 2 Immunity and Its Impact on COVID-19 Infection in the Airways. Viruses 2023; 15:402. [PMID: 36851616 PMCID: PMC9967553 DOI: 10.3390/v15020402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/17/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Type 2 immune responses are characterized by elevated type 2 cytokines and blood eosinophilia. Emerging evidence suggests that people with chronic type 2 inflammatory lung diseases are not particularly susceptible to SARS-CoV-2 infection. Intriguingly, recent in vitro, ex vivo research demonstrates type 2 cytokines, particularly IL-13, reduce the risk of SARS-CoV-2 infection in the airway epithelium. IL-13 treatment in airway epithelial cells followed by SARS-CoV-2 diminished viral entry, replication, spread, and cell death. IL-13 reduces the expression of the angiotensin-converting enzyme 2 (ACE2) receptor in the airway epithelium and transmembrane serine protease 2 (TMPRSS2), particularly in ciliated cells. It also alters the cellular composition toward a secretory-cell-rich phenotype reducing total ciliated cells and, thus, reducing viral tropism. IL-13 enhances Muc5ac mucin and glycocalyx secretion in the periciliary layer, which acts as a physical barrier to restrict virus attachment. Moreover, type 2 airway immune cells, such as M2 alveolar macrophages, CD4+ tissue-resident memory T cells, and innate lymphoid 2 cells, may also rescue type 2 airways from SARS-CoV-2-induced adverse effects. In this review, we discuss recent findings that demonstrate how type 2 immunity alters immune responses against SARS-CoV-2 and its consequences on COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Prabuddha S. Pathinayake
- School of Medicine and Public Health, The University of Newcastle and Immune Health Program Hunter Medical Research Institute, Newcastle, NSW 2308, Australia
| | - Nikhil T. Awatade
- School of Medicine and Public Health, The University of Newcastle and Immune Health Program Hunter Medical Research Institute, Newcastle, NSW 2308, Australia
| | - Peter A. B. Wark
- School of Medicine and Public Health, The University of Newcastle and Immune Health Program Hunter Medical Research Institute, Newcastle, NSW 2308, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
19
|
Akuthota P. Asthma Exacerbations: Patient Features and Potential Long-Term Implications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:253-263. [PMID: 37464125 DOI: 10.1007/978-3-031-32259-4_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Asthma exacerbations occur in the context of a complex interplay between external exposures and host factors. Respiratory tract viral infections, in particular rhinovirus, are dominant initiators of exacerbations, with allergens and other inhalation exposures as additional key contributors. The presence of underlying type II inflammation, with associated biomarker elevations, is a major driver of exacerbation risk and mechanism, as evidenced by the consistent reduction of exacerbations seen with biologics targeting these pathways. Several genetic polymorphisms are associated with exacerbations, and while they may individually have small effects, they are cumulatively important and magnified by environmental exposures. A history of exacerbations predicts future exacerbations with potentially negative implications on long-term lung health.
Collapse
Affiliation(s)
- Praveen Akuthota
- Division of Pulmonary, Critical Care, Sleep Medicine, & Physiology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
20
|
Essaidi-Laziosi M, Royston L, Boda B, Pérez-Rodriguez FJ, Piuz I, Hulo N, Kaiser L, Clément S, Huang S, Constant S, Tapparel C. Altered cell function and increased replication of rhinoviruses and EV-D68 in airway epithelia of asthma patients. Front Microbiol 2023; 14:1106945. [PMID: 36937308 PMCID: PMC10014885 DOI: 10.3389/fmicb.2023.1106945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/18/2023] [Indexed: 03/05/2023] Open
Abstract
Introduction Rhinovirus (RV) infections constitute one of the main triggers of asthma exacerbations and an important burden in pediatric yard. However, the mechanisms underlying this association remain poorly understood. Methods In the present study, we compared infections of in vitro reconstituted airway epithelia originating from asthmatic versus healthy donors with representative strains of RV-A major group and minor groups, RV-C, RV-B, and the respiratory enterovirus EV-D68. Results We found that viral replication was higher in tissues derived from asthmatic donors for all tested viruses. Viral receptor expression was comparable in non-infected tissues from both groups. After infection, ICAM1 and LDLR were upregulated, while CDHR3 was downregulated. Overall, these variations were related to viral replication levels. The presence of the CDHR3 asthma susceptibility allele (rs6967330) was not associated with increased RV-C replication. Regarding the tissue response, a significantly higher interferon (IFN) induction was demonstrated in infected tissues derived from asthmatic donors, which excludes a defect in IFN-response. Unbiased transcriptomic comparison of asthmatic versus control tissues revealed significant modifications, such as alterations of cilia structure and motility, in both infected and non-infected tissues. These observations were supported by a reduced mucociliary clearance and increased mucus secretion in non-infected tissues from asthmatic donors. Discussion Altogether, we demonstrated an increased permissiveness and susceptibility to RV and respiratory EV infections in HAE derived from asthmatic patients, which was associated with a global alteration in epithelial cell functions. These results unveil the mechanisms underlying the pathogenesis of asthma exacerbation and suggest interesting therapeutic targets.
Collapse
Affiliation(s)
- Manel Essaidi-Laziosi
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Léna Royston
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Francisco Javier Pérez-Rodriguez
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Infectious Diseases, Geneva University Hospital, Geneva, Switzerland
| | - Isabelle Piuz
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nicolas Hulo
- Service for Biomathematical and Biostatistical Analyses, Institute of Genetics and Genomics, University of Geneva, Geneva, Switzerland
| | - Laurent Kaiser
- Division of Infectious Diseases, Geneva University Hospital, Geneva, Switzerland
| | - Sophie Clément
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Song Huang
- Epithelix Sàrl, Plan les Ouates, Geneva, Switzerland
| | | | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- *Correspondence: Caroline Tapparel,
| |
Collapse
|
21
|
Bartlett NW, Feghali-Bostwick C, Gunst SJ. Call for Papers: "Targeting Airway Immunity in Lung Disease". Am J Physiol Lung Cell Mol Physiol 2023; 324:L48-L52. [PMID: 36472349 DOI: 10.1152/ajplung.00375.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Nathan W Bartlett
- Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Carol Feghali-Bostwick
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Susan J Gunst
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
22
|
Nicholas B, Lee HH, Guo J, Cicmil M, Blume C, Malefyt RDW, Djukanović R. Immunomodulatory regulator blockade in a viral exacerbation model of severe asthma. Front Immunol 2022; 13:973673. [PMID: 36479132 PMCID: PMC9720166 DOI: 10.3389/fimmu.2022.973673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/03/2022] [Indexed: 11/22/2022] Open
Abstract
Asthmatics are more susceptible to viral infections than healthy individuals and are known to have impaired innate anti-viral defences. Influenza A virus causes significant morbidity and mortality in this population. Immuno-modulatory regulators (IMRs) such as PD-1 are activated on T cells following viral infection as part of normal T cell activation responses, and then subside, but remain elevated in cases of chronic exposure to virus, indicative of T cell exhaustion rather than activation. There is evidence that checkpoint inhibition can enhance anti-viral responses during acute exposure to virus through enhancement of CD8+T cell function. Although elevated PD-1 expression has been described in pulmonary tissues in other chronic lung diseases, the role of IMRs in asthma has been relatively unexplored as the basis for immune dysfunction. We first assessed IMR expression in the peripheral circulation and then quantified changes in IMR expression in lung tissue in response to ex-vivo influenza infection. We found that the PD-1 family members are not significantly altered in the peripheral circulation in individuals with severe asthma but are elevated in pulmonary tissues following ex-vivo influenza infection. We then applied PD-1 Mab inhibitor treatment to bronchial biopsy tissues infected with influenza virus and found that PD-1 inhibition was ineffective in asthmatics, but actually increased infection rates in healthy controls. This study, therefore, suggests that PD-1 therapy would not produce harmful side-effects when applied in people with severe asthma, but could have important, as yet undescribed, negative effects on anti-viral responses in healthy individuals that warrant further investigation.
