1
|
Morena D, Lumbreras S, Rodríguez JM, Campos C, Castillo M, Benavent M, Izquierdo JL. Chronic Respiratory Diseases as a Risk Factor for Herpes Zoster Infection. Arch Bronconeumol 2023; 59:797-804. [PMID: 37734964 DOI: 10.1016/j.arbres.2023.08.010] [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: 07/07/2023] [Revised: 08/20/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023]
Abstract
INTRODUCTION Herpes zoster (HZ) is a condition that results from the reactivation of the varicella zoster virus (VZV). Several diseases have been reported to increase the risk of developing HZ and postherpetic neuralgia (PHN). The objective of this study is to analyze the prevalence and risk factors for HZ and PHN in the most frequent chronic respiratory diseases, which are chronic obstructive pulmonary disease (COPD), asthma, lung cancer and obstructive sleep apnea (OSA). METHODS We conducted an observational, retrospective, non-interventional study between January 2012 and December 2020 based on data from the Castilla-La Mancha Regional Health System in Spain. We used the Savana Manager 3.0 artificial intelligence-enabled system to collect information from electronic medical records. RESULTS 31765 subjects presented a diagnosis of HZ. Mean age was 64.5 years (95%CI 64.3-64.7), and 58.2% were women. The prevalence of HZ showed an increasing trend in patients over the age of 50. A risk analysis adjusted for sex and comorbidities in COPD, asthma, lung cancer and OSA presented a higher risk of developing HZ in the first three (OR 1.16 [95%CI 1.13-1.19], 1.67 [1.63-1.71], 1.68 [1.60-1.76], respectively), which further increased in all three when associated with comorbidities. Regarding postherpetic neuralgia, an increased risk was only observed related to COPD and lung cancer (OR 1.24 [95%CI 1.23-1.25], 1.14 [1.13-1.16], respectively), further increasing when associated with comorbidities. CONCLUSIONS In a standard clinical practice setting, the most prevalent respiratory diseases (asthma, COPD and lung cancer) are related to a higher risk of HZ and PHN. These data are fundamental to assess the potential impact of vaccination in this population.
Collapse
Affiliation(s)
- Diego Morena
- Servicio de Neumología, Hospital Universitario de Guadalajara, Guadalajara, Spain; Programa de Doctorado en Ciencias de la Salud, Universidad de Alcalá, Madrid, Spain.
| | | | - José Miguel Rodríguez
- Servicio de Neumología, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Carolina Campos
- Servicio de Neumología, Hospital Universitario de Guadalajara, Guadalajara, Spain
| | - María Castillo
- Servicio de Neumología, Hospital Universitario de Guadalajara, Guadalajara, Spain
| | | | - José Luis Izquierdo
- Servicio de Neumología, Hospital Universitario de Guadalajara, Guadalajara, Spain; Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Madrid, Spain
| |
Collapse
|
2
|
Safonova E, Yawn BP, Welte T, Wang C. Risk factors for herpes zoster: should people with asthma or COPD be vaccinated? Respir Res 2023; 24:35. [PMID: 36709298 PMCID: PMC9884420 DOI: 10.1186/s12931-022-02305-1] [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: 03/09/2022] [Accepted: 12/23/2022] [Indexed: 01/30/2023] Open
Abstract
Without vaccination, an estimated 1 in 3 individuals will develop herpes zoster (HZ) in their lifetime. Increased risk of HZ is attributed to impaired cell-mediated immunity, as observed in age-related immunosenescence or in individuals immunocompromised due to disease or immunosuppressive treatments. Most vaccination guidelines recommend HZ vaccination in all adults ≥ 50 years of age, although Shingrix® was recently approved by the U.S. Food and Drug Administration for use in individuals aged ≥ 18 years who are or will be at increased risk of HZ due to immunodeficiency or immunosuppression caused by known disease or therapy, followed by approval by the European Medicines Agency for use in immunocompromised individuals aged ≥ 18 years. Chronic respiratory diseases are also risk factors for HZ. A new meta-analysis reported 24% and 41% increased risks of HZ in those with asthma and chronic obstructive pulmonary disorder (COPD), respectively, compared with healthy controls. Asthma and COPD increase a person's risk of HZ and associated complications at any age and may be further elevated in those receiving inhaled corticosteroids. Despite the increased risks, there is evidence that HZ vaccination uptake in those aged ≥ 50 years with COPD may be lower compared with the age-matched general population, potentially indicating a lack of awareness of HZ risk factors among clinicians and patients. The 2022 Global Initiative for Chronic Lung Disease report recognizes that Centers for Disease Control and Prevention recommended to vaccinate those aged ≥ 50 years against HZ, although health systems should consider the inclusion of all adults with asthma or COPD into their HZ vaccination programs. Further research into HZ vaccine efficacy/effectiveness and safety in younger populations is needed to inform vaccination guidelines.
Collapse
Affiliation(s)
| | | | - Tobias Welte
- Hannover School of Medicine and German Center for Lung Research, Hannover, Germany
| | - Chengbin Wang
- GSK Vaccines, Rockville, MD, USA.
- Novavax Inc., Gaithersburg, MD, USA.
| |
Collapse
|
3
|
Cavaliere C, Segatto M, Ciofalo A, Colizza A, Minni A, Messineo D, Lambiase A, Greco A, de Vincentiis M, Masieri S. Benralizumab reduces eosinophils and inflammatory markers in patients with severe eosinophilic asthma and chronic rhinosinusitis with nasal polyps: A pilot real-life study. Immunol Lett 2022; 248:70-77. [PMID: 35752279 DOI: 10.1016/j.imlet.2022.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022]
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) and Severe Eosinophilic Asthma (SEA) are both frequently sustained by eosinophilic inflammation and are probably the manifestation of a unique disease of upper and lower respiratory tract. We retrospectively observed 11 patients with severe CRSwNP and concomitant SEA under add-on therapy with benralizumab evaluating symptoms using Sino Nasal Outcome Test-22 (SNOT-22), Visual Analogue Scale (VAS), and Asthma Control Test (ACT) and Nasal polyp size by endoscopic and radiological score by Nasal Polyp Score (NPS) and Lund-Mackay Score (LMS). At 6 and 12 months, the expression of cationic eosinophil protein (ECP), Interleukin 17 (IL-17), Interferon gamma (INF-γ), and vascular endothelial growth factor (VEGF) was measured by nasal scraping to assess mucosal inflammation. After 12 months of benralizumab treatment, SNOT-22 decreased from 45 (23-97) to 14 (5-53) (p < 0.05), total VAS of rhinologic symptoms decreased from 30 (17-44) to 9 (5-37) (p ≤ 0.01) and ACT score increased from 10 (5-15) to 24 (20-25) (p ≤ 0.01). NPS decreased from 5 (3-6) to 3 (2-4) after 6 months (p < 0.05) and to 2 (2-3) after one year respectively (p < 0.05) and LMS total score from 21 (15-24) to 17 (8-21) (p ≤ 0.01) after 12 months from starting treatment. Nasal mucosa scraping found differences in INF-γ and VEGF expression in patients compared to 10 healthy subjects, with a normalization of these markers during eosinophils depletion induced by benralizumab. This is the first pilot real-life study conducted with an anti-IL5R monoclonal antibody in severe eosinophilic asthma and severe CRSwNP patients showing that this treatment can induce benefit both diseases not only from the clinical, but also from the inflammatory point of view. Moreover, our research pointed out that INF-γ and VEGF may represent potential response biomarker.
Collapse
Affiliation(s)
- Carlo Cavaliere
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy.
| | - Marco Segatto
- Department of Biosciences and Territory, University of Molise, Pesche, IS, Italy
| | - Andrea Ciofalo
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Andrea Colizza
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Antonio Minni
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Daniela Messineo
- Radiology, Oncology, and Anatomopathological Department, Sapienza University of Rome, Rome, Italy
| | | | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, Rome, Italy
| | - Marco de Vincentiis
- Department of Oral and Maxillo-Facial Sciences, Sapienza University of Rome, Rome, Italy
| | - Simonetta Masieri
- Department of Oral and Maxillo-Facial Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
4
|
Liew KY, Koh SK, Hooi SL, Ng MKL, Chee HY, Harith HH, Israf DA, Tham CL. Rhinovirus-Induced Cytokine Alterations With Potential Implications in Asthma Exacerbations: A Systematic Review and Meta-Analysis. Front Immunol 2022; 13:782936. [PMID: 35242128 PMCID: PMC8886024 DOI: 10.3389/fimmu.2022.782936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/13/2022] [Indexed: 12/01/2022] Open
Abstract
Background Rhinovirus (RV) infections are a major cause of asthma exacerbations. Unlike other respiratory viruses, RV causes minimal cytotoxic effects on airway epithelial cells and cytokines play a critical role in its pathogenesis. However, previous findings on RV-induced cytokine responses were largely inconsistent. Thus, this study sought to identify the cytokine/chemokine profiles induced by RV infection and their correlations with airway inflammatory responses and/or respiratory symptoms using systematic review, and to determine whether a quantitative difference exists in cytokine levels between asthmatic and healthy individuals via meta-analysis. Methods Relevant articles were obtained from PubMed, Scopus, and ScienceDirect databases. Studies that compared RV-induced cytokine responses between asthmatic and healthy individuals were included in the systematic review, and their findings were categorized based on the study designs, which were ex vivo primary bronchial epithelial cells (PBECs), ex vivo peripheral blood mononuclear cells (PBMCs), and human experimental studies. Data on cytokine levels were also extracted and analyzed using Review Manager 5.4. Results Thirty-four articles were included in the systematic review, with 18 of these further subjected to meta-analysis. Several studies reported the correlations between the levels of cytokines, such as IL-8, IL-4, IL-5, and IL-13, and respiratory symptoms. Evidence suggests that IL-25 and IL-33 may be the cytokines that promote type 2 inflammation in asthmatics after RV infection. Besides that, a meta-analysis revealed that PBECs from children with atopic asthma produced significantly lower levels of IFN-β [Effect size (ES): -0.84, p = 0.030] and IFN-λ (ES: -1.00, p = 0.002), and PBECs from adult atopic asthmatics produced significantly lower levels of IFN-β (ES: -0.68, p = 0.009), compared to healthy subjects after RV infection. A trend towards a deficient production of IFN-γ (ES: -0.56, p = 0.060) in PBMCs from adult atopic asthmatics was observed. In lower airways, asthmatics also had significantly lower baseline IL-15 (ES: -0.69, p = 0.020) levels. Conclusion Overall, RV-induced asthma exacerbations are potentially caused by an imbalance between Th1 and Th2 cytokines, which may be contributed by defective innate immune responses at cellular levels. Exogenous IFNs delivery may be beneficial as a prophylactic approach for RV-induced asthma exacerbations. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=184119, identifier CRD42020184119.
