1
|
Yao Y, Yang Y, Ji M, Qin Q, Xu K, Xia Z, Liu H, Yuan L, Yuan Y, Qin L, Du X, Wang L, Zhou K, Wu X, Wang W, Qing B, Xiang Y, Qu X, Yang M, Qin X, Liu C. Airway epithelial-derived exosomes induce acute asthma exacerbation after respiratory syncytial virus infection. MedComm (Beijing) 2024; 5:e621. [PMID: 38938285 PMCID: PMC11208743 DOI: 10.1002/mco2.621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/29/2024] Open
Abstract
Acute asthma exacerbation refers to the progressive deterioration of asthma symptoms that is always triggered by virus infection represented by respiratory syncytial virus (RSV). After RSV infection, exaggerated Th2-mediated pulmonary inflammation is the critical pathological response of asthmatic patients with acute exacerbation. Significantly, airway epithelial cells, being the primary targets of RSV infection, play a crucial role in controlling the pulmonary inflammatory response by releasing airway epithelial cell-derived exosomes (AEC-Exos), which potentially influence the development of asthma. However, the specific role of AEC-Exos in acute asthma exacerbation after RSV infection remains obscure. The purpose of this study was to determine the distinct function of AEC-Exos in exacerbating acute asthma following RSV infection. Blockade of exosomes by GW reduce the enhanced pulmonary inflammation significantly. Specifically, the enhanced Th2 inflammation was induced by AEC-Exos thorough transportation of hsa-miR-155-5p-Sirtuin 1 (SIRT1) pathway during acute asthma exacerbation. Targeted inhibition of hsa-miR-155-5p blocks the exaggerated Th2 inflammation effectively in mice with acute asthma exacerbation. In summary, our study showed that during acute asthma exacerbation after RSV infection, AEC-Exos promote the enhanced Th2 inflammation through transportation of increased hsa-miR-155-5p, which was mediated partly through SIRT1-mediated pathway. hsa-miR-155-5p is a potential biomarker for early prediction of acute asthma exacerbation.
Collapse
Affiliation(s)
- Ye Yao
- Department of Respiratory MedicineNational Clinical Research Center for Respiratory DiseasesXiangya HospitalCentral South UniversityChangshaChina
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Yu Yang
- Department of Respiratory MedicineNational Clinical Research Center for Respiratory DiseasesXiangya HospitalCentral South UniversityChangshaChina
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Ming Ji
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Qingwu Qin
- Department of Pulmonary and Critical Care Medicinethe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Kun Xu
- Department of preventive medicine, School of MedicineHunan Normal UniversityChangshaChina
| | - Zhenkun Xia
- Department of Thoracic Surgerythe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Huijun Liu
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Lin Yuan
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Yunchang Yuan
- Department of Thoracic Surgerythe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Ling Qin
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
- Basic and Clinical Research Laboratory of Major Respiratory DiseasesCentral South UniversityChangshaHunanChina
| | - Xizi Du
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Leyuan Wang
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Kai Zhou
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Xinyu Wu
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Weijie Wang
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Bei Qing
- Department of Thoracic Surgerythe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yang Xiang
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Xiangping Qu
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Ming Yang
- Centre for Asthma and Respiratory DiseaseSchool of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of Newcastle and Hunter Medical Research InstituteCallaghanNew South WalesAustralia
| | - Xiaoqun Qin
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
| | - Chi Liu
- Department of Respiratory MedicineNational Clinical Research Center for Respiratory DiseasesXiangya HospitalCentral South UniversityChangshaChina
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaHunanChina
- Basic and Clinical Research Laboratory of Major Respiratory DiseasesCentral South UniversityChangshaHunanChina
| |
Collapse
|
2
|
Macchione M, Yoshizaki K, Frias DP, Maier K, Smelan J, Prado CM, Mauad T. Fragrances as a trigger of immune responses in different environments. Toxicol In Vitro 2024; 96:105769. [PMID: 38142785 DOI: 10.1016/j.tiv.2023.105769] [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: 08/03/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Fragrances can cause allergic skin reactions, expressed as allergic contact dermatitis and reactions in the respiratory tract that range from acute temporary upper airway irritation to obstructive lung disease. These adverse health effects may result from the stimulation of a specific (adaptive) immune response. Th1 cells, which essentially produce interleukin-2 (IL-2) and interferon-γ (IFN-γ), play a key role in allergic contact dermatitis and also on allergic sensitization to common allergens (e.g., nickel and fragrance). It has been shown that fragrance allergy leads to Th2/Th22 production of IL-4, IL-5 and IL-13, controlling the development of IgE and mediating hypersensitivity reactions in the lung, such as asthma. Cytokines released during immune response modulate the expression of cytochrome P450 (CYPs) proteins, which can result in alterations of the pharmacological effects of substances in inflammatory diseases. The mechanisms linking environment and immunity are still not completely understood but it is known that aryl hydrocarbon receptor (AhR) is a sensor with conserved ligand-activated transcription factor, highly expressed in cells that controls complex transcriptional programs which are ligand and cell type specific, with CYPs as targeted genes. This review focuses on these important aspects of immune responses of the skin and respiratory tract cells, describing some in vitro models applied to evaluate the mechanisms involved in fragrance-induced allergy.
Collapse
Affiliation(s)
- M Macchione
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil.
| | - K Yoshizaki
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| | - D P Frias
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| | - K Maier
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| | - J Smelan
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| | - C M Prado
- Federal University of Sao Paulo, Santos, Brazil
| | - T Mauad
- Laboratory of Experimental Environmental Pathology, Department of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| |
Collapse
|
3
|
Chen Y, Zhu M, Hu J, He S, Li S, Liu B, Yang J. IL-27 Alleviates Airway Inflammation and Airway Hyperresponsiveness in Asthmatic Mice by Targeting the CD39/ATP Axis of Dendritic Cells. Inflammation 2024; 47:807-821. [PMID: 38117410 DOI