Collapse
Affiliation(s)
- Ben Nicholas
- Division of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Hampshire, United Kingdom,*Correspondence: Ben Nicholas,
| | - Hyun-Hee Lee
- Oncology & Immunology Discovery, Merck Research Laboratories, Boston, MA, United States
| | - Jane Guo
- Oncology & Immunology Discovery, Merck Research Laboratories, Boston, MA, United States
| | - Milenko Cicmil
- Oncology & Immunology Discovery, Merck Research Laboratories, Boston, MA, United States
| | - Cornelia Blume
- Division of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Hampshire, United Kingdom
| | | | - Ratko Djukanović
- Division of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Hampshire, United Kingdom
| |
Collapse
|
23
|
Immunopathology of Differing Viral Infection in Allergic Asthma Disease. Immunol Allergy Clin North Am 2022; 42:715-726. [DOI: 10.1016/j.iac.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
24
|
Roberts G. Is it time to consider allergen immunotherapy earlier in the management of allergic asthma? Eur Respir J 2022; 60:60/5/2201686. [PMID: 36396157 DOI: 10.1183/13993003.01686-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Graham Roberts
- University of Southampton Faculty of Medicine, Southampton, UK .,Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,David Hide Asthma and Allergy Centre, St Mary's Hospital, Isle of Wight, UK
| |
Collapse
|
25
|
Woehlk C, Von Bülow A, Ghanizada M, Søndergaard MB, Hansen S, Porsbjerg C. Allergen immunotherapy effectively reduces the risk of exacerbations and lower respiratory tract infections in both seasonal and perennial allergic asthma: a nationwide epidemiological study. Eur Respir J 2022; 60:13993003.00446-2022. [PMID: 35618279 DOI: 10.1183/13993003.00446-2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/10/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Allergic asthma is associated with increased risk of respiratory tract infections and exacerbations. It remains unclear whether this susceptibility is conditioned by seasonal or by perennial allergy. AIM To investigate perennial allergy compared with seasonal allergy as a risk factor for lower respiratory tract infections and exacerbations in asthma and whether this risk can be reduced by allergen immunotherapy (AIT). METHODOLOGY This is a prospective register-based nationwide study of 18-44-year-olds treated with AIT during 1995-2014. Based on the type of AIT and use of anti-asthmatic drugs, patients were subdivided into two groups: perennial allergic asthma (PAA) versus seasonal allergic asthma (SAA). Data on antibiotics against lower respiratory tract infections (LRTI) and oral corticosteroids for exacerbations were analysed before starting AIT (baseline) and 3 years after completing AIT (follow-up). RESULTS We identified 2688 patients with asthma treated with AIT, of whom 1249 had PAA and 1439 had SAA. At baseline, patients with SAA had more exacerbations (23.8% versus 16.5%, p≤0.001), but there were no differences in LRTI. During the 3-year follow-up, we observed a highly significant reduction of exacerbations with an average decrease of 57% in PAA and 74% in SAA. In addition, we observed a significant reduction of LRTI in both PAA and SAA: 17% and 20% decrease, respectively. CONCLUSION AIT effectively reduced the risk of exacerbations and lower respiratory tract infections in both seasonal and perennial allergic asthma. Perennial allergy is seemingly not a stronger risk factor for respiratory infections and exacerbations than seasonal allergy.
Collapse
Affiliation(s)
- Christian Woehlk
- Respiratory Research Unit, Dept Respiratory Medicine, Copenhagen, Denmark
| | - Anna Von Bülow
- Respiratory Research Unit, Dept Respiratory Medicine, Copenhagen, Denmark
| | - Muzhda Ghanizada
- Respiratory Research Unit, Dept Respiratory Medicine, Copenhagen, Denmark
| | | | - Susanne Hansen
- Respiratory Research Unit, Dept Respiratory Medicine, Copenhagen, Denmark
| | - Celeste Porsbjerg
- Respiratory Research Unit, Dept Respiratory Medicine, Copenhagen, Denmark
| |
Collapse
|
26
|
Kim SR. Viral Infection and Airway Epithelial Immunity in Asthma. Int J Mol Sci 2022; 23:9914. [PMID: 36077310 PMCID: PMC9456547 DOI: 10.3390/ijms23179914] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 12/19/2022] Open
Abstract
Viral respiratory tract infections are associated with asthma development and exacerbation in children and adults. In the course of immune responses to viruses, airway epithelial cells are the initial platform of innate immunity against viral invasion. Patients with severe asthma are more vulnerable than those with mild to moderate asthma to viral infections. Furthermore, in most cases, asthmatic patients tend to produce lower levels of antiviral cytokines than healthy subjects, such as interferons produced from immune effector cells and airway epithelial cells. The epithelial inflammasome appears to contribute to asthma exacerbation through overactivation, leading to self-damage, despite its naturally protective role against infectious pathogens. Given the mixed and complex immune responses in viral-infection-induced asthma exacerbation, this review examines the diverse roles of airway epithelial immunity and related potential therapeutic targets and discusses the mechanisms underlying the heterogeneous manifestations of asthma exacerbations.
Collapse
Affiliation(s)
- So Ri Kim
- Division of Respiratory Medicine and Allergy, Department of Internal Medicine, Medical School of Jeonbuk National University, 20 Geonji-ro, Deokjin-gu, Jeonju 54907, Korea
| |
Collapse
|
27
|
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: 5] [Impact Index Per Article: 1.7] [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
|
28
|
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
|
29
|
Shrestha P, Wi CI, Liu H, King KS, Ryu E, Kwon JH, Sohn S, Park M, Juhn Y. Risk of pneumonia in asthmatic children using inhaled corticosteroids: a nested case-control study in a birth cohort. BMJ Open 2022; 12:e051926. [PMID: 35273042 PMCID: PMC8915358 DOI: 10.1136/bmjopen-2021-051926] [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] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Inhaled corticosteroids (ICSs) are important in asthma management, but there are concerns regarding associated risk of pneumonia. While studies in asthmatic adults have shown inconsistent results, this risk in asthmatic children is unclear. OBJECTIVE Our aim was to determine the association of ICS use with pneumonia risk in asthmatic children. METHODS A nested case-control study was performed in the Mayo Clinic Birth Cohort. Asthmatic children (<18 years) with a physician diagnosis of asthma were identified from electronic medical records of children born at Mayo Clinic from 1997 to 2016 and followed until 31 December 2017. Pneumonia cases defined by Infectious Disease Society of America were 1:1 matched with controls without pneumonia by age, sex and asthma index date. Exposure was defined as ICS prescription at least 90 days prior to pneumonia. Associations of ICS use, type and dose (low, medium and high) with pneumonia risk were analysed using conditional logistic regression. RESULTS Of the 2108 asthmatic children eligible for the study (70% mild intermittent and 30% persistent asthma), 312 children developed pneumonia during the study period. ICS use overall was not associated with risk of pneumonia (adjusted OR: 0.94, 95% CI: 0.62 to 1.41). Poorly controlled asthma was significantly associated with the risk of pneumonia (OR: 2.03, 95% CI: 1.35 to 3.05; p<0.001). No ICS type or dose was associated with risk of pneumonia. CONCLUSION ICS use in asthmatic children was not associated with risk of pneumonia but poorly controlled asthma was. Future asthma studies may need to include pneumonia as a potential outcome of asthma management.
Collapse
Affiliation(s)
- Pragya Shrestha
- Precision Population Science Lab, Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Chung-Il Wi
- Precision Population Science Lab, Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Hongfang Liu
- Artificial Intelligence and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Katherine S King
- Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | - Euijung Ryu
- Computational Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jung Hyun Kwon
- Precision Population Science Lab, Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Pediatrics, Korea University Medical Center, Seoul, Republic of Korea
| | - Sunghwan Sohn
- Artificial Intelligence and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Miguel Park
- Division of Allergic Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Young Juhn
- Precision Population Science Lab, Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
30
|
Makrinioti H, Bush A, Gern J, Johnston SL, Papadopoulos N, Feleszko W, Camargo CA, Hasegawa K, Jartti T. The Role of Interferons in Driving Susceptibility to Asthma Following Bronchiolitis: Controversies and Research Gaps. Front Immunol 2021; 12:761660. [PMID: 34925333 PMCID: PMC8677668 DOI: 10.3389/fimmu.2021.761660] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022] Open
Abstract
Bronchiolitis is the most common cause of hospitalization in infancy and is associated with a higher risk for the development of childhood asthma. However, not all children hospitalized with bronchiolitis will develop asthma. The mechanisms underlying asthma development following bronchiolitis hospitalization are complex. Immune responses to respiratory viruses may underlie both bronchiolitis severity and long-term sequela (such as asthma). Interferons (IFNs) are important components of innate immune responses to respiratory viruses and could influence both asthma development and asthma exacerbations. However, the nature of the relationship between interferon production and wheezing illnesses is controversial. For example, low peripheral blood IFN responses at birth have been linked with recurrent wheeze and asthma development. In contrast, there is evidence that severe illnesses (e.g., hospitalization for bronchiolitis) are associated with increased IFN responses during acute infection (bronchiolitis hospitalization) and a higher risk for subsequent asthma diagnosis. Furthermore, mechanistic studies suggest that bronchial epithelial cells from asthmatic children have impaired IFN responses to respiratory viruses, which may enable increased viral replication followed by exaggerated secondary IFN responses. This review aims to discuss controversies around the role of IFNs as drivers of susceptibility to asthma development following bronchiolitis hospitalization. Past evidence from both mechanistic and cohort studies are discussed. We will highlight knowledge gaps that can inform future research study design.