Collapse
Affiliation(s)
- Kong Yen Liew
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Sue Kie Koh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Suet Li Hooi
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | | | - Hui-Yee Chee
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hanis Hazeera Harith
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
5
|
Chen A, Diaz-Soto MP, Sanmamed MF, Adams T, Schupp JC, Gupta A, Britto C, Sauler M, Yan X, Liu Q, Nino G, Cruz CSD, Chupp GL, Gomez JL. Single-cell characterization of a model of poly I:C-stimulated peripheral blood mononuclear cells in severe asthma. Respir Res 2021; 22:122. [PMID: 33902571 PMCID: PMC8074196 DOI: 10.1186/s12931-021-01709-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/08/2021] [Indexed: 11/22/2022] Open
Abstract
Background Asthma has been associated with impaired interferon response. Multiple cell types have been implicated in such response impairment and may be responsible for asthma immunopathology. However, existing models to study the immune response in asthma are limited by bulk profiling of cells. Our objective was to Characterize a model of peripheral blood mononuclear cells (PBMCs) of patients with severe asthma (SA) and its response to the TLR3 agonist Poly I:C using two single-cell methods. Methods Two complementary single-cell methods, DropSeq for single-cell RNA sequencing (scRNA-Seq) and mass cytometry (CyTOF), were used to profile PBMCs of SA patients and healthy controls (HC). Poly I:C-stimulated and unstimulated cells were analyzed in this study. Results PBMCs (n = 9414) from five SA (n = 6099) and three HC (n = 3315) were profiled using scRNA-Seq. Six main cell subsets, namely CD4 + T cells, CD8 + T cells, natural killer (NK) cells, B cells, dendritic cells (DCs), and monocytes, were identified. CD4 + T cells were the main cell type in SA and demonstrated a pro-inflammatory profile characterized by increased JAK1 expression. Following Poly I:C stimulation, PBMCs from SA had a robust induction of interferon pathways compared with HC. CyTOF profiling of Poly I:C stimulated and unstimulated PBMCs (n = 160,000) from the same individuals (SA = 5; HC = 3) demonstrated higher CD8 + and CD8 + effector T cells in SA at baseline, followed by a decrease of CD8 + effector T cells after poly I:C stimulation. Conclusions Single-cell profiling of an in vitro model using PBMCs in patients with SA identified activation of pro-inflammatory pathways at baseline and strong response to Poly I:C, as well as quantitative changes in CD8 + effector cells. Thus, transcriptomic and cell quantitative changes are associated with immune cell heterogeneity in this model to evaluate interferon responses in severe asthma. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01709-9.
Collapse
Affiliation(s)
- Ailu Chen
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Maria P Diaz-Soto
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Miguel F Sanmamed
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Taylor Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Jonas C Schupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Amolika Gupta
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Clemente Britto
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Maor Sauler
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Xiting Yan
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Qing Liu
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Gustavo Nino
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, Department of Pediatrics, George Washington University, Washington, DC, USA
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Geoffrey L Chupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Jose L Gomez
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA.
| |
Collapse
|
6
|
Larsson O, Sunnergren O, Bachert C, Kumlien Georén S, Cardell LO. The SP-TLR axis, which locally primes the nasal mucosa, is impeded in patients with allergic rhinitis. Clin Transl Allergy 2021; 11:e12009. [PMID: 33900054 PMCID: PMC8099340 DOI: 10.1002/clt2.12009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 11/24/2022] Open
Abstract
Background Substance P (SP) and toll‐like receptors (TLRs) contribute to airway disease, particularly during viral infection. We recently demonstrated that SP can act as an initial response to viral stimuli in the upper airway by upregulating TLRs in the nasal epithelia (the SP‐TLR axis). Patients with allergic rhinitis (AR) suffer from prolonged airway infections. The aim of the present study was to examine if patients with AR exhibit a disturbance in the SP‐TLR axis. Method Human nasal biopsies and human nasal epithelial cells (HNEC) from healthy volunteers and patients with AR were cultured in the presence of SP. Epithelial expression of TLR4, neutral endopeptidase (NEP) and neurokinin 1 (NK1) were evaluated with flow cytometry and/or quantitative polymerase chain reaction after 30 min to 24 h. The effect of SP on nasal lipopolysaccharide‐induced interleukin‐8 (IL‐8) release was investigated. Results SP stimulation of tissue from healthy volunteers resulted in a transient increase of the TLR4 expression, whereas stimulation of AR patient‐derived material led to a delayed and prolonged upregulation of TLR4. NEP expression in HNEC was lower in AR than healthy controls whereas NK1 receptor expression was increased. SP pretreatment increased TLR4‐dependent IL‐8 expression in healthy controls, but not in AR. Conclusions SP‐induced regulation of TLR4 in the human nasal mucosa is disturbed in AR. An altered SP‐mediated innate immune response may contribute to the dysfunctional and often prolonged responses to infection in AR.
Collapse
Affiliation(s)
- Olivia Larsson
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ola Sunnergren
- Department of Otorhinolaryngology, Ryhov County Hospital, Jönköping, Sweden
| | - Claus Bachert
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - Susanna Kumlien Georén
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Lars Olaf Cardell
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.,Department of ENT Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
7
|
van der Heide SL, Xi Y, Upham JW. Natural Killer Cells and Host Defense Against Human Rhinoviruses Is Partially Dependent on Type I IFN Signaling. Front Cell Infect Microbiol 2020; 10:510619. [PMID: 33194777 PMCID: PMC7609819 DOI: 10.3389/fcimb.2020.510619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
Rhinovirus (RV), the causative agent of the common cold, causes only mild upper respiratory tract infections in healthy individuals, but can cause longer lasting and more severe pulmonary infections in people with chronic lung diseases and in the setting of immune suppression or immune deficiency. RV-infected lung structural cells release type I interferon (IFN-I), initiating the immune response, leading to protection against viruses in conjunction with migratory immune cells. However, IFN-I release is deficient in some people with asthma. Innate immune cells, such as natural killer (NK) cells, are proposed to play major roles in the control of viral infections, and may contribute to exacerbations of chronic lung diseases, such as asthma. In this study, we characterized the NK cell response to RV infection using an in vitro model of infection in healthy individuals, and determined the extent to which IFN-I signaling mediates this response. The results indicate that RV stimulation in vitro induces NK cell activation in healthy donors, leading to degranulation and the release of cytotoxic mediators and cytokines. IFN-I signaling was partly responsible for NK cell activation and functional responses to RV. Overall, our findings suggest the involvement of NK cells in the control of RV infection in healthy individuals. Further understanding of NK cell regulation may deepen our understanding of the mechanisms that contribute to susceptibility to RV infections in asthma and other chronic lung diseases.
Collapse
Affiliation(s)
- Saskia L van der Heide
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Yang Xi
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - John W Upham
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Department of Respiratory Medicine, Princess Alexandra Hospital, Brisbane, QLD, Australia
| |
Collapse
|
8
|
Murray LM, Yerkovich ST, Ferreira MA, Upham JW. Risks for cold frequency vary by sex: role of asthma, age, TLR7 and leukocyte subsets. Eur Respir J 2020; 56:13993003.02453-2019. [PMID: 32513781 DOI: 10.1183/13993003.02453-2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/17/2020] [Indexed: 11/05/2022]
Abstract
Viral respiratory infections are usually benign but can trigger asthma exacerbations. The factors associated with upper respiratory tract infection (cold) frequency are not fully understood, nor is it clear whether such factors differ between women and men.To determine which immunological and clinical variables associate with the frequency of self-reported respiratory infections (colds), 150 asthma cases and 151 controls were recruited. Associations between antiviral immune response variables: toll-like receptor (TLR)7/8 gene expression, plasmacytoid dendritic cell (pDC) numbers and interferon-α, tumour necrosis factor and interleukin-12 production, and asthma were then examined that might explain cold frequency.People with asthma cases reported more colds per year (median 3 versus 2; p<0.001) and had lower baseline TLR7 gene expression (odds ratio 0.12; p=0.02) than controls. Associations between many variables and cold frequency differed between women and men. In women, high blood neutrophil counts (β=0.096, p=0.002), and younger age (β=-0.017, p<0.001), but not exposure to children, were independently associated with more frequent colds. In men, low TLR7 expression (β=-0.96, p=0.041) and high CLEC4C gene expression (a marker of pDC; β=0.88, p=0.008) were independently associated with more frequent colds. Poor asthma symptom control was independently associated with reduced TLR8 gene expression (β=-1.4, p=0.036) and high body mass index (β=0.041, p=0.004).Asthma, age and markers of inflammation and antiviral immunity in peripheral blood are associated with frequent colds. Interestingly, the variables associated with cold frequency differed between women and men.
Collapse
Affiliation(s)
- Liisa M Murray
- Diamantina Institute, The University of Queensland, Brisbane, Australia
| | | | | | - John W Upham
- Diamantina Institute, The University of Queensland, Brisbane, Australia.,Respiratory and Sleep Medicine, Princess Alexandra Hospital, Brisbane, Australia
| |
Collapse
|
9
|
Rich HE, Antos D, Melton NR, Alcorn JF, Manni ML. Insights Into Type I and III Interferons in Asthma and Exacerbations. Front Immunol 2020; 11:574027. [PMID: 33101299 PMCID: PMC7546400 DOI: 10.3389/fimmu.2020.574027] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/25/2020] [Indexed: 01/16/2023] Open
Abstract
Asthma is a highly prevalent, chronic respiratory disease that impacts millions of people worldwide and causes thousands of deaths every year. Asthmatics display different phenotypes with distinct genetic components, environmental causes, and immunopathologic signatures, and are broadly characterized into type 2-high or type 2-low (non-type 2) endotypes by linking clinical characteristics, steroid responsiveness, and molecular pathways. Regardless of asthma severity and adequate disease management, patients may experience acute exacerbations of symptoms and a loss of disease control, often triggered by respiratory infections. The interferon (IFN) family represents a group of cytokines that play a central role in the protection against and exacerbation of various infections and pathologies, including asthma. Type I and III IFNs in particular play an indispensable role in the host immune system to fight off pathogens, which seems to be altered in both pediatric and adult asthmatics. Impaired IFN production leaves asthmatics susceptible to infection and with uncontrolled type 2 immunity, promotes airway hyperresponsiveness (AHR), and inflammation which can lead to asthma exacerbations. However, IFN deficiency is not observed in all asthmatics, and alterations in IFN expression may be independent of type 2 immunity. In this review, we discuss the link between type I and III IFNs and asthma both in general and in specific contexts, including during viral infection, co-infection, and bacterial/fungal infection. We also highlight several studies which examine the potential role for type I and III IFNs as asthma-related therapies.
Collapse
Affiliation(s)
- Helen E Rich
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Danielle Antos
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Natalie R Melton
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - John F Alcorn
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Michelle L Manni
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
10
|
Kim SY, Oh DJ, Choi HG. Asthma increases the risk of herpes zoster: a nested case-control study using a national sample cohort. Allergy Asthma Clin Immunol 2020; 16:52. [PMID: 32944021 PMCID: PMC7491354 DOI: 10.1186/s13223-020-00453-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 06/15/2020] [Indexed: 01/20/2023] Open
Abstract
Background This study aimed to complement previous studies on the risk of herpes zoster in the asthmatic adult population. Methods The Korean Health Insurance Review and Assessment Service—National Sample Cohort (HIRA-NSC) from 2002 through 2013 was used. A total of 64,152 participants with herpes zoster were matched for age, sex, income, region of residence, hypertension, diabetes, and dyslipidemia with 239,780 participants who were included as a control group. In both the herpes zoster and control groups, previous history of asthma were investigated. The crude and adjusted odds ratios (ORs) and 95% confidence intervals (CI) of asthma for herpes zoster were analyzed using unconditional logistic regression analysis. Subgroup analyses were conducted according to age and sex. Results Approximately 16.2% (9728/59,945) and 12.8% (30,752/239,780) of participants in the herpes zoster and control groups, respectively, had a previous history of asthma (P < 0.001). The herpes zoster group demonstrated a 1.32-times higher odds of asthma than the control group (95% CI 1.28–1.35, P < 0.001). The increased odds of asthma in the herpes zoster group persisted in all the age and sex subgroups. Conclusions The odds for asthma were higher in the herpes zoster group.
Collapse
Affiliation(s)
- So Young Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Dong Jun Oh
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyo Geun Choi
- Department of Otorhinolaryngology-Head & Neck Surgery, Hallym University College of Medicine, Sacred Heart Hospital, 22, Gwanpyeong-ro 170beon-gil, Dongan-gu, Anyang-Si, Gyeonggi-do 14068 Republic of Korea.,Hallym Data Science Laboratory, Hallym University College of Medicine, Anyang, Republic of Korea
| |
Collapse
|
11
|
Zhu J, Message SD, Mallia P, Kebadze T, Contoli M, Ward CK, Barnathan ES, Mascelli MA, Kon OM, Papi A, Stanciu LA, Edwards MR, Jeffery PK, Johnston SL. Bronchial mucosal IFN-α/β and pattern recognition receptor expression in patients with experimental rhinovirus-induced asthma exacerbations. J Allergy Clin Immunol 2019; 143:114-125.e4. [PMID: 29698627 PMCID: PMC6320262 DOI: 10.1016/j.jaci.2018.04.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 03/12/2018] [Accepted: 04/03/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The innate immune system senses viral infection through pattern recognition receptors (PRRs), leading to type I interferon production. The role of type I interferon and PPRs in rhinovirus-induced asthma exacerbations in vivo are uncertain. OBJECTIVES We sought to compare bronchial mucosal type I interferon and PRR expression at baseline and after rhinovirus infection in atopic asthmatic patients and control subjects. METHODS Immunohistochemistry was used to detect expression of IFN-α, IFN-β, and the PRRs: Toll-like receptor 3, melanoma differentiation-associated gene 5, and retinoic acid-inducible protein I in bronchial biopsy specimens from 10 atopic asthmatic patients and 15 nonasthmatic nonatopic control subjects at baseline and on day 4 and 6 weeks after rhinovirus infection. RESULTS We observed IFN-α/β deficiency in the bronchial epithelium at 3 time points in asthmatic patients in vivo. Lower epithelial IFN-α/β expression was related to greater viral load, worse airway symptoms, airway hyperresponsiveness, and reductions in lung function during rhinovirus infection. We found lower frequencies of bronchial subepithelial monocytes/macrophages expressing IFN-α/β in asthmatic patients during infection. Interferon deficiency at baseline was not accompanied by deficient PRR expression in asthmatic patients. Both epithelial and subepithelial PRR expression were induced during rhinovirus infection. Rhinovirus infection-increased numbers of subepithelial interferon/PRR-expressing inflammatory cells were related to greater viral load, airway hyperresponsiveness, and reductions in lung function. CONCLUSIONS Bronchial epithelial IFN-α/β expression and numbers of subepithelial IFN-α/β-expressing monocytes/macrophages during infection were both deficient in asthmatic patients. Lower epithelial IFN-α/β expression was associated with adverse clinical outcomes after rhinovirus infection in vivo. Increases in numbers of subepithelial cells expressing interferon/PRRs during infection were also related to greater viral load/illness severity.
Collapse
Affiliation(s)
- Jie Zhu
- Airway Disease Infection, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Simon D Message
- Airway Disease Infection, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom; Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Patrick Mallia
- Airway Disease Infection, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom; Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Tatiana Kebadze
- Airway Disease Infection, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Marco Contoli
- Airway Disease Infection, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom; Imperial College Healthcare NHS Trust, London, United Kingdom; Research Centre on Asthma and COPD, University of Ferrara, Ferrara, Italy
| | | | | | | | - Onn M Kon
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Alberto Papi
- Research Centre on Asthma and COPD, University of Ferrara, Ferrara, Italy
| | - Luminita A Stanciu
- Airway Disease Infection, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Michael R Edwards
- Airway Disease Infection, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Peter K Jeffery
- Airway Disease Infection, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Sebastian L Johnston
- Airway Disease Infection, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom; Imperial College Healthcare NHS Trust, London, United Kingdom.
| |
Collapse
|
12
|
Farahnak S, Chronopoulos J, Martin JG. Nucleic Acid Sensing in Allergic Disorders. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 345:1-33. [PMID: 30904191 DOI: 10.1016/bs.ircmb.2018.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Recent advances indicate that there is crosstalk between allergic disorders and nucleic acid sensing. Triggers that activate inflammatory mechanisms via nucleic acid sensors affect both allergic phenotypes and anti-viral responses, depending on the timing and the order of exposure. Viral respiratory infections, such as those caused by the rhinovirus, influenza, and respiratory syncytial virus, are the most frequent cause of significant asthma exacerbations through effects mediated predominantly by TLR3. However, agonists of other nucleic acid sensors, such as TLR7/8 and TLR9 agonists, may inhibit allergic inflammation and reduce clinical manifestations of disease. The allergic state can predispose the immune system to both exaggerated responses to viral infections or protection from anti-viral inflammatory responses. TH2 cytokines appear to alter the epithelium, leading to defective viral clearance or exaggerated responses to viral infections. However, a TH2 skewed allergic response may be protective against a TH1-dependent inflammatory anti-viral response. This review briefly introduces the receptors involved in nucleic acid sensing, addresses mechanisms by which nucleic acid sensing and allergic responses can counteract one another, and discusses the strategies in experimental settings, both in animal and human studies, to harness the nucleic acid sensing machinery for the intervention of allergic disorders.
Collapse
Affiliation(s)
- Soroor Farahnak
- Meakins Christie Laboratories, Research Institute of the McGill University Health Centre and McGill University, Montreal, QC, Canada
| | - Julia Chronopoulos
- Meakins Christie Laboratories, Research Institute of the McGill University Health Centre and McGill University, Montreal, QC, Canada
| | - James G Martin
- Meakins Christie Laboratories, Research Institute of the McGill University Health Centre and McGill University, Montreal, QC, Canada.
| |
Collapse
|
13
|
Boeck A, Landgraf-Rauf K, Vogelsang V, Siemens D, Prazeres da Costa O, Klucker E, von Mutius E, Buch T, Mansmann U, Schaub B. Ca 2+ and innate immune pathways are activated and differentially expressed in childhood asthma phenotypes. Pediatr Allergy Immunol 2018; 29:823-833. [PMID: 30102794 DOI: 10.1111/pai.12971] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Asthma is the most common chronic disease in children. Underlying immunologic mechanisms-in particular of different phenotypes-are still just partly understood. The objective of the study was the identification of distinct cellular pathways in allergic asthmatics (AA) and nonallergic asthmatics (NA) vs healthy controls (HC). METHODS Peripheral blood mononuclear cells (PBMCs) of steroid-naïve children (n(AA/NA/HC) = 35/13/34)) from the CLARA study (n = 275) were stimulated (anti-CD3/CD28, LpA) or kept unstimulated. Gene expression was investigated by transcriptomics and quantitative RT-PCR. Differentially regulated pathways between phenotypes were assessed after adjustment for sex and age (KEGG pathways). Networks based on correlations of gene expression were built using force-directed graph drawing. RESULTS Allergic asthmatics vs NA and asthmatics overall vs HC showed significantly different expression of Ca2+ and innate immunity-associated pathways. PCR analysis confirmed significantly increased Ca2+ -associated gene regulation (ORMDL3 and ATP2A3) in asthmatics vs HC, most prominent in AA. Innate immunity receptors (LY75, TLR7), relevant for virus infection, were also upregulated in AA and NA compared to HC. AA and NA could be differentiated by increased ATP2A3 and FPR2 in AA, decreased CLEC4E in AA, and increased IFIH1 expression in NA following anti-CD3/28 stimulation vs unstimulated (fold change). CONCLUSIONS Ca2+ regulation and innate immunity response pattern to viruses were activated in PBMCs of asthmatics. Asthma phenotypes were differentially characterized by distinct regulation of ATP2A3 and expression of innate immune receptors (FPR2, CLEC4E, IFIH1). These genes may present promising targets for future in-depth investigation with the long-term goal of more phenotype-specific therapeutic interventions in asthmatics.
Collapse
Affiliation(s)
- Andreas Boeck
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Katja Landgraf-Rauf
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany.,Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Vanessa Vogelsang
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Diana Siemens
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | | | - Elisabeth Klucker
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Erika von Mutius
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany.,Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute for Asthma and Allergy Prevention, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Ulrich Mansmann
- Institute for Medical Information Processing, Biometry and Epidemiology, LMU, Munich, Germany
| | - Bianca Schaub
- Department of Asthma & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany.,Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
14
|
Lo D, Kennedy JL, Kurten RC, Panettieri RA, Koziol-White CJ. Modulation of airway hyperresponsiveness by rhinovirus exposure. Respir Res 2018; 19:208. [PMID: 30373568 PMCID: PMC6206673 DOI: 10.1186/s12931-018-0914-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/17/2018] [Indexed: 01/12/2023] Open
Abstract
Rhinovirus (RV) exposure has been implicated in childhood development of wheeze evoking asthma and exacerbations of underlying airways disease. Studies such as the Copenhagen Prospective Studies on Asthma in Childhood (COPSAC) and Childhood Origins of ASThma (COAST) have identified RV as a pathogen inducing severe respiratory disease. RVs also modulate airway hyperresponsiveness (AHR), a key characteristic of such diseases. Although potential factors underlying mechanisms by which RV induces AHR have been postulated, the precise mechanisms of AHR following RV exposure remain elusive. A challenge to RV-related research stems from inadequate models for study. While human models raise ethical concerns and are relatively difficult in terms of subject recruitment, murine models are limited by susceptibility of infection to the relatively uncommon minor group (RV-B) serotypes, strains that are generally associated with infrequent clinical respiratory virus infections. Although a transgenic mouse strain that has been developed has enhanced susceptibility for infection with the common major group (RV-A) serotypes, few studies have focused on RV in the context of allergic airways disease rather than understanding RV-induced AHR. Recently, the receptor for the virulent RV-C CDHR3, was identified, but a dearth of studies have examined RV-C-induced effects in humans. Currently, the mechanisms by which RV infections modulate airway smooth muscle (ASM) shortening or excitation-contraction coupling remain elusive. Further, only one study has investigated the effects of RV on bronchodilatory mechanisms, with only speculation as to mechanisms underlying RV-mediated modulation of bronchoconstriction.