Collapse
Affiliation(s)
- Heidi Makrinioti
- West Middlesex University Hospital, Chelsea, and Westminster Foundation Trust, London, United Kingdom.,Imperial Centre for Paediatrics and Child Health, Imperial College, London, United Kingdom
| | - Andrew Bush
- Imperial Centre for Paediatrics and Child Health, Imperial College, London, United Kingdom.,National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - James Gern
- Department of Paediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Wisconsin, WI, United States
| | | | - Nikolaos Papadopoulos
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.,Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Wojciech Feleszko
- Department of Paediatric Pneumology and Allergy, The Medical University of Warsaw, Warsaw, Poland
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Tuomas Jartti
- Department of Paediatrics, Turku University Hospital and Turku University, Turku, Finland.,Research Unit for Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology and Ophthalmology (PEDEGO), Medical Research Center, University of Oulu, Oulu, Finland.,Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
31
|
Custovic A, Siddiqui S, Saglani S. Considering biomarkers in asthma disease severity. J Allergy Clin Immunol 2021; 149:480-487. [PMID: 34942235 DOI: 10.1016/j.jaci.2021.11.021] [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: 10/05/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 10/19/2022]
Abstract
Amongst patients with asthma, reliance on the type/dose of prescribed medication and symptom control does not adequately capture those at risk of adverse outcomes, and we need biomarkers for risk and treatment stratification which are consistently accurate, readily quantifiable and reproducible. The majority of patients with severe asthma, regardless of age, have predominant type-2 (T2) inflammation mediated disease, making airway/blood eosinophils, FeNO, periostin and/or allergic sensitization potentially important biomarkers for severe disease. In both adult and pediatric asthma, there is scope to improve prediction of severe attacks by using a composite T2 biomarkers of blood eosinophils and FeNO. Technological advances in component-resolved diagnostics (CRD) microarray technologies coupled with the development of interpretation software offer a possibility to use CRD as biomarkers of asthma severity amongst sensitized asthmatics. Genetic predisposition and polygenic risk scores of relevant traits (e.g., lung function, host immune responses, biomarkers of exposure from the indoor and outdoor environment, infection and microbial dysbiosis) may also contribute to prediction algorithms. We challenge the idea that asthma can be accurately defined in an individual patient by a discrete and static "endotype" (e.g., T2-high asthma). As we traverse the new era of molecular endotyping in asthma, we need to understand how relevant mechanisms impact patient outcomes, and in parallel develop new tools and approaches to stratify therapies and define individual patient trajectories.
Collapse
Affiliation(s)
- Adnan Custovic
- National Heart and Lung Institute, Imperial College London, UK.
| | - Salman Siddiqui
- Department of Respiratory Sciences, University of Leicester and NIHR Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester UK
| | - Sejal Saglani
- National Heart and Lung Institute, Imperial College London, UK
| |
Collapse
|
32
|
Watkinson RL, Looi K, Laing IA, Cianferoni A, Kicic A. Viral Induced Effects on a Vulnerable Epithelium; Lessons Learned From Paediatric Asthma and Eosinophilic Oesophagitis. Front Immunol 2021; 12:773600. [PMID: 34912343 PMCID: PMC8666438 DOI: 10.3389/fimmu.2021.773600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/05/2021] [Indexed: 01/07/2023] Open
Abstract
The epithelium is integral to the protection of many different biological systems and for the maintenance of biochemical homeostasis. Emerging evidence suggests that particular children have epithelial vulnerabilities leading to dysregulated barrier function and integrity, that resultantly contributes to disease pathogenesis. These epithelial vulnerabilities likely develop in utero or in early life due to various genetic, epigenetic and environmental factors. Although various epithelia are uniquely structured with specific function, prevalent allergic-type epithelial diseases in children potentially have common or parallel disease processes. These include inflammation and immune response dysregulation stemming from atypical epithelial barrier function and integrity. Two diseases where aetiology and pathogenesis are potentially linked to epithelial vulnerabilities include Paediatric Asthma and Eosinophilic Oesophagitis (EoE). For example, rhinovirus C (RV-C) is a known risk factor for paediatric asthma development and is known to disrupt respiratory epithelial barrier function causing acute inflammation. In addition, EoE, a prevalent atopic condition of the oesophageal epithelium, is characterised by similar innate immune and epithelial responses to viral injury. This review examines the current literature and identifies the gaps in the field defining viral-induced effects on a vulnerable respiratory epithelium and resulting chronic inflammation, drawing from knowledge generated in acute wheezing illness, paediatric asthma and EoE. Besides highlighting the importance of epithelial structure and barrier function in allergic disease pathogenesis regardless of specific epithelial sub-types, this review focuses on the importance of examining other parallel allergic-type disease processes that may uncover commonalities driving disease pathogenesis. This in turn may be beneficial in the development of common therapeutics for current clinical management and disease prevention in the future.
Collapse
Affiliation(s)
- Rebecca L Watkinson
- Division of Paediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia.,Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Kevin Looi
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia.,School of Public Health, Curtin University, Bentley, WA, Australia
| | - Ingrid A Laing
- Division of Paediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia.,Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Antonella Cianferoni
- Pediatrics Department, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Anthony Kicic
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia.,School of Public Health, Curtin University, Bentley, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
33
|
Krammer S, Sicorschi Gutu C, Grund JC, Chiriac MT, Zirlik S, Finotto S. Regulation and Function of Interferon-Lambda (IFNλ) and Its Receptor in Asthma. Front Immunol 2021; 12:731807. [PMID: 34899691 PMCID: PMC8660125 DOI: 10.3389/fimmu.2021.731807] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/12/2021] [Indexed: 12/22/2022] Open
Abstract
Asthma is a chronic respiratory disease affecting people of all ages, especially children, worldwide. Origins of asthma are suggested to be placed in early life with heterogeneous clinical presentation, severity and pathophysiology. Exacerbations of asthma disease can be triggered by many factors, including viral respiratory tract infections. Rhinovirus (RV) induced respiratory infections are the predominant cause of the common cold and also play a crucial role in asthma development and exacerbations. Rhinovirus mainly replicates in epithelial cells lining the upper and lower respiratory tract. Type III interferons, also known as interferon-lambda (IFNλ), are potent immune mediators of resolution of infectious diseases but they are known to be involved in autoimmune diseases as well. The protective role of type III IFNs in antiviral, antibacterial, antifungal and antiprotozoal functions is of major importance for our innate immune system. The IFNλ receptor (IFNλR) is expressed in selected types of cells like epithelial cells, thus orchestrating a specific immune response at the site of viruses and bacteria entry into the body. In asthma, IFNλ restricts the development of TH2 cells, which are induced in the airways of asthmatic patients. Several studies described type III IFNs as the predominant type of interferon increased after infection caused by respiratory viruses. It efficiently reduces viral replication, viral spread into the lungs and viral transmission from infected to naive individuals. Several reports showed that bronchial epithelial cells from asthmatic subjects have a deficient response of type III interferon after RV infection ex vivo. Toll like Receptors (TLRs) recognize pathogen-associated molecular patterns (PAMPs) expressed on infectious agents, and induce the development of antiviral and antibacterial immunity. We recently discovered that activation of TLR7/8 resulted in enhanced IFNλ receptor mRNA expression in PBMCs of healthy and asthmatic children, opening new therapeutic frontiers for rhinovirus-induced asthma. This article reviews the recent advances of the literature on the regulated expression of type III Interferons and their receptor in association with rhinovirus infection in asthmatic subjects.
Collapse
Affiliation(s)
- Susanne Krammer
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Cristina Sicorschi Gutu
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Janina C Grund
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mircea T Chiriac
- Medical Clinic 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sabine Zirlik
- Medical Clinic 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany.,Medical Clinic 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
34
|
Murray LM, Thillaiyampalam G, Xi Y, Cristino AS, Upham JW. Whole transcriptome analysis of high and low IFN-α producers reveals differential response patterns following rhinovirus stimulation. Clin Transl Immunology 2021; 10:e1356. [PMID: 34868584 PMCID: PMC8599968 DOI: 10.1002/cti2.1356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 09/01/2021] [Accepted: 10/26/2021] [Indexed: 11/29/2022] Open
Abstract
Objectives Viral respiratory infections cause considerable morbidity and economic loss. While rhinoviruses (RV) typically cause little more than the common cold, they can produce severe infections and disease exacerbations in susceptible individuals, such as those with asthma. Variations in the regulation of key antiviral cytokines, particularly type I interferon (IFN‐α and IFN‐β), may contribute to RV susceptibility. To understand this variability, we compared the transcriptomes of high and low type I IFN producers. Methods Blood mononuclear cells from 238 individuals with or without asthma were cultured in the presence or absence of RV. Those samples demonstrating high or low RV‐stimulated IFN‐α production (N = 75) underwent RNA‐sequencing. Results Gene expression patterns were similar in samples from healthy participants and those with asthma. At baseline, the high IFN‐α producer group showed higher expression of genes associated with plasmacytoid dendritic cells, the innate immune response and vitamin D activation, but lower expression of oxidative stress pathways than the low IFN‐α producer group. After RV stimulation, the high IFN‐α producer group showed higher expression of genes found in immune response biological pathways and lower expression of genes linked to developmental and catabolic processes when compared to the low IFN‐α producer group. Conclusions These differences suggest that the high IFN‐α group has a higher level of immune system readiness, resulting in a more intense and perhaps more focussed pathogen‐specific immune response. These results contribute to a better understanding of the variability in type I IFN production between individuals.
Collapse
Affiliation(s)
- Liisa M Murray
- Diamantina Institute The University of Queensland Brisbane QLD Australia
| | - Gayathri Thillaiyampalam
- Diamantina Institute The University of Queensland Brisbane QLD Australia.,Griffith Institute for Drug Discovery Griffith University Brisbane QLD Australia
| | - Yang Xi
- Diamantina Institute The University of Queensland Brisbane QLD Australia
| | - Alexandre S Cristino
- Diamantina Institute The University of Queensland Brisbane QLD Australia.,Griffith Institute for Drug Discovery Griffith University Brisbane QLD Australia
| | - John W Upham
- Diamantina Institute The University of Queensland Brisbane QLD Australia.,Respiratory and Sleep Medicine Princess Alexandra Hospital Brisbane QLD Australia
| |
Collapse
|
35
|
Coleman C, Doyle-Meyers LA, Russell-Lodrigue KE, Golden N, Threeton B, Song K, Pierre G, Baribault C, Bohm RP, Maness NJ, Kolls JK, Rappaport J, Mudd JC. Similarities and Differences in the Acute-Phase Response to SARS-CoV-2 in Rhesus Macaques and African Green Monkeys. Front Immunol 2021; 12:754642. [PMID: 34691074 PMCID: PMC8527883 DOI: 10.3389/fimmu.2021.754642] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022] Open
Abstract
Understanding SARS-CoV-2 immune pathology is critical for the development of effective vaccines and treatments. Here, we employed unbiased serial whole-blood transcriptome profiling by weighted gene network correlation analysis (WGCNA) at pre-specified timepoints of infection to understand SARS-CoV-2-related immune alterations in a cohort of rhesus macaques (RMs) and African green monkeys (AGMs) presenting with varying degrees of pulmonary pathology. We found that the bulk of transcriptional changes occurred at day 3 post-infection and normalized to pre-infection levels by 3 weeks. There was evidence of coordination of transcriptional networks in blood (defined by WGCNA) and the nasopharyngeal SARS-CoV-2 burden as well as the absolute monocyte count. Pathway analysis of gene modules revealed prominent regulation of type I and type II interferon stimulated genes (ISGs) in both RMs and AGMs, with the latter species exhibiting a greater breadth of ISG upregulation. Notably, pathways relating to neutrophil degranulation were enriched in blood of SARS-CoV-2 infected AGMs, but not RMs. Our results elude to hallmark similarities as well as differences in the RM and AGM acute response to SARS-CoV-2 infection, and may help guide the selection of particular NHP species in modeling aspects of COVID-19 disease outcome.
Collapse
Affiliation(s)
- Celeste Coleman
- Department of Immunology and Microbiology, Tulane National Primate Research Center, Covington, LA, United States.,Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Lara A Doyle-Meyers
- Department of Immunology and Microbiology, Tulane National Primate Research Center, Covington, LA, United States.,Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Kasi E Russell-Lodrigue
- Department of Immunology and Microbiology, Tulane National Primate Research Center, Covington, LA, United States.,Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Nadia Golden
- Department of Immunology and Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Breanna Threeton
- Department of Immunology and Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Kejing Song
- Center for Translational Research in Infection and Inflammation, Department of Pediatrics and Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Genevieve Pierre
- Center for Translational Research in Infection and Inflammation, Department of Pediatrics and Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Carl Baribault
- Center for Research & Scientific Computing, Tulane University Information Technology, New Orleans, LA, United States
| | - Rudolf P Bohm
- Department of Immunology and Microbiology, Tulane National Primate Research Center, Covington, LA, United States.,Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Nicholas J Maness
- Department of Immunology and Microbiology, Tulane National Primate Research Center, Covington, LA, United States.,Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Department of Pediatrics and Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jay Rappaport
- Department of Immunology and Microbiology, Tulane National Primate Research Center, Covington, LA, United States.,Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Joseph C Mudd
- Department of Immunology and Microbiology, Tulane National Primate Research Center, Covington, LA, United States.,Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
36
|
Yang Z, Mitländer H, Vuorinen T, Finotto S. Mechanism of Rhinovirus Immunity and Asthma. Front Immunol 2021; 12:731846. [PMID: 34691038 PMCID: PMC8526928 DOI: 10.3389/fimmu.2021.731846] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/20/2021] [Indexed: 12/30/2022] Open
Abstract
The majority of asthma exacerbations in children are caused by Rhinovirus (RV), a positive sense single stranded RNA virus of the Picornavirus family. The host has developed virus defense mechanisms that are mediated by the upregulation of interferon-activated signaling. However, the virus evades the immune system by inducing immunosuppressive cytokines and surface molecules like programmed cell death protein 1 (PD-1) and its ligand (PD-L1) on immunocompetent cells. Initially, RV infects epithelial cells, which constitute a physiologic mucosal barrier. Upon virus entrance, the host cell immediately recognizes viral components like dsRNA, ssRNA, viral glycoproteins or CpG-DNA by host pattern recognition receptors (PRRs). Activation of toll like receptors (TLR) 3, 7 and 8 within the endosome and through MDA-5 and RIG-I in the cytosol leads to the production of interferon (IFN) type I and other antiviral agents. Every cell type expresses IFNAR1/IFNAR2 receptors thus allowing a generalized antiviral activity of IFN type I resulting in the inhibition of viral replication in infected cells and preventing viral spread to non-infected cells. Among immune evasion mechanisms of the virus, there is downregulation of IFN type I and its receptor as well as induction of the immunosuppressive cytokine TGF-β. TGF-β promotes viral replication and is associated with induction of the immunosuppression signature markers LAP3, IDO and PD-L1. This article reviews the recent advances on the regulation of interferon type I expression in association with RV infection in asthmatics and the immunosuppression induced by the virus.