Collapse
Affiliation(s)
- Dennis Lo
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Joshua L Kennedy
- Department of Pediatrics, Division of Allergy and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Richard C Kurten
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Reynold A Panettieri
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Cynthia J Koziol-White
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
15
|
Gill MA, Liu AH, Calatroni A, Krouse RZ, Shao B, Schiltz A, Gern JE, Togias A, Busse WW. Enhanced plasmacytoid dendritic cell antiviral responses after omalizumab. J Allergy Clin Immunol 2018; 141:1735-1743.e9. [PMID: 28870461 PMCID: PMC6013066 DOI: 10.1016/j.jaci.2017.07.035] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/21/2017] [Accepted: 07/12/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Atopy and viral respiratory tract infections synergistically promote asthma exacerbations. IgE cross-linking inhibits critical virus-induced IFN-α responses of plasmacytoid dendritic cells (pDCs), which can be deficient in patients with allergic asthma. OBJECTIVE We sought to determine whether reducing IgE levels in vivo with omalizumab treatment increases pDC antiviral IFN-α responses in inner-city children with asthma. METHODS PBMCs and pDCs isolated from children with exacerbation-prone asthma before and during omalizumab treatment were stimulated ex vivo with rhinovirus and influenza in the presence or absence of IgE cross-linking. IFN-α levels were measured in supernatants, and mRNA expression of IFN-α pathway genes was determined by using quantitative RT-PCR (qRT-PCR) in cell pellets. FcεRIα protein levels and mRNA expression were measured in unstimulated cells by using flow cytometry and qRT-PCR, respectively. Changes in these outcomes and associations with clinical outcomes were analyzed, and statistical modeling was used to identify risk factors for asthma exacerbations. RESULTS Omalizumab treatment increased rhinovirus- and influenza-induced PBMC and rhinovirus-induced pDC IFN-α responses in the presence of IgE cross-linking and reduced pDC surface FcεRIα expression. Omalizumab-induced reductions in pDC FcεRIα levels were significantly associated with a lower asthma exacerbation rate during the outcome period and correlated with increases in PBMC IFN-α responses. PBMC FcεRIα mRNA expression measured on study entry significantly improved an existing model of exacerbation prediction. CONCLUSIONS These findings indicate that omalizumab treatment augments pDC IFN-α responses and attenuates pDC FcεRIα protein expression and provide evidence that these effects are related. These results support a potential mechanism underlying clinical observations that allergic sensitization is associated with increased susceptibility to virus-induced asthma exacerbations.
Collapse
Affiliation(s)
- Michelle A Gill
- Department of Pediatrics, Division of Infectious Diseases, and Immunology, University of Texas Southwestern Medical Center, Dallas, Tex; Department of Internal Medicine, Division of Allergy & Immunology, University of Texas Southwestern Medical Center, Dallas, Tex.
| | - Andrew H Liu
- Children's Hospital Colorado and the Department of Pediatrics, Division of Pulmonary Medicine, University of Colorado School of Medicine, Aurora, Colo
| | | | | | - Baomei Shao
- Department of Internal Medicine, Division of Allergy & Immunology, University of Texas Southwestern Medical Center, Dallas, Tex
| | - Allison Schiltz
- Children's Hospital Colorado and the Department of Pediatrics, Division of Pulmonary Medicine, University of Colorado School of Medicine, Aurora, Colo
| | - James E Gern
- Department of Medicine, Division of Allergy & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Alkis Togias
- National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - William W Busse
- Department of Medicine, Division of Allergy & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| |
Collapse
|
16
|
Gomez JL, Diaz MP, Nino G, Britto CJ. Impaired type I interferon regulation in the blood transcriptome of recurrent asthma exacerbations. BMC Med Genomics 2018; 11:21. [PMID: 29486764 PMCID: PMC5830339 DOI: 10.1186/s12920-018-0340-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/21/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Asthma exacerbations are an important cause of morbidity in asthma. Respiratory infections are often involved in asthma exacerbations in both children and adults. Some individuals with asthma have increased susceptibility to viral infections and as a result increased rates of asthma exacerbations. We sought to identify a transcriptomic signature in the blood associated with asthma exacerbations triggered by respiratory infections (AETRI) and determine its association with increased risk for asthma exacerbations. METHODS We conducted a two-step study using publicly available, previously generated transcriptomic signatures in peripheral blood mononuclear cells (PBMCs) from asthmatics to identify novel markers of increased risk for asthma exacerbations. In the 1st step, we identified an in vitro PBMC signature in response to rhinovirus. In the 2nd step, we used the in vitro signature to filter PBMC transcripts in response to asthma exacerbations in an independent in vivo cohort. Three different subgroups were identified and studied in the in vivo cohort: 1. Single AETRI; 2. Multiple AETRIs; and 3. Single non-infectious asthma exacerbations. We performed pathway and network analyses in all independent comparisons. We also performed an immunologic gene set enrichment analysis (GSEA) of the comparison between single AETRI and non-infectious asthma exacerbations. RESULTS The in vitro signature identified 4354 differentially expressed genes (DEGs) with a fold change (FC) ≥ 1.2, false discovery rate (FDR) < 0.05. Subsequent analyses filtered by this in vitro signature on an independent cohort of adult asthma identified 238 DEGs (FC≥1.1, FDR < 0.1) in subjects with a single AETRI and no DEGs in single non-infectious asthma exacerbations. A comparison between the response in subjects with single and multiple AETRIs identified two discordant gene subsets. In the largest discordant subset (n = 63 genes) we identified an impaired type I interferon and STAT1 response in multiple AETRIs during the acute phase of the exacerbation and an upregulated STAT1 response at baseline. The STAT1 upregulation at baseline in subjects with multiple AETRIs was accompanied by upregulation of pro-inflammatory molecules including IL-15, interferon-stimulated genes (ISGs), several toll-like receptors 2, - 4, - 5 and - 8 and a triggering receptor expressed on myeloid cells 1 (TREM1) network. CONCLUSIONS Subjects with asthma and multiple AETRIs display a pro-inflammatory signature at baseline, associated with elevated STAT, IL-15 and ISGs, and an impaired STAT1 response during acute asthma exacerbations.
Collapse
Affiliation(s)
- Jose L. Gomez
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Maria P. Diaz
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Gustavo Nino
- Division of Pulmonary and Sleep Medicine, Children’s National Medical Center, Washington, DC USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC USA
- Center for Genetic Medicine, Children’s National Medical Center, Washington, DC USA
| | - Clemente J. Britto
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
17
|
Landgraf-Rauf K, Boeck A, Siemens D, Klucker E, Vogelsang V, Schmidt S, Kunze S, Weissenbacher C, Graessel A, Schmidt-Weber C, von Mutius E, Schedel M, Schaub B. IRF-1 SNPs influence the risk for childhood allergic asthma: A critical role for pro-inflammatory immune regulation. Pediatr Allergy Immunol 2018; 29:34-41. [PMID: 29047170 DOI: 10.1111/pai.12821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/15/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Allergic and non-allergic childhood asthma has been characterized by distinct immune mechanisms. While interferon regulating factor 1 (IRF-1) polymorphisms (SNPs) influence atopy risk, the effect of SNPs on asthma phenotype-specific immune mechanisms is unclear. We assessed whether IRF-1 SNPs modify distinct immune-regulatory pathways in allergic and non-allergic childhood asthma (AA/NA). METHODS In the CLARA study, asthma was characterized by doctor's diagnosis and AA vs NA by positive or negative specific IgE. Children were genotyped for four tagging SNPs within IRF-1 (n = 172). mRNA expression was measured with qRT-PCR. Gene expression was analyzed depending on genetic variants within IRF-1 and phenotype including haplotype estimation and an allelic risk score. RESULTS Carrying the risk alleles of IRF-1 in rs10035166, rs2706384, or rs2070721 was associated with increased risk for AA. Carrying the non-risk allele in rs17622656 was associated with lower risk for AA but not NA. In AA carrying the risk alleles, an increased pro-inflammatory expression of ICAM3, IRF-8, XBP-1, IFN-γ, RGS13, RORC, and TSC2 was observed. NOD2 expression was decreased in AA with risk alleles in rs2706384 and rs10035166 and with risk haplotype. Further, AA with risk haplotype showed increased IL-13 secretion. NA with risk allele in rs2070721 compared to non-risk allele in rs17622656 showed significantly upregulated calcium, innate, mTOR, neutrophil, and inflammatory-associated genes. CONCLUSION IRF-1 polymorphisms influence the risk for childhood allergic asthma being associated with increased pro-inflammatory gene regulation. Thus, it is critical to implement IRF-1 genetics in immune assessment for childhood asthma phenotypes.
Collapse
Affiliation(s)
- Katja Landgraf-Rauf
- Department of Pulmonary & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany.,Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research, Munich, Germany
| | - Andreas Boeck
- Department of Pulmonary & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Diana Siemens
- Department of Pulmonary & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Elisabeth Klucker
- Department of Pulmonary & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Vanessa Vogelsang
- Department of Pulmonary & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Susanne Schmidt
- Department of Pulmonary & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Sonja Kunze
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Claudia Weissenbacher
- Department of Endocrinology, University Children's Hospital Munich, LMU Munich, Munich, Germany
| | - Anke Graessel
- ZAUM - Center of Allergy and Environment, Technische Universität and Helmholtz Center Munich, Munich, Germany
| | - Carsten Schmidt-Weber
- ZAUM - Center of Allergy and Environment, Technische Universität and Helmholtz Center Munich, Munich, Germany
| | - Erika von Mutius
- Department of Pulmonary & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany.,Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research, Munich, Germany
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Bianca Schaub
- Department of Pulmonary & Allergy, University Children's Hospital Munich, LMU Munich, Munich, Germany.,Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research, Munich, Germany
| |
Collapse
|
18
|
Vargas MH, Becerril-Ángeles M, Medina-Reyes IS, Rascón-Pacheco RA. Altitude above 1500 m is a major determinant of asthma incidence. An ecological study. Respir Med 2017; 135:1-7. [PMID: 29414446 DOI: 10.1016/j.rmed.2017.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/17/2017] [Accepted: 12/18/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Previous studies suggest an inverse correlation between asthma and altitude. In the present work, we performed an in-depth analysis of asthma incidence in the 758 Mexican counties covered by the largest medical institution in the country (∼37.5 million insured subjects), and evaluated its relationships with altitude and other factors. METHODS Asthma incidence in each county was calculated from new cases diagnosed by family physicians. Other variables in the same counties, including selected diseases, geographical variables, and socioeconomic factors, were also obtained and their association with asthma was evaluated through bivariate and multivariate analyses. RESULTS Median asthma incidence was 296.2 × 100,000 insured subjects, but tended to be higher in those counties located on or near the coast. When asthma incidence was plotted against altitude, a two-stage pattern was evident: asthma rates were relatively stable in counties located below an altitude of ∼1500 m, while these rates progressively decreased as altitude increased beyond this level (rS = -0.51, p < .001). Multivariate analysis showed that, once each variable was adjusted by the potential influence of the others, asthma incidence was inversely correlated with altitude (standardized β coefficient, -0.577), helminthiasis (-0.173), pulmonary tuberculosis (-0.130), and latitude (-0.126), and was positively correlated with acute respiratory tract infection (0.382), pneumonia (0.289), type 2 diabetes (0.138), population (0.108), and pharyngotonsillitis (0.088), all with a p ≤ .001. CONCLUSION Our study showed that altitude higher than ∼1500 m comprises a major factor in determining asthma incidence, with the risk of new-onset asthma decreasing as altitude increases. Other less influential conditions were also identified.