Collapse
Affiliation(s)
- Zuqin Yang
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Hannah Mitländer
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tytti Vuorinen
- Medical Microbiology, Turku University Hospital, Institut of Biomedicine, University of Turku, Turku, Finland
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
37
|
Kan-O K, Washio Y, Fujimoto T, Shiroyama N, Nakano T, Wakamatsu K, Takata S, Yoshida M, Fujita M, Matsumoto K. Differences in the spectrum of respiratory viruses and detection of human rhinovirus C in exacerbations of adult asthma and chronic obstructive pulmonary disease. Respir Investig 2021; 60:129-136. [PMID: 34580039 DOI: 10.1016/j.resinv.2021.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/08/2021] [Accepted: 08/27/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Viral respiratory infections are a common cause of acute exacerbations of chronic obstructive pulmonary disease (AECOPD) and asthma. We conducted a multicenter prospective study to determine the differences in the spectrum of viruses between adults with asthma exacerbations and AECOPD and assessed the prevalence and impact of human rhinovirus (HRV)-C in adults, which is more pathogenic in children with asthma than other HRV species. METHODS Nasopharyngeal and serum samples and clinical information were collected from 64 outpatients with adult asthma exacerbations and 44 outpatients with AECOPD between April 2018 and March 2020. Viral pathogens and HRV strains were identified from nasal samples by multiplex PCR and VP4/VP2 nested PCR. RESULTS Viral pathogens were identified in 31 patients with asthma exacerbations (48.4%) and 17 patients with AECOPD (38.6%). The most commonly detected viruses were HRV/enterovirus followed by human metapneumovirus (hMPV) in patients with asthma exacerbations, and hMPV followed by parainfluenza virus in patients with AECOPD. HRV-C was the HRV species most commonly associated with both asthma exacerbations and AECOPD. Clinical characteristics, baseline lung function, serum inflammatory chemokines, hospitalization, and systemic steroid use did not differ between HRV-C-positive patients and those positive for other HRV species. CONCLUSIONS Exacerbation-associated spectrum of viruses differed between adults with asthma exacerbations and AECOPD. HRV-C was the HRV species most often observed in adult asthma exacerbations and AECOPD, although it did not worsen patients' clinical outcomes relative to those of patients with other HRVs. Underlying disease-specific factors may be responsible for susceptibility to respiratory viruses. TRIAL REGISTRATION UMIN-CTR UMIN000031934.
Collapse
Affiliation(s)
- Keiko Kan-O
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan; Department of Endoscopic Diagnostics and Therapeutics, Kyushu University Hospital, Fukuoka, 812-8582, Japan.
| | - Yasuyoshi Washio
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Tsuguto Fujimoto
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Natsuko Shiroyama
- Department of Respiratory Medicine, Fukuoka University School of Medicine, Fukuoka, 814-0180, Japan
| | - Takako Nakano
- Department of Respiratory Medicine, National Hospital Organization Fukuokahigashi Medical Center, Koga, 811-3195, Japan
| | - Kentaro Wakamatsu
- Department of Respiratory Medicine, National Hospital Organization Omuta National Hospital, Omuta, 837-0911, Japan
| | - Shohei Takata
- Department of Respiratory Medicine, National Hospital Organization Fukuokahigashi Medical Center, Koga, 811-3195, Japan
| | - Makoto Yoshida
- Division of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, Fukuoka, 811-1394, Japan
| | - Masaki Fujita
- Department of Respiratory Medicine, Fukuoka University School of Medicine, Fukuoka, 814-0180, Japan
| | - Koichiro Matsumoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| |
Collapse
|
38
|
Kwon JH, Wi CI, Seol HY, Park M, King K, Ryu E, Sohn S, Liu H, Juhn YJ. Risk, Mechanisms and Implications of Asthma-Associated Infectious and Inflammatory Multimorbidities (AIMs) among Individuals With Asthma: a Systematic Review and a Case Study. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2021; 13:697-718. [PMID: 34486256 PMCID: PMC8419637 DOI: 10.4168/aair.2021.13.5.697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/15/2021] [Indexed: 11/25/2022]
Abstract
Our prior work and the work of others have demonstrated that asthma increases the risk of a broad range of both respiratory (e.g., pneumonia and pertussis) and non-respiratory (e.g., zoster and appendicitis) infectious diseases as well as inflammatory diseases (e.g., celiac disease and myocardial infarction [MI]), suggesting the systemic disease nature of asthma and its impact beyond the airways. We call these conditions asthma-associated infectious and inflammatory multimorbidities (AIMs). At present, little is known about why some people with asthma are at high-risk of AIMs, and others are not, to the extent to which controlling asthma reduces the risk of AIMs and which specific therapies mitigate the risk of AIMs. These questions represent a significant knowledge gap in asthma research and unmet needs in asthma care, because there are no guidelines addressing the identification and management of AIMs. This is a systematic review on the association of asthma with the risk of AIMs and a case study to highlight that 1) AIMs are relatively under-recognized conditions, but pose major health threats to people with asthma; 2) AIMs provide insights into immunological and clinical features of asthma as a systemic inflammatory disease beyond a solely chronic airway disease; and 3) it is time to recognize AIMs as a distinctive asthma phenotype in order to advance asthma research and improve asthma care. An improved understanding of AIMs and their underlying mechanisms will bring valuable and new perspectives improving the practice, research, and public health related to asthma.
Collapse
Affiliation(s)
- Jung Hyun Kwon
- Precision Population Science Lab, Department of Pediatrics and Adolescence Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Pediatrics, Korea University College of Medicine, Seoul, Korea
| | - Chung-Il Wi
- Precision Population Science Lab, Department of Pediatrics and Adolescence Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hee Yun Seol
- Precision Population Science Lab, Department of Pediatrics and Adolescence Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Miguel Park
- Division of Allergy and Immunology, Mayo Clinic, Rochester, MN, USA
| | - Katherine King
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Euijung Ryu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Sunghwan Sohn
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Hongfang Liu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Young J Juhn
- Precision Population Science Lab, Department of Pediatrics and Adolescence Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
39
|
Coultas JA, Cafferkey J, Mallia P, Johnston SL. Experimental Antiviral Therapeutic Studies for Human Rhinovirus Infections. J Exp Pharmacol 2021; 13:645-659. [PMID: 34276229 PMCID: PMC8277446 DOI: 10.2147/jep.s255211] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/01/2021] [Indexed: 12/17/2022] Open
Abstract
Rhinovirus infection is common and usually causes mild, self-limiting upper respiratory tract symptoms. Rhinoviruses can cause exacerbation of chronic respiratory diseases, such as asthma or chronic obstructive pulmonary disease, leading to a significant burden of morbidity and mortality. There has been a great deal of progress in efforts to understand the immunological basis of rhinovirus infection. However, despite a number of in vitro and in vivo attempts, there have been no effective treatments developed. This review article summarises the up to date virological and immunological understanding of these infections. We discuss the challenges researchers face, and key solutions, in their work to investigate potential therapies including in vivo rhinovirus challenge studies. Finally, we explore past and present experimental therapeutic strategies employed in the treatment of rhinovirus infections and highlight promising areas of future work.
Collapse
Affiliation(s)
- James A Coultas
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Cafferkey
- Respiratory Medicine, St Mary's Hospital, Imperial College Healthcare Foundation Trust, London, UK
| | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
40
|
Girkin J, Loo SL, Esneau C, Maltby S, Mercuri F, Chua B, Reid AT, Veerati PC, Grainge CL, Wark PAB, Knight D, Jackson D, Demaison C, Bartlett NW. TLR2-mediated innate immune priming boosts lung anti-viral immunity. Eur Respir J 2021; 58:13993003.01584-2020. [PMID: 33303547 DOI: 10.1183/13993003.01584-2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/27/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND We assessed whether Toll-like receptor (TLR)2 activation boosts the innate immune response to rhinovirus infection, as a treatment strategy for virus-induced respiratory diseases. METHODS We employed treatment with a novel TLR2 agonist (INNA-X) prior to rhinovirus infection in mice, and INNA-X treatment in differentiated human bronchial epithelial cells derived from asthmatic-donors. We assessed viral load, immune cell recruitment, cytokines, type I and III interferon (IFN) production, as well as the lung tissue and epithelial cell immune transcriptome. RESULTS We show, in vivo, that a single INNA-X treatment induced innate immune priming characterised by low-level IFN-λ, Fas ligand, chemokine expression and airway lymphocyte recruitment. Treatment 7 days before infection significantly reduced lung viral load, increased IFN-β/λ expression and inhibited neutrophilic inflammation. Corticosteroid treatment enhanced the anti-inflammatory effects of INNA-X. Treatment 1 day before infection increased expression of 190 lung tissue immune genes. This tissue gene expression signature was absent with INNA-X treatment 7 days before infection, suggesting an alternate mechanism, potentially via establishment of immune cell-mediated mucosal innate immunity. In vitro, INNA-X treatment induced a priming response defined by upregulated IFN-λ, chemokine and anti-microbial gene expression that preceded an accelerated response to infection enriched for nuclear factor (NF)-κB-regulated genes and reduced viral loads, even in epithelial cells derived from asthmatic donors with intrinsic delayed anti-viral immune response. CONCLUSION Airway epithelial cell TLR2 activation induces prolonged innate immune priming, defined by early NF-κB activation, IFN-λ expression and lymphocyte recruitment. This response enhanced anti-viral innate immunity and reduced virus-induced airway inflammation.