Collapse
Affiliation(s)
- Mario H Vargas
- Unidad de Investigación Médica en Enfermedades Respiratorias, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico; Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Martín Becerril-Ángeles
- Departamento de Alergia e Inmunología Clínica, Hospital de Especialidades, Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social, Mexico City, Mexico.
| | - Ismael Seth Medina-Reyes
- División de Información Epidemiológica, Coordinación de Vigilancia Epidemiológica y Apoyo en Contingencias, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Ramón Alberto Rascón-Pacheco
- División de Información Epidemiológica, Coordinación de Vigilancia Epidemiológica y Apoyo en Contingencias, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
19
|
Johnston SL. IFN Deficiency in Asthma Attacks. Is Restoring Toll-like Receptor-7 Expression a New Treatment Approach in Severe Asthma? Am J Respir Crit Care Med 2017; 194:1-3. [PMID: 27367880 DOI: 10.1164/rccm.201602-0223ed] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Sebastian L Johnston
- 1 National Heart and Lung Institute Imperial College London London, United Kingdom
| |
Collapse
|
20
|
Gardeux V, Berghout J, Achour I, Schissler AG, Li Q, Kenost C, Li J, Shang Y, Bosco A, Saner D, Halonen MJ, Jackson DJ, Li H, Martinez FD, Lussier YA. A genome-by-environment interaction classifier for precision medicine: personal transcriptome response to rhinovirus identifies children prone to asthma exacerbations. J Am Med Inform Assoc 2017; 24:1116-1126. [PMID: 29016970 PMCID: PMC6080688 DOI: 10.1093/jamia/ocx069] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 05/01/2017] [Accepted: 06/29/2017] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To introduce a disease prognosis framework enabled by a robust classification scheme derived from patient-specific transcriptomic response to stimulation. MATERIALS AND METHODS Within an illustrative case study to predict asthma exacerbation, we designed a stimulation assay that reveals individualized transcriptomic response to human rhinovirus. Gene expression from peripheral blood mononuclear cells was quantified from 23 pediatric asthmatic patients and stimulated in vitro with human rhinovirus. Responses were obtained via the single-subject gene set testing methodology "N-of-1-pathways." The classifier was trained on a related independent training dataset (n = 19). Novel visualizations of personal transcriptomic responses are provided. RESULTS Of the 23 pediatric asthmatic patients, 12 experienced recurrent exacerbations. Our classifier, using individualized responses and trained on an independent dataset, obtained 74% accuracy (area under the receiver operating curve of 71%; 2-sided P = .039). Conventional classifiers using messenger RNA (mRNA) expression within the viral-exposed samples were unsuccessful (all patients predicted to have recurrent exacerbations; accuracy of 52%). DISCUSSION Prognosis based on single time point, static mRNA expression alone neglects the importance of dynamic genome-by-environment interplay in phenotypic presentation. Individualized transcriptomic response quantified at the pathway (gene sets) level reveals interpretable signals related to clinical outcomes. CONCLUSION The proposed framework provides an innovative approach to precision medicine. We show that quantifying personal pathway-level transcriptomic response to a disease-relevant environmental challenge predicts disease progression. This genome-by-environment interaction assay offers a noninvasive opportunity to translate omics data to clinical practice by improving the ability to predict disease exacerbation and increasing the potential to produce more effective treatment decisions.
Collapse
Affiliation(s)
- Vincent Gardeux
- Department of Medicine, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
| | - Joanne Berghout
- Department of Medicine, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
| | - Ikbel Achour
- Department of Medicine, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
| | - A Grant Schissler
- Department of Medicine, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
- Interdisciplinary Program in Statistics, University of Arizona, Tucson, AZ, USA
| | - Qike Li
- Department of Medicine, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
- Interdisciplinary Program in Statistics, University of Arizona, Tucson, AZ, USA
| | - Colleen Kenost
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
| | - Jianrong Li
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
| | - Yuan Shang
- Department of Medicine, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, USA
| | - Anthony Bosco
- Telethon Institute for Child Health Research, Perth, Australia
| | - Donald Saner
- Department of Medicine, University of Arizona, Tucson, AZ, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
- Banner Health, Phoenix, AZ, USA
| | | | - Daniel J Jackson
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, WI, USA
| | - Haiquan Li
- Department of Medicine, University of Arizona, Tucson, AZ, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
| | - Fernando D Martinez
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| | - Yves A Lussier
- Department of Medicine, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
- UA Cancer Center, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
21
|
Xi Y, Troy NM, Anderson D, Pena OM, Lynch JP, Phipps S, Bosco A, Upham JW. Critical Role of Plasmacytoid Dendritic Cells in Regulating Gene Expression and Innate Immune Responses to Human Rhinovirus-16. Front Immunol 2017; 8:1351. [PMID: 29118754 PMCID: PMC5660993 DOI: 10.3389/fimmu.2017.01351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/03/2017] [Indexed: 11/16/2022] Open
Abstract
Though human rhinoviruses (HRVs) are usually innocuous viruses, they can trigger serious consequences in certain individuals, especially in the setting of impaired interferon (IFN) synthesis. Plasmacytoid dendritic cells (pDCs) are key IFN producing cells, though we know little about the role of pDC in HRV-induced immune responses. Herein, we used gene expression microarrays to examine HRV-activated peripheral blood mononuclear cells (PBMCs) from healthy people, in combination with pDC depletion, to assess whether observed gene expression patterns were pDC dependent. As expected, pDC depletion led to a major reduction in IFN-α release. This was associated with profound differences in gene expression between intact PBMC and pDC-depleted PBMC, and major changes in upstream regulators: 70–80% of the HRV activated genes appeared to be pDC dependent. Real-time PCR confirmed key changes in gene expression, in which the following selected genes were shown to be highly pDC dependent: the transcription factor IRF7, both IL-27 chains (IL-27p28 and EBI3), the alpha chain of the IL-15 receptor (IL-15RA) and the IFN-related gene IFI27. HRV-induced IL-6, IFN-γ, and IL-27 protein synthesis were also highly pDC dependent. Supplementing pDC-depleted cultures with recombinant IL-15, IFN-γ, IL-27, or IL-6 was able to restore the IFN-α response, thereby compensating for the absence of pDC. Though pDC comprise only a minority population of migratory leukocytes, our findings highlight the profound extent to which these cells contribute to the immune response to HRV.
Collapse
Affiliation(s)
- Yang Xi
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Niamh M Troy
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Denise Anderson
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Olga M Pena
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jason P Lynch
- Respiratory Immunology Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Simon Phipps
- Respiratory Immunology Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Anthony Bosco
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - John W Upham
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia.,Department of Respiratory Medicine, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| |
Collapse
|
22
|
Boorsma CE, van der Veen TA, Putri KSS, de Almeida A, Draijer C, Mauad T, Fejer G, Brandsma CA, van den Berge M, Bossé Y, Sin D, Hao K, Reithmeier A, Andersson G, Olinga P, Timens W, Casini A, Melgert BN. A Potent Tartrate Resistant Acid Phosphatase Inhibitor to Study the Function of TRAP in Alveolar Macrophages. Sci Rep 2017; 7:12570. [PMID: 28974738 PMCID: PMC5626781 DOI: 10.1038/s41598-017-12623-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 09/13/2017] [Indexed: 12/03/2022] Open
Abstract
The enzyme tartrate resistant acid phosphatase (TRAP, two isoforms 5a and 5b) is highly expressed in alveolar macrophages, but its function there is unclear and potent selective inhibitors of TRAP are required to assess functional aspects of the protein. We found higher TRAP activity/expression in lungs of patients with chronic obstructive pulmonary disease (COPD) and asthma compared to controls and more TRAP activity in lungs of mice with experimental COPD or asthma. Stimuli related to asthma and/or COPD were tested for their capacity to induce TRAP. Receptor activator of NF-κb ligand (RANKL) and Xanthine/Xanthine Oxidase induced TRAP mRNA expression in mouse macrophages, but only RANKL also induced TRAP activity in mouse lung slices. Several Au(III) coordination compounds were tested for their ability to inhibit TRAP activity and [Au(4,4′-dimethoxy-2,2′-bipyridine)Cl2][PF6] (AubipyOMe) was found to be the most potent inhibitor of TRAP5a and 5b activity reported to date (IC50 1.3 and 1.8 μM respectively). AubipyOMe also inhibited TRAP activity in murine macrophage and human lung tissue extracts. In a functional assay with physiological TRAP substrate osteopontin, AubipyOMe inhibited mouse macrophage migration over osteopontin-coated membranes. In conclusion, higher TRAP expression/activity are associated with COPD and asthma and TRAP is involved in regulating macrophage migration.
Collapse
Affiliation(s)
- Carian E Boorsma
- University of Groningen, Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - T Anienke van der Veen
- University of Groningen, Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - Kurnia S S Putri
- University of Groningen, Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute for Pharmacy, Groningen, The Netherlands
| | | | - Christina Draijer
- University of Groningen, Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - Thais Mauad
- São Paulo University, Department of Pathology, São Paulo, Brazil
| | - Gyorgy Fejer
- University of Plymouth, School of Biomedical and Healthcare Sciences, Peninsula Schools of Medicine and Dentistry, Plymouth, United Kingdom
| | - Corry-Anke Brandsma
- University of Groningen, University Medical Center Groningen, Department of Pathology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - Yohan Bossé
- Laval University, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Molecular Medicine, Québec, Canada
| | - Don Sin
- University of British Columbia, James Hogg Research Center, Providence Heart+Lung Institute, St. Paul's Hospital, Vancouver, British Columbia, Canada.,University of British Columbia, Respiratory Division, Department of Medicine, Vancouver, British Columbia, Canada
| | - Ke Hao
- Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Anja Reithmeier
- Karolinska Institute, Department of Laboratory Medicine (LABMED), H5, Division of Pathology, F46, Karolinska University hospital, Huddinge, Stockholm, Sweden
| | - Göran Andersson
- Karolinska Institute, Department of Laboratory Medicine (LABMED), H5, Division of Pathology, F46, Karolinska University hospital, Huddinge, Stockholm, Sweden
| | - Peter Olinga
- University of Groningen, Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute for Pharmacy, Groningen, The Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - Angela Casini
- University of Groningen, Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, Groningen, The Netherlands. .,School of Chemistry, Cardiff University, Cardiff, United Kingdom.
| | - Barbro N Melgert
- University of Groningen, Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, Groningen, The Netherlands. .,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands.
| |
Collapse
|
23
|
Chen ACH, Xi Y, Carroll M, Petsky HL, Gardiner SJ, Pizzutto SJ, Yerkovich ST, Baines KJ, Gibson PG, Hodge S, Masters IB, Buntain HM, Chang AB, Upham JW. Cytokine responses to two common respiratory pathogens in children are dependent on interleukin-1β. ERJ Open Res 2017; 3:00025-2017. [PMID: 29204435 PMCID: PMC5703357 DOI: 10.1183/23120541.00025-2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 07/07/2017] [Indexed: 11/05/2022] Open
Abstract
Protracted bacterial bronchitis (PBB) in young children is a common cause of prolonged wet cough and may be a precursor to bronchiectasis in some children. Although PBB and bronchiectasis are both characterised by neutrophilic airway inflammation and a prominent interleukin (IL)-1β signature, the contribution of the IL-1β pathway to host defence is not clear. This study aimed to compare systemic immune responses against common pathogens in children with PBB, bronchiectasis and control children and to determine the importance of the IL-1β pathway. Non-typeable Haemophilus influenzae (NTHi) stimulation of peripheral blood mononuclear cells (PBMCs) from control subjects (n=20), those with recurrent PBB (n=20) and bronchiectasis (n=20) induced high concentrations of IL-1β, IL-6, interferon (IFN)-γ and IL-10. Blocking with an IL-1 receptor antagonist (IL-1Ra) modified the cellular response to pathogens, inhibiting cytokine synthesis by NTHi-stimulated PBMCs and rhinovirus-stimulated PBMCs (in a separate PBB cohort). Inhibition of IFN-γ production by IL-1Ra was observed across multiple cell types, including CD3+ T cells and CD56+ NK cells. Our findings highlight the extent to which IL-1β regulates the cellular immune response against two common respiratory pathogens. While blocking the IL-1β pathway has the potential to reduce inflammation, this may come at the cost of protective immunity against NTHi and rhinovirus.
Collapse
Affiliation(s)
- Alice C-H Chen
- Diamantina Institute, The University of Queensland, Brisbane, Australia.,These authors contributed equally
| | - Yang Xi
- Diamantina Institute, The University of Queensland, Brisbane, Australia.,These authors contributed equally
| | - Melanie Carroll
- Diamantina Institute, The University of Queensland, Brisbane, Australia
| | - Helen L Petsky
- Queensland University of Technology, CCHR, Brisbane, Australia
| | | | - Susan J Pizzutto
- Child Health Division, Menzies School of Health Research, Charles Darwin Hospital, Darwin, Australia
| | | | | | | | | | - Ian B Masters
- Respiratory and Sleep Medicine, Lady Cilento Children's Hospital, Brisbane, Australia
| | | | - Anne B Chang
- Queensland University of Technology, CCHR, Brisbane, Australia.,Child Health Division, Menzies School of Health Research, Charles Darwin Hospital, Darwin, Australia
| | - John W Upham
- Diamantina Institute, The University of Queensland, Brisbane, Australia
| |
Collapse
|
24
|
Spalluto CM, Singhania A, Cellura D, Woelk CH, Sanchez-Elsner T, Staples KJ, Wilkinson TMA. IFN-γ Influences Epithelial Antiviral Responses via Histone Methylation of the RIG-I Promoter. Am J Respir Cell Mol Biol 2017; 57:428-438. [PMID: 28481620 DOI: 10.1165/rcmb.2016-0392oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The asthmatic lung is prone to respiratory viral infections that exacerbate the symptoms of the underlying disease. Recent work has suggested that a deficient T-helper cell type 1 response in early life may lead to these aberrant antiviral responses. To study the development of long-term dysregulation of innate responses, which is a hallmark of asthma, we investigated whether the inflammatory environment of the airway epithelium can modulate antiviral gene expression via epigenetic mechanisms. We primed AALEB cells, a human bronchial epithelial cell line, with IFN-γ and IL-13, and subsequently infected the cells with respiratory syncytial virus (RSV). We then analyzed the expression of innate antiviral genes and their epigenetic markers. Priming epithelial cells with IFN-γ reduced the RSV viral load. Microarray analysis identified that IFN-γ priming enhanced retinoic acid-inducible gene (RIG)-I mRNA expression, and this expression correlated with epigenetic changes at the RIG-I promoter that influenced its transcription. Using chromatin immunoprecipitation, we observed a reduction of trimethylated histone 3 lysine 9 at the RIG-I promoter. Addition of inhibitor BIX-01294 to this model indicated an involvement of lysine methyltransferase G9a in RIG-I epigenetic regulation. These data suggest that prior exposure to IFN-γ may leave an epigenetic mark on the chromatin that enhances airway cells' ability to resist infection, possibly via epigenetic upregulation of RIG-I. These observations provide further evidence for a crucial role of IFN-γ in the development of mature antiviral responses within a model of respiratory infection. Further clinical validation is required to determine whether this effect in early life leads to changes in antiviral responses associated with asthma.
Collapse
Affiliation(s)
- C Mirella Spalluto
- 1 Clinical and Experimental Sciences and
- 2 Wessex Investigational Sciences Hub, Faculty of Medicine, University of Southampton, and
| | | | | | | | | | - Karl J Staples
- 1 Clinical and Experimental Sciences and
- 2 Wessex Investigational Sciences Hub, Faculty of Medicine, University of Southampton, and
| | - Tom M A Wilkinson
- 1 Clinical and Experimental Sciences and
- 2 Wessex Investigational Sciences Hub, Faculty of Medicine, University of Southampton, and
- 3 Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
25
|
Farne HA, Johnston SL. Immune mechanisms of respiratory viral infections in asthma. Curr Opin Immunol 2017; 48:31-37. [PMID: 28818789 DOI: 10.1016/j.coi.2017.07.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/12/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022]
Abstract
The more severe pathology respiratory viral infections produce in asthma sufferers is a result of a dysregulated immune response. Excess type 2 inflammation is a well-described feature of virally induced asthma exacerbations, with growing evidence that production of antiviral interferons may also be impaired. However, the mechanisms underlying these are little understood. This review summarizes the current understanding and recent discoveries of the cellular and molecular events that follow viral infections in asthma. In particular, we discuss differences in viral sensing and intracellular signalling pathways upstream of interferon induction in asthma, and the role of epithelial-derived cytokines in orchestrating type 2 immunopathology, including type 2 innate lymhpoid cells (ILC2s).
Collapse
Affiliation(s)
- Hugo A Farne
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, UK
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, UK.
| |
Collapse
|
26
|
Airway Epithelial Orchestration of Innate Immune Function in Response to Virus Infection. A Focus on Asthma. Ann Am Thorac Soc 2017; 13 Suppl 1:S55-63. [PMID: 27027954 DOI: 10.1513/annalsats.201507-421mg] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Asthma is a very common respiratory condition with a worldwide prevalence predicted to increase. There are significant differences in airway epithelial responses in asthma that are of particular interest during exacerbations. Preventing exacerbations is a primary aim when treating asthma because they often necessitate unscheduled healthcare visits and hospitalizations and are a significant cause of morbidity and mortality. The most common cause of asthma exacerbations is a respiratory virus infection, of which the most likely type is rhinovirus infection. This article focuses on the role played by the epithelium in orchestrating the innate immune responses to respiratory virus infection. Recent studies show impaired bronchial epithelial cell innate antiviral immune responses, as well as augmentation of a pro-Th2 response characterized by the epithelial-derived cytokines IL-25 and IL-33, crucial in maintaining the Th2 cytokine response to virus infection in asthma. A better understanding of the mechanisms of these abnormal immune responses has the potential to lead to the development of novel therapeutic targets for virus-induced exacerbations. The aim of this article is to highlight current knowledge regarding the role of viruses and immune modulation in the asthmatic epithelium and to discuss exciting areas for future research and novel treatments.
Collapse
|
27
|
Głobińska A, Pawełczyk M, Piechota-Polańczyk A, Olszewska-Ziąber A, Moskwa S, Mikołajczyk A, Jabłońska A, Zakrzewski PK, Brauncajs M, Jarzębska M, Taka S, Papadopoulos NG, Kowalski ML. Impaired virus replication and decreased innate immune responses to viral infections in nasal epithelial cells from patients with allergic rhinitis. Clin Exp Immunol 2016; 187:100-112. [PMID: 27667736 DOI: 10.1111/cei.12869] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2016] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to assess the immune response to parainfluenza virus type 3 (PIV3), rhinovirus 1B (RV1B) and intracellular Toll-like receptors (TLR) agonists in nasal epithelial cells (NECs) from patients with allergic rhinitis and healthy controls. NECs were obtained from eight patients with allergic rhinitis (AR) and 11 non-atopic healthy controls (HC) by nasal scraping, grown to confluence and exposed to PIV3, RV1B infection or TLR-3 and TLR-7/8 agonists. Interferon (IFN)-λ1, IFN-α, IFN-β and regulated on activation, normal T expressed and secreted (RANTES) release into the cell culture supernatants was assessed at 8, 24 and 48 h upon infection or 8 and 24 h after stimulation with poly(I:C) and R848. mRNA levels of IFNs, RANTES, interferon regulatory transcription factor (IRF)3, IRF7 and viral gene copy number were determined using real-time polymerase chain reaction (RT-PCR). PIV3 but not RV1B replication 48 h after infection was significantly lower (P < 0·01) in NECs from AR patients compared to HC. PIV3 infection induced significantly less IFN-λ1 (both protein and mRNA) in NECs from AR compared to HC. IFN-β mRNA expression and RANTES protein release and mRNA expression tended to be smaller in AR compared HC cells in response to both viruses. Stimulation with TLR-3 agonist [poly (I:C)] induced similar IFN-λ1 and RANTES generation in AR and HC subjects. Viral infections in NECs induced IRF7 expression, which correlated with IFN and RANTES expression. These data suggest that virus proliferation rates and the immune response profile are different in nasal epithelial cells from patients with allergic rhinitis compared to healthy individuals.
Collapse
Affiliation(s)
- A Głobińska
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - M Pawełczyk
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - A Piechota-Polańczyk
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - A Olszewska-Ziąber
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - S Moskwa
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland.,Microbiology and Laboratory Medical Immunology Department, Medical University of Łódź, Łódź, Poland
| | - A Mikołajczyk
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - A Jabłońska
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - P K Zakrzewski
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - M Brauncajs
- Microbiology and Laboratory Medical Immunology Department, Medical University of Łódź, Łódź, Poland
| | - M Jarzębska
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - S Taka
- Allergy Department, Second Paediatric Clinic, University of Athens, Athens, Greece
| | - N G Papadopoulos
- Allergy Department, Second Paediatric Clinic, University of Athens, Athens, Greece
| | - M L Kowalski
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| |
Collapse
|
28
|
Abstract
Chronic airway diseases are a significant cause of morbidity and mortality worldwide, and their prevalence is predicted to increase in the future. Respiratory viruses are the most common cause of acute pulmonary infection, and there is clear evidence of their role in acute exacerbations of inflammatory airway diseases such as asthma and chronic obstructive pulmonary disease. Studies have reported impaired host responses to virus infection in these diseases, and a better understanding of the mechanisms of these abnormal immune responses has the potential to lead to the development of novel therapeutic targets for virus-induced exacerbations. The aim of this article is to review the current knowledge regarding the role of viruses and immune modulation in acute exacerbations of chronic pulmonary diseases and to discuss exciting areas for future research and novel treatments.