Collapse
Affiliation(s)
- Jason Girkin
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,These authors contributed equally
| | - Su-Ling Loo
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,These authors contributed equally
| | - Camille Esneau
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Steven Maltby
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | | | - Brendon Chua
- Dept of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Andrew T Reid
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Punnam Chander Veerati
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Chris L Grainge
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Darryl Knight
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - David Jackson
- Dept of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | | | - Nathan W Bartlett
- Viral Immunology and Respiratory Disease group, University of Newcastle, Newcastle, Australia .,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| |
Collapse
|
41
|
Weidner J, Bartel S, Kılıç A, Zissler UM, Renz H, Schwarze J, Schmidt‐Weber CB, Maes T, Rebane A, Krauss‐Etschmann S, Rådinger M. Spotlight on microRNAs in allergy and asthma. Allergy 2021; 76:1661-1678. [PMID: 33128813 PMCID: PMC8246745 DOI: 10.1111/all.14646] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/16/2020] [Accepted: 10/25/2020] [Indexed: 12/14/2022]
Abstract
In past 10 years, microRNAs (miRNAs) have gained scientific attention due to their importance in the pathophysiology of allergic diseases and their potential as biomarkers in liquid biopsies. They act as master post‐transcriptional regulators that control most cellular processes. As one miRNA can target several mRNAs, often within the same pathway, dysregulated expression of miRNAs may alter particular cellular responses and contribute, or lead, to the development of various diseases. In this review, we give an overview of the current research on miRNAs in allergic diseases, including atopic dermatitis, allergic rhinitis, and asthma. Specifically, we discuss how individual miRNAs function in the regulation of immune responses in epithelial cells and specialized immune cells in response to different environmental factors and respiratory viruses. In addition, we review insights obtained from experiments with murine models of allergic airway and skin inflammation and offer an overview of studies focusing on miRNA discovery using profiling techniques and bioinformatic modeling of the network effect of multiple miRNAs. In conclusion, we highlight the importance of research into miRNA function in allergy and asthma to improve our knowledge of the molecular mechanisms involved in the pathogenesis of this heterogeneous group of diseases.
Collapse
Affiliation(s)
- Julie Weidner
- Department of Internal Medicine and Clinical Nutrition Krefting Research Centre Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Sabine Bartel
- Department of Pathology and Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Ayse Kılıç
- Channing Division of Network Medicine Brigham and Women's Hospital Boston MA USA
| | - Ulrich M. Zissler
- Center for Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
| | - Harald Renz
- Institut für Laboratoriumsmedizin und Pathobiochemie Philipps University of Marburg Marburg Germany
| | - Jürgen Schwarze
- Centre for Inflammation Research The University of Edinburgh Edinburgh UK
| | - Carsten B. Schmidt‐Weber
- Center for Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
| | - Tania Maes
- Department of Respiratory Medicine Ghent University Ghent Belgium
| | - Ana Rebane
- Institute of Biomedicine and Translational Medicine University of Tartu Tartu Estonia
| | - Susanne Krauss‐Etschmann
- Research Center Borstel Borstel Germany
- Institute of Experimental Medicine Christian‐Albrechts University Kiel Kiel Germany
| | - Madeleine Rådinger
- Department of Internal Medicine and Clinical Nutrition Krefting Research Centre Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| |
Collapse
|
42
|
Mast Cell Proteases Tryptase and Chymase Induce Migratory and Morphological Alterations in Bronchial Epithelial Cells. Int J Mol Sci 2021; 22:ijms22105250. [PMID: 34065716 PMCID: PMC8156481 DOI: 10.3390/ijms22105250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/07/2021] [Accepted: 05/13/2021] [Indexed: 12/19/2022] Open
Abstract
Chronic respiratory diseases are often characterized by impaired epithelial function and remodeling. Mast cells (MCs) are known to home into the epithelium in respiratory diseases, but the MC-epithelial interactions remain less understood. Therefore, this study aimed to investigate the effect of MC proteases on bronchial epithelial morphology and function. Bronchial epithelial cells were stimulated with MC tryptase and/or chymase. Morphology and epithelial function were performed using cell tracking analysis and holographic live-cell imaging. Samples were also analyzed for motility-associated gene expression. Immunocytochemistry was performed to compare cytoskeletal arrangement. Stimulated cells showed strong alterations on gene, protein and functional levels in several parameters important for maintaining epithelial function. The most significant increases were found in cell motility, cellular speed and cell elongation compared to non-stimulated cells. Also, cell morphology was significantly altered in chymase treated compared to non-stimulated cells. In the current study, we show that MC proteases can induce cell migration and morphological and proliferative alterations in epithelial cells. Thus, our data imply that MC release of proteases may play a critical role in airway epithelial remodeling and disruption of epithelial function.
Collapse
|
43
|
COPD Is Associated with Elevated IFN-β Production by Bronchial Epithelial Cells Infected with RSV or hMPV. Viruses 2021; 13:v13050911. [PMID: 34069223 PMCID: PMC8156254 DOI: 10.3390/v13050911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 12/22/2022] Open
Abstract
IFN treatment may be a viable option for treating COPD exacerbations based on evidence of IFN deficiency in COPD. However, in vitro studies have used primarily influenza and rhinoviruses to investigate IFN responses. This study aims to investigate the susceptibility to infection and IFN response of primary bronchial epithelial cells (BECs) from COPD donors to infection with RSV and hMPV. BECs from five COPD and five healthy donors were used to establish both submerged monolayer and well-differentiated (WD) cultures. Two isolates of both RSV and hMPV were used to infect cells. COPD was not associated with elevated susceptibility to infection and there was no evidence of an intrinsic defect in IFN production in either cell model to either virus. Conversely, COPD was associated with significantly elevated IFN-β production in response to both viruses in both cell models. Only in WD-BECs infected with RSV was elevated IFN-β associated with reduced viral shedding. The role of elevated epithelial cell IFN-β production in the pathogenesis of COPD is not clear and warrants further investigation. Viruses vary in the responses that they induce in BECs, and so conclusions regarding antiviral responses associated with disease cannot be made based on single viral infections.
Collapse
|
44
|
Fitzpatrick AM, Chipps BE, Holguin F, Woodruff PG. T2-"Low" Asthma: Overview and Management Strategies. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 8:452-463. [PMID: 32037109 DOI: 10.1016/j.jaip.2019.11.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
Although the term "asthma" has been applied to all patients with airway lability and variable chest symptoms for centuries, phenotypes of asthma with distinct clinical and molecular features that may warrant different treatment approaches are well recognized. Patients with type 2 (T2)-"high" asthma are characterized by upregulation of T2 immune pathways (ie, IL-4 and IL-13 gene sets) and eosinophilic airway inflammation, whereas these features are absent in patients with T2-"low" asthma and may contribute to poor responsiveness to corticosteroid treatment. This review details definitions and clinical features of T2-"low" asthma, potential mechanisms and metabolic aspects, pediatric considerations, and potential treatment approaches. Priority research questions for T2-"low" asthma are also discussed.
Collapse
Affiliation(s)
| | - Bradley E Chipps
- Capital Allergy and Respiratory Disease Center, Sacramento, Calif
| | - Fernando Holguin
- University of Colorado, Pulmonary Sciences and Critical Care Medicine, Denver, Colo
| | - Prescott G Woodruff
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and the Cardiovascular Research Institute, University of California, San Francisco, Calif
| |
Collapse
|
45
|
Krug J, Kiefer A, Koelle J, Vuorinen T, Xepapadaki P, Stanic B, Chiriac MT, Akdis M, Zimmermann T, Papadopoulos NG, Finotto S. TLR7/8 regulates type I and type III interferon signalling in rhinovirus 1b-induced allergic asthma. Eur Respir J 2021; 57:13993003.01562-2020. [PMID: 33303556 DOI: 10.1183/13993003.01562-2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Interferon (IFN) responses have been reported to be defective in rhinovirus (RV)-induced asthma. The heterodimeric receptor of type I IFN (IFN-α/β) is composed of IFN-αR1 and IFN-αR2. Ligand binding to the IFN-α/β receptor complex activates signal transducer and activator of transcription (STAT) proteins STAT1 and STAT2 intracellularly. Although type III IFN (IFN-λ) binds to a different receptor containing IFN-λR1 and interleukin-10R2, its triggering leads to activation of the same downstream transcription factors. Here, we analysed the effects of RV on IFN type I and III receptors, and asked about possible Toll-like receptor 7/8 (TLR7/8) agonist R848-mediated IFN-αR1 and IFN-λR1 regulation. METHODS We measured IFN-α, IFN-β and IFN-λ and their receptor levels in peripheral blood mononuclear cell (PBMC) supernatants and cell pellets stimulated with RV1b and R848 in two cohorts of children with and without asthma recruited at pre-school age (PreDicta) and at primary school age (AGENDAS) as well as in cell supernatants from total lung cells isolated from mice. RESULTS We observed that R848 induced IFN-λR mRNA expression in PBMCs of healthy and asthmatic children, but suppressed IFN-αR mRNA levels. In murine lung cells, RV1b alone and together with R848 suppressed IFN-αR protein in T-cells compared with controls and in total lung IFN-λR mRNA compared with RV1b infection alone. CONCLUSIONS In PBMCs from pre-school age children, IFN-αR mRNA was reduced and IFN-λR1 mRNA was induced upon treatment with the TLR7/8 agonist R848, thus suggesting new avenues for induction of antiviral immune responses in paediatric asthma.
Collapse
Affiliation(s)
- Jasmin Krug
- Dept of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Alexander Kiefer
- Dept of Allergy and Pneumology, Children's Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Koelle
- Dept of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Paraskevi Xepapadaki
- Dept of Allergy, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Barbara Stanic
- Musculoskeletal Infection, AO Research Institute Davos, Davos Platz, Switzerland
| | - Mircea T Chiriac
- I Medical Clinic, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos Wolfgang, Switzerland
| | - Theodor Zimmermann
- Dept of Allergy and Pneumology, Children's Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Nikolaos G Papadopoulos
- Dept of Allergy, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.,Centre for Respiratory Medicine and Allergy, University of Manchester, Manchester, UK
| | - Susetta Finotto
- Dept of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
46
|
Damialis A, Gilles S, Sofiev M, Sofieva V, Kolek F, Bayr D, Plaza MP, Leier-Wirtz V, Kaschuba S, Ziska LH, Bielory L, Makra L, Del Mar Trigo M, Traidl-Hoffmann C. Higher airborne pollen concentrations correlated with increased SARS-CoV-2 infection rates, as evidenced from 31 countries across the globe. Proc Natl Acad Sci U S A 2021; 118:e2019034118. [PMID: 33798095 PMCID: PMC7999946 DOI: 10.1073/pnas.2019034118] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pollen exposure weakens the immunity against certain seasonal respiratory viruses by diminishing the antiviral interferon response. Here we investigate whether the same applies to the pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is sensitive to antiviral interferons, if infection waves coincide with high airborne pollen concentrations. Our original hypothesis was that more airborne pollen would lead to increases in infection rates. To examine this, we performed a cross-sectional and longitudinal data analysis on SARS-CoV-2 infection, airborne pollen, and meteorological factors. Our dataset is the most comprehensive, largest possible worldwide from 130 stations, across 31 countries and five continents. To explicitly investigate the effects of social contact, we additionally considered population density of each study area, as well as lockdown effects, in all possible combinations: without any lockdown, with mixed lockdown-no lockdown regime, and under complete lockdown. We found that airborne pollen, sometimes in synergy with humidity and temperature, explained, on average, 44% of the infection rate variability. Infection rates increased after higher pollen concentrations most frequently during the four previous days. Without lockdown, an increase of pollen abundance by 100 pollen/m3 resulted in a 4% average increase of infection rates. Lockdown halved infection rates under similar pollen concentrations. As there can be no preventive measures against airborne pollen exposure, we suggest wide dissemination of pollen-virus coexposure dire effect information to encourage high-risk individuals to wear particle filter masks during high springtime pollen concentrations.
Collapse
Affiliation(s)
- Athanasios Damialis
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany;
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Stefanie Gilles
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Mikhail Sofiev
- Finnish Meteorological Institute, Helsinki FI-00101, Finland
| | | | - Franziska Kolek
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Daniela Bayr
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Maria P Plaza
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Vivien Leier-Wirtz
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Sigrid Kaschuba
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Lewis H Ziska
- Mailman School of Public Health, Columbia University, New York, NY 10032
| | - Leonard Bielory
- Center for Environmental Prediction, Rutgers University, New Brunswick, NJ 08901
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ 08854
- Medicine, Allergy, Immunology and Ophthalmology Department, Hackensack Meridian School of Medicine, Nutley, NJ 07110
- New Jersey Center of Science, Technology and Mathematics, Kean University, Union, NJ 07083
| | - László Makra
- Institute of Economics and Rural Development, Faculty of Agriculture, University of Szeged, Szeged 6720, Hungary
| | - Maria Del Mar Trigo
- Department of Botany and Plant Physiology, University of Malaga, Malaga 29016, Spain
| | - Claudia Traidl-Hoffmann
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| |
Collapse
|
47
|
Damialis A, Gilles S, Sofiev M, Sofieva V, Kolek F, Bayr D, Plaza MP, Leier-Wirtz V, Kaschuba S, Ziska LH, Bielory L, Makra L, Del Mar Trigo M, Traidl-Hoffmann C. Higher airborne pollen concentrations correlated with increased SARS-CoV-2 infection rates, as evidenced from 31 countries across the globe. Proc Natl Acad Sci U S A 2021; 118. [PMID: 33798095 DOI: 10.1073/pnas.2019034118/-/dcsupplemental] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Pollen exposure weakens the immunity against certain seasonal respiratory viruses by diminishing the antiviral interferon response. Here we investigate whether the same applies to the pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is sensitive to antiviral interferons, if infection waves coincide with high airborne pollen concentrations. Our original hypothesis was that more airborne pollen would lead to increases in infection rates. To examine this, we performed a cross-sectional and longitudinal data analysis on SARS-CoV-2 infection, airborne pollen, and meteorological factors. Our dataset is the most comprehensive, largest possible worldwide from 130 stations, across 31 countries and five continents. To explicitly investigate the effects of social contact, we additionally considered population density of each study area, as well as lockdown effects, in all possible combinations: without any lockdown, with mixed lockdown-no lockdown regime, and under complete lockdown. We found that airborne pollen, sometimes in synergy with humidity and temperature, explained, on average, 44% of the infection rate variability. Infection rates increased after higher pollen concentrations most frequently during the four previous days. Without lockdown, an increase of pollen abundance by 100 pollen/m3 resulted in a 4% average increase of infection rates. Lockdown halved infection rates under similar pollen concentrations. As there can be no preventive measures against airborne pollen exposure, we suggest wide dissemination of pollen-virus coexposure dire effect information to encourage high-risk individuals to wear particle filter masks during high springtime pollen concentrations.