Collapse
|
29
|
Nuolivirta K, Törmänen S, Teräsjärvi J, Vuononvirta J, Koponen P, Korppi M, Helminen M, Peltola V, He Q. Post-bronchiolitis wheezing is associated with toll-like receptor 9 rs187084 gene polymorphism. Sci Rep 2016; 6:31165. [PMID: 27498757 PMCID: PMC4976338 DOI: 10.1038/srep31165] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/15/2016] [Indexed: 11/29/2022] Open
Abstract
Innate immunity receptors play a critical role in host defence, as well as in allergy and asthma. The aim of this exploratory study was to evaluate whether there are associations between TLR7 rs179008, TLR8 rs2407992, TLR9 rs187084 or TLR10 rs4129009 polymorphisms and viral findings, clinical characteristics or subsequent wheezing in infants with bronchiolitis. In all, 135 full-term infants were hospitalized for bronchiolitis at age less than 6 months: 129 of them were followed-up until the age of 1.5 years. The outcome measures were repeated wheezing, use of inhaled corticosteroids, atopic dermatitis during the first 1.5 years of life and total serum immunoglobulin E (IgE). There were no significant associations between the genotypes or allele frequencies of TLR7 rs179008, TLR8 rs2407992, TLR9 rs187084 or TLR10 rs4129009 polymorphisms and clinical characteristics or the severity of bronchiolitis during hospitalization. During follow-up, repeated wheezing was more common in children with TLR9 rs187084 variant genotype CC (30.5%) than in children with TLR9 wild-type genotype TT (12.2%) (p = 0.02, aOR 2.73, 95% CI 1.02–7.29). The TLR10 rs4129009 minor allele G was associated with elevated total serum IgE. TLR9 rs187084 gene polymorphism may be associated with post-bronchiolitis wheezing, and TLR10 rs4129009 gene polymorphism may be associated with atopy.
Collapse
Affiliation(s)
- Kirsi Nuolivirta
- Department of Pediatrics, Seinäjoki Central Hospital, Seinäjoki, Finland
| | - Sari Törmänen
- Center for Child Health Research, Tampere University and University Hospital, Tampere, Finland
| | - Johanna Teräsjärvi
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Juho Vuononvirta
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Petri Koponen
- Center for Child Health Research, Tampere University and University Hospital, Tampere, Finland
| | - Matti Korppi
- Center for Child Health Research, Tampere University and University Hospital, Tampere, Finland
| | - Merja Helminen
- Center for Child Health Research, Tampere University and University Hospital, Tampere, Finland
| | - Ville Peltola
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and Child and Youth Research Institute, University of Turku, Turku, Finland
| | - Qiushui He
- Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland.,Department of Medical Microbiology, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Peng YH, Fang HY, Wu BR, Kao CH, Chen HJ, Hsia TC, Liao WC. Adult asthma is associated with an increased risk of herpes zoster: A population-based cohort study. J Asthma 2016; 54:250-257. [PMID: 27410999 DOI: 10.1080/02770903.2016.1211142] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The objective of this study was to determine whether a new diagnosis of asthma is associated with a later diagnosis of herpes zoster (HZ) in a nationwide, retrospective, non-age limited, population-based cohort. METHODS We used data from the National Health Insurance Research Database in Taiwan. The asthma group consisted of all 40 069 patients in the database with newly diagnosed asthma and using asthma medications from 2000 through 2005. The nonasthma group comprised 40 069 age- and sex-matched patients without any asthma diagnosis. Cox proportional hazards regression analysis was applied to calculate the hazard ratio of HZ in the patients with asthma relative to those without asthma. RESULTS During a mean follow-up period of 8.77 years, the risk of HZ was 1.48-fold higher in the asthma group compared with that in the nonasthma group after adjustment for sex, age, comorbidities, inhaled and systemic corticosteroid use, and annual outpatient department visits to dermatologists. Additional stratified analyses revealed that the risk of HZ was significantly higher in patients of both sexes and those aged older than 21 years. CONCLUSIONS Newly diagnosed adult patients with asthma have a significantly higher risk of developing HZ than do those without asthma.
Collapse
Affiliation(s)
- Yi-Hao Peng
- a Department of Respiratory Therapy , Asia University Hospital, Asia University , Taichung , Taiwan.,b Department of Public Health , China Medical University , Taichung , Taiwan.,c Department of Respiratory Therapy , China Medical University , Taichung , Taiwan
| | - Hsin-Yu Fang
- d Department of Dermatology , China Medical University Hospital, China Medical University , Taichung , Taiwan
| | - Biing-Ru Wu
- e Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, China Medical University , Taichung , Taiwan
| | - Chia-Hung Kao
- f Graduate Institute of Clinical Medical Science and School of Medicine, College of Medicine, China Medical University , Taichung , Taiwan.,g Department of Nuclear Medicine and PET Center , China Medical University Hospital, China Medical University , Taichung , Taiwan
| | - Hsuan-Ju Chen
- h Management Office for Health Data, China Medical University Hospital, China Medical University Hospital , Taichung , Taiwan
| | - Te-Chun Hsia
- c Department of Respiratory Therapy , China Medical University , Taichung , Taiwan.,e Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, China Medical University , Taichung , Taiwan
| | - Wei-Chih Liao
- e Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, China Medical University , Taichung , Taiwan
| |
Collapse
|
31
|
Holding the Inflammatory System in Check: TLRs and Their Targeted Therapy in Asthma. Mediators Inflamm 2016; 2016:2180417. [PMID: 27274620 PMCID: PMC4870363 DOI: 10.1155/2016/2180417] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/13/2016] [Indexed: 02/06/2023] Open
Abstract
Inflammation is a complex biological response to detrimental stimuli and can be a double-edged sword. Inflammation plays a protective role in removing pathogenic factors, but dysregulated inflammation is associated with several major fatal diseases such as asthma, cancer, and cardiovascular diseases. Asthma is a complex heterogenous disease caused by genetic and environmental factors. TLRs are the primary proteins associated with the innate and adaptive immune responses to these fatal factors and play an important role in recognizing pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), which initiates the downstream immune response. Due to the complex TLRs cascade and nowadays unsuccessful control in asthma, new studies are focused on TLRs and other potential targets in TLR cascade to minimize airway inflammation.
Collapse
|
32
|
Hall SC, Agrawal DK. Toll-like receptors, triggering receptor expressed on myeloid cells family members and receptor for advanced glycation end-products in allergic airway inflammation. Expert Rev Respir Med 2016; 10:171-84. [PMID: 26678062 PMCID: PMC4955846 DOI: 10.1586/17476348.2016.1133303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Asthma is a chronic disorder of the airways characterized by cellular infiltration, airway hyper-responsive and airway inflammation. Innate immune cells are the first line of defense against endogenous and exogenous signals in the airways and as such possess a diverse array of pattern recognition receptors. Toll-like receptors are crucial sentinels which when activated, can either promote or ameliorate the inflammatory response in predisposed individuals. The recently discovered triggering receptor expressed on myeloid cells family members are emerging mediators of inflammation. These receptors are believed to modulate inflammatory responses by collaborating with classic PRRs. Endogenous signals like HMGB-1, signaling through the receptor for advanced glycation end products, also promotes inflammation, however, its contribution to inflammation in the airways is not well known. Here, we discuss the role of each receptor in airway inflammation and highlight potential synergistic mechanisms, which contribute to disease pathogenesis in allergic asthma.
Collapse
Affiliation(s)
- Sannette C. Hall
- Department of Biomedical Science, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K. Agrawal
- Department of Biomedical Science, Creighton University School of Medicine, Omaha, NE, USA
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE, USA
- Center for Clinical and Translational Science Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
33
|
Human rhinovirus-induced inflammatory responses are inhibited by phosphatidylserine containing liposomes. Mucosal Immunol 2016; 9:1303-16. [PMID: 26906404 PMCID: PMC4883656 DOI: 10.1038/mi.2015.137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 11/25/2015] [Indexed: 02/04/2023]
Abstract
Human rhinovirus (HRV) infections are major contributors to the healthcare burden associated with acute exacerbations of chronic airway disease, such as chronic obstructive pulmonary disease and asthma. Cellular responses to HRV are mediated through pattern recognition receptors that may in part signal from membrane microdomains. We previously found Toll-like receptor signaling is reduced, by targeting membrane microdomains with a specific liposomal phosphatidylserine species, 1-stearoyl-2-arachidonoyl-sn-glycero-3-phospho-L-serine (SAPS). Here we explored the ability of this approach to target a clinically important pathogen. We determined the biochemical and biophysical properties and stability of SAPS liposomes and studied their ability to modulate rhinovirus-induced inflammation, measured by cytokine production, and rhinovirus replication in both immortalized and normal primary bronchial epithelial cells. SAPS liposomes rapidly partitioned throughout the plasma membrane and internal cellular membranes of epithelial cells. Uptake of liposomes did not cause cell death, but was associated with markedly reduced inflammatory responses to rhinovirus, at the expense of only modest non-significant increases in viral replication, and without impairment of interferon receptor signaling. Thus using liposomes of phosphatidylserine to target membrane microdomains is a feasible mechanism for modulating rhinovirus-induced signaling, and potentially a prototypic new therapy for viral-mediated inflammation.
Collapse
|
34
|
Simpson JL, Carroll M, Yang IA, Reynolds PN, Hodge S, James AL, Gibson PG, Upham JW. Reduced Antiviral Interferon Production in Poorly Controlled Asthma Is Associated With Neutrophilic Inflammation and High-Dose Inhaled Corticosteroids. Chest 2015; 149:704-13. [PMID: 26836898 DOI: 10.1016/j.chest.2015.12.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/26/2015] [Accepted: 12/01/2015] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Asthma is a heterogeneous chronic inflammatory disease in which host defense against respiratory viruses such as human rhinovirus (HRV) may be abnormal. This is a matter of some controversy, with some investigators reporting reduced type I interferon (IFN) synthesis and others suggesting that type I IFN synthesis is relatively normal in asthma. OBJECTIVE The objective of this study was to examine the responsiveness of circulating mononuclear cells to HRV in a large cohort of participants with poorly controlled asthma and determine whether IFN-α and IFN-β synthesis varies across different inflammatory phenotypes. METHODS Eligible adults with asthma (n = 86) underwent clinical assessment, sputum induction, and blood sampling. Asthma inflammatory subtypes were defined by sputum cell count, and supernatant assessed for IL-1β. Peripheral blood mononuclear cells (PBMCs) were exposed to HRV serotype 1b, and IFN-α and IFN-β release was measured by enzyme-linked immunosorbent assay. RESULTS Participants (mean age, 59 years; atopy, 76%) had suboptimal asthma control (mean asthma control questionnaire 6, 1.7). In those with neutrophilic asthma (n = 12), HRV1b-stimulated PBMCs produced significantly less IFN-α than PBMCs from participants with eosinophilic (n = 35) and paucigranulocytic asthma (n = 35). Sputum neutrophil proportion and the dose of inhaled corticosteroids were independent predictors of reduced IFN-α production after HRV1b exposure. CONCLUSIONS Antiviral type I IFN production is impaired in those with neutrophilic airway inflammation and in those prescribed high doses of inhaled corticosteroids. Our study is an important step toward identifying those with poorly controlled asthma who might respond best to inhaled IFN therapy during exacerbations.