Collapse
Affiliation(s)
- Athanasios Damialis
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany;
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Stefanie Gilles
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Mikhail Sofiev
- Finnish Meteorological Institute, Helsinki FI-00101, Finland
| | | | - Franziska Kolek
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Daniela Bayr
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Maria P Plaza
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Vivien Leier-Wirtz
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Sigrid Kaschuba
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Lewis H Ziska
- Mailman School of Public Health, Columbia University, New York, NY 10032
| | - Leonard Bielory
- Center for Environmental Prediction, Rutgers University, New Brunswick, NJ 08901
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ 08854
- Medicine, Allergy, Immunology and Ophthalmology Department, Hackensack Meridian School of Medicine, Nutley, NJ 07110
- New Jersey Center of Science, Technology and Mathematics, Kean University, Union, NJ 07083
| | - László Makra
- Institute of Economics and Rural Development, Faculty of Agriculture, University of Szeged, Szeged 6720, Hungary
| | - Maria Del Mar Trigo
- Department of Botany and Plant Physiology, University of Malaga, Malaga 29016, Spain
| | - Claudia Traidl-Hoffmann
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| |
Collapse
|
48
|
Ballester B, Milara J, Cortijo J. The role of mucin 1 in respiratory diseases. Eur Respir Rev 2021; 30:30/159/200149. [PMID: 33536260 DOI: 10.1183/16000617.0149-2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/04/2020] [Indexed: 01/21/2023] Open
Abstract
Recent evidence has demonstrated that mucin 1 (MUC1) is involved in many pathological processes that occur in the lung. MUC1 is a transmembrane protein mainly expressed by epithelial and hematopoietic cells. It has a receptor-like structure, which can sense the external environment and activate intracellular signal transduction pathways through its cytoplasmic domain. The extracellular domain of MUC1 can be released to the external environment, thus acting as a decoy barrier to mucosal pathogens, as well as serving as a serum biomarker for the diagnosis and prognosis of several respiratory diseases such as lung cancer and interstitial lung diseases. Furthermore, bioactivated MUC1-cytoplasmic tail (CT) has been shown to act as an anti-inflammatory molecule in several airway infections and mediates the expression of anti-inflammatory genes in lung diseases such as chronic rhinosinusitis, chronic obstructive pulmonary disease and severe asthma. Bioactivated MUC1-CT has also been reported to interact with several effectors linked to cellular transformation, contributing to the progression of respiratory diseases such as lung cancer and pulmonary fibrosis. In this review, we summarise the current knowledge of MUC1 as a promising biomarker and drug target for lung disease.
Collapse
Affiliation(s)
- Beatriz Ballester
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA .,CIBERES, Health Institute Carlos III, Valencia, Spain.,Both authors contributed equally to this work
| | - Javier Milara
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Pharmacy Unit, Consorcio Hospital General de Valencia, Valencia, Spain.,Pharmacology Dept, University Jaume I, Castellon, Spain.,Both authors contributed equally to this work
| | - Julio Cortijo
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Research and teaching Unit, Consorcio Hospital General de Valencia, Valencia, Spain.,Dept of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
49
|
McCrae C, Olsson M, Gustafson P, Malmgren A, Aurell M, Fagerås M, Da Silva CA, Cavallin A, Paraskos J, Karlsson K, Wingren C, Monk P, Marsden R, Harrison T. INEXAS: A Phase 2 Randomized Trial of On-demand Inhaled Interferon Beta-1a in Severe Asthmatics. Clin Exp Allergy 2021; 51:273-283. [PMID: 33091192 PMCID: PMC7984268 DOI: 10.1111/cea.13765] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/18/2020] [Accepted: 10/17/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Upper respiratory tract infections (URTIs) are important triggers for asthma exacerbations. We hypothesized that inhalation of the anti-viral cytokine, interferon (IFN)-β, during URTI, could prevent these exacerbations. OBJECTIVE To evaluate the efficacy of on-demand inhaled IFN-β1a (AZD9412) to prevent severe asthma exacerbations following symptomatic URTI. METHODS This was a randomized, double-blind, placebo-controlled trial in which patients with severe asthma (GINA 4-5; n = 121) reporting URTI symptoms were randomized to 14 days of once-daily nebulized AZD9412 or placebo. The primary endpoint was severe exacerbations during treatment. Secondary endpoints included 6-item asthma control questionnaire (ACQ-6) and lung function. Exploratory biomarkers included IFN-response markers in serum and sputum, blood leucocyte counts and serum inflammatory cytokines. RESULTS Following a pre-planned interim analysis, the trial was terminated early due to an unexpectedly low exacerbation rate. Asthma worsenings were generally mild and tended to peak at randomization, possibly contributing to the lack of benefit of AZD9412 on other asthma endpoints. Numerically, AZD9412 did not reduce severe exacerbation rate, ACQ-6, asthma symptom scores or reliever medication use. AZD9412 improved lung function (morning peak expiratory flow; mPEF) by 19.7 L/min. Exploratory post hoc analyses indicated a greater mPEF improvement by AZD9412 in patients with high blood eosinophils (>0.3 × 109 /L) at screening and low serum interleukin-18 relative change at pre-treatment baseline. Pharmacodynamic effect of AZD9412 was confirmed using IFN-response markers. CONCLUSIONS & CLINICAL RELEVANCE Colds did not have the impact on asthma patients that was expected and, due to the low exacerbation rate, the trial was stopped early. On-demand AZD9412 treatment did not numerically reduce the number of exacerbations, but did attenuate URTI-induced worsening of mPEF. Severe asthma patients with high blood eosinophils or low serum interleukin-18 response are potential subgroups for further investigation of inhaled IFN-β1a.
Collapse
Affiliation(s)
- Christopher McCrae
- Translational Science and Experimental Medicine, Research and Early DevelopmentRespiratory & Immunology, BioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
- Krefting Research CentreDepartment of Internal Medicine and Clinical NutritionInstitute of MedicineUniversity of GothenburgGothenburgSweden
| | - Marita Olsson
- Early Biometrics and Statistical InnovationData Science and AI, BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Per Gustafson
- BioPharmaceutical MedicalAstraZenecaGothenburgSweden
| | - Anna Malmgren
- Early Respiratory & Immunology Projects DepartmentBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Malin Aurell
- Early Respiratory & Immunology Clinical DevelopmentBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Malin Fagerås
- BioPharmaceutical MedicalAstraZenecaGothenburgSweden
| | - Carla A. Da Silva
- Early Respiratory & Immunology Clinical DevelopmentBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Anders Cavallin
- Translational Science and Experimental MedicineEarly Cardiovascular, Renal and Metabolism (CVRM)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Jonathan Paraskos
- Point of Care Diagnostics, Precision MedicineOncology R&DAstraZenecaCambridgeUK
| | - Karin Karlsson
- Translational Science and Experimental MedicineEarly Cardiovascular, Renal and Metabolism (CVRM)BioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Cecilia Wingren
- Translational Science and Experimental Medicine, Research and Early DevelopmentRespiratory & Immunology, BioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
| | - Phillip Monk
- Synairgen Research LtdSouthampton University HospitalSouthamptonUK
| | - Richard Marsden
- Synairgen Research LtdSouthampton University HospitalSouthamptonUK
| | - Tim Harrison
- Nottingham NIHR Biomedical Research CentreUniversity of NottinghamNottingham City HospitalNottinghamUK
| |
Collapse
|
50
|
Jazaeri S, Goldsmith AM, Jarman CR, Lee J, Hershenson MB, Lewis TC. Nasal interferon responses to community rhinovirus infections are similar in controls and children with asthma. Ann Allergy Asthma Immunol 2021; 126:690-695.e1. [PMID: 33515711 DOI: 10.1016/j.anai.2021.01.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/12/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Rhinovirus (RV) is the main cause of asthma exacerbations in children. Some studies reported that persons with asthma have attenuated interferon (IFN) responses to experimental RV infection compared with healthy individuals. However, responses to community-acquired RV infections in controls and children with asthma have not been compared. OBJECTIVE To evaluate nasal cytokine responses after natural RV infections in people with asthma and healthy children. METHODS We compared nasal cytokine expression among controls and children with asthma during healthy, virus-negative surveillance weeks and self-reported RV-positive sick weeks. A total of 14 controls and 21 patients with asthma were studied. Asthma disease severity was based on symptoms and medication use. Viral genome was detected by multiplex polymerase chain reaction. Nasal cytokine protein levels were determined by multiplex assays. RESULTS Two out of 47 surveillance weeks tested positive for RV, illustrating an asymptomatic infection rate of 5%. A total of 38 of 47 sick weeks (81%) tested positive for the respiratory virus. Of these, 33 (87%) were positive for RV. During well weeks, nasal interleukin 8 (IL-8), IL-12, and IL-1β levels were higher in children with asthma than controls. Compared with healthy virus-negative surveillance weeks, IL-8, IL-13, and interferon beta increased during colds only in patients with asthma. In both controls and children with asthma, the nasal levels of interferon gamma, interferon lambda-1, IL-1β, IL-8, and IL-10 increased during RV-positive sick weeks. During RV infection, IL-8, IL-1β, and tumor necrosis factor-α levels were strongly correlated. CONCLUSION In both controls and patients with asthma, natural RV infection results in robust type II and III IFN responses.
Collapse
Affiliation(s)
| | - Adam M Goldsmith
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Caitlin R Jarman
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Julie Lee
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Marc B Hershenson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Toby C Lewis
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|