Collapse
Affiliation(s)
- Jodie L Simpson
- Department of Respiratory and Sleep Medicine, Hunter Medical Research Institute, New Lambton, NSW, Australia; Priority Research Centre for Asthma and Respiratory Disease, The University of Newcastle, Callaghan, NSW, Australia.
| | - Melanie Carroll
- School of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Ian A Yang
- School of Medicine, The University of Queensland, St Lucia, QLD, Australia; Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, QLD, Australia
| | - Paul N Reynolds
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia; Lung Research Laboratory, Hanson Institute, Adelaide, SA, Australia
| | - Sandra Hodge
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia; Lung Research Laboratory, Hanson Institute, Adelaide, SA, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, WA, Australia; School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia
| | - Peter G Gibson
- Department of Respiratory and Sleep Medicine, Hunter Medical Research Institute, New Lambton, NSW, Australia; Priority Research Centre for Asthma and Respiratory Disease, The University of Newcastle, Callaghan, NSW, Australia; Woolcock Institute of Medical Research, Sydney, NSW, Australia
| | - John W Upham
- School of Medicine, The University of Queensland, St Lucia, QLD, Australia; Department of Respiratory Medicine, Princess Alexandra Hospital, Woolloongabba, Brisbane, QLD, Australia
| |
Collapse
|
35
|
Hui DS, Lee N, Chan PKS. Update in Viral Infections 2014. Am J Respir Crit Care Med 2015; 192:676-81. [PMID: 26371813 DOI: 10.1164/rccm.201503-0637up] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- David S Hui
- 1 Department of Medicine and Therapeutics.,2 Stanley Ho Center for Emerging Infectious Diseases, and
| | - Nelson Lee
- 1 Department of Medicine and Therapeutics.,2 Stanley Ho Center for Emerging Infectious Diseases, and
| | - Paul K S Chan
- 2 Stanley Ho Center for Emerging Infectious Diseases, and.,3 Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong
| |
Collapse
|
36
|
Rhinovirus stimulated IFN-α production: how important are plasmacytoid DCs, monocytes and endosomal pH? Clin Transl Immunology 2015; 4:e46. [PMID: 26682054 PMCID: PMC4673444 DOI: 10.1038/cti.2015.27] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 09/30/2015] [Accepted: 09/30/2015] [Indexed: 01/05/2023] Open
Abstract
Human rhinovirus (HRV) infection is a major cause of asthma exacerbations, which appears to be linked to a defective innate immune response to infection. Although the type I interferons (IFN-α and IFN-β) have a critical role in protecting against most viral infections, the cells responsible for IFN production in response to HRV and the relative importance of pattern recognition receptors located in endosomes has not been fully elucidated. In the current study we demonstrate that, using intracellular flow cytometry, >90% of the IFN-α-producing cells in human blood mononuclear cells following HRV16 exposure are plasmacytoid dendritic cells, whereas monocytes and myeloid dendritic cells contribute only 10% and <1%, respectively, of the IFN-α production. Bafilomycin and chloroquine, agents that inhibit the function of endosomal toll-like receptors (TLRs), significantly reduced the capacity of TLR3-, TLR7- and TLR-9-stimulated cells to produce IFN-α and the IFN-induced chemokine CXCL10 (IP-10). In contrast, only bafilomycin (but not chloroquine) effectively suppressed HRV16-stimulated IFN-α and IP-10 production, whereas neither bafilomycin or chloroquine inhibited HRV16-stimulated interleukin-6 release. Attempts to block IFN-α production with commercially available TLR-specific oligonucleotides were unsuccessful due to major ‘off-target' effects. These findings suggest that among circulating haemopoietic cells, plasmacytoid dendritic cells and TLRs located within endosomes are critical for inducing efficient IFN-I production in response to HRVs.
Collapse
|
37
|
Understanding the Association of Human Rhinovirus with Asthma. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 23:6-10. [PMID: 26376925 DOI: 10.1128/cvi.00414-15] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Human rhinoviruses are ubiquitous seasonal pathogens. They have known associations with first onset of wheezing illnesses in children and with asthma exacerbations in patients of all ages. It is not yet certain whether human rhinoviruses play a direct role in the pathogenesis of asthma by activating deleterious inflammatory responses or if they only serve as a catalyst to accelerate the disease in genetically predisposed individuals. There have been previously demonstrated reductions in the development of the asthmatic phenotype with passive immunization against respiratory syncytial virus; however, in the case of rhinovirus, there are barriers to effective vaccine development, such as the lack of a common antigenic target due to alterations of surface markers among subtypes. It remains to be determined whether certain subtypes of human rhinovirus are more asthmagenic and therefore worthy of greater attention as vaccine candidates, but several studies have suggested that RV-C and certain RV-A strains may be more strongly linked with asthma.
Collapse
|
38
|
Gonzales-van Horn SR, Farrar JD. Interferon at the crossroads of allergy and viral infections. J Leukoc Biol 2015; 98:185-94. [PMID: 26026068 PMCID: PMC4501675 DOI: 10.1189/jlb.3ru0315-099r] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/07/2015] [Accepted: 05/12/2015] [Indexed: 01/01/2023] Open
Abstract
IFN-α/β was first described as a potent inhibitor of viral replication, but it is now appreciated that IFN signaling plays a pleiotropic role in regulating peripheral T cell functions. Recently, IFN-α/β was shown to block human Th2 development by suppressing the transcription factor GATA3. This effect is consistent with the role for IFN-α/β in suppressing allergic inflammatory processes by blocking granulocyte activation and IL-4-mediated B cell isotype switching to IgE. With the consideration of recent studies demonstrating a defect in IFN-α/β secretion in DCs and epithelial cells from individuals with severe atopic diseases, there is an apparent reciprocal negative regulatory loop in atopic individuals, whereby the lack of IFN-α/β secretion by innate cells contributes to the development of allergic Th2 cells. Is it possible to overcome these events by treating with IFN-α/β or by inducing its secretion in vivo? In support of this approach, case studies have documented the therapeutic potential of IFN-α/β in treating steroid-resistant allergic asthma and other atopic diseases. Additionally, individuals with asthma who are infected with HCV and respond to IFN therapy showed a reduction in symptoms and severity of asthma attacks. These findings support a model, whereby allergic and antiviral responses are able to cross-regulate each other, as IgER cross-linking of pDCs prevents IFN-α/β production in response to viral infection. The clinical importance of upper-respiratory viruses in the context of allergic asthma supports the need to understand how these pathways intersect and to identify potential therapeutic targets.
Collapse
Affiliation(s)
| | - J David Farrar
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
39
|
Kim SH, Lim KH, Park HK, Lee SY, Kim SH, Kang HR, Park HW, Chang YS, Cho SH. Reduced IRF7 response to rhinovirus unrelated with DNA methylation in peripheral mononuclear cells of adult asthmatics. Asia Pac Allergy 2015; 5:114-22. [PMID: 25938076 PMCID: PMC4415177 DOI: 10.5415/apallergy.2015.5.2.114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 04/12/2015] [Indexed: 02/04/2023] Open
Abstract
Background Human rhinoviruses are the major cause of asthma exacerbation in both children and adults. Recently, impaired antiviral interferon (IFN) response in asthmatics has been indicated as a primary reason of the susceptibility to respiratory virus, but the mechanism of defective IFN production is little understood to date. The expression of IFN regulatory factor 7 (IRF7), a transcriptional factor for virus-induced type I IFN production is known to be regulated epigenetically by DNA methylation. Objective We aimed to investigate the expression of IFN-α, IFN-β, and IRF7 in response to rhinovirus infection in the adult asthmatics and evaluate DNA methylation status of IRF7 gene promotor. Methods Twenty symptomatic adult asthmatics and 10 healthy subjects were enrolled and peripheral blood was collected from each subject. Peripheral blood mononuclear cells (PBMCs) were isolated, cultured, and ex vivo stimulated with rhinovirus-16. The mRNA expressions of IFN-α, IFN-β, and IRF7 were analyzed using real time quantitative polymerase chain reaction. Genomic DNA was isolated from untreated PBMCs and the methylation status of IRF7 gene promotor was investigated using bisulfite pyrosequencing. Results The mean age of asthmatics was 45.4 ± 15.7 years and 40% was male, which were not different with those of control group. Asthmatics showed significantly decreased mRNA expressions (relative expression to beta-actin) of IFN-α and IFN-β compared with normal control. The mRNA expression of IRF7 in the asthmatics was also significantly lower than those in the normal control. No significant difference of DNA methylation was observed between asthmatics and controls in all analyzed positions of IRF7 promotor CpG loci. Conclusion The mRNA expression of type I IFN in response to rhinovirus was impaired in the PBMCs of adult asthmatics. The mRNA expression of IRF7, transcriptional factor inducing type I IFN was also reduced, but not caused by altered DNA methylation in the IRF7 gene promotor.
Collapse
Affiliation(s)
- Sae-Hoon Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea. ; Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 463-707, Korea
| | - Kyung-Hwan Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea
| | - Han-Ki Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea
| | - Suh-Young Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea. ; Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 463-707, Korea
| | - Soon-Hee Kim
- Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam 463-707, Korea
| | - Hye-Ryun Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea
| | - Heung-Woo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea
| | - Yoon-Seok Chang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea. ; Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 463-707, Korea
| | - Sang-Heon Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea
| |
Collapse
|
40
|
Phenotypic responses of differentiated asthmatic human airway epithelial cultures to rhinovirus. PLoS One 2015; 10:e0118286. [PMID: 25706956 PMCID: PMC4338293 DOI: 10.1371/journal.pone.0118286] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 01/12/2015] [Indexed: 12/19/2022] Open
Abstract
Objectives Human airway epithelial cells are the principal target of human rhinovirus (HRV), a common cold pathogen that triggers the majority of asthma exacerbations. The objectives of this study were 1) to evaluate an in vitro air liquid interface cultured human airway epithelial cell model for HRV infection, and 2) to identify gene expression patterns associated with asthma intrinsically and/or after HRV infection using this model. Methods Air-liquid interface (ALI) human airway epithelial cell cultures were prepared from 6 asthmatic and 6 non-asthmatic donors. The effects of rhinovirus RV-A16 on ALI cultures were compared. Genome-wide gene expression changes in ALI cultures following HRV infection at 24 hours post exposure were further analyzed using RNA-seq technology. Cellular gene expression and cytokine/chemokine secretion were further evaluated by qPCR and a Luminex-based protein assay, respectively. Main Results ALI cultures were readily infected by HRV. RNA-seq analysis of HRV infected ALI cultures identified sets of genes associated with asthma specific viral responses. These genes are related to inflammatory pathways, epithelial structure and remodeling and cilium assembly and function, including those described previously (e.g. CCL5, CXCL10 and CX3CL1, MUC5AC, CDHR3), and novel ones that were identified for the first time in this study (e.g. CCRL1). Conclusions ALI-cultured human airway epithelial cells challenged with HRV are a useful translational model for the study of HRV-induced responses in airway epithelial cells, given that gene expression profile using this model largely recapitulates some important patterns of gene responses in patients during clinical HRV infection. Furthermore, our data emphasize that both abnormal airway epithelial structure and inflammatory signaling are two important asthma signatures, which can be further exacerbated by HRV infection.
Collapse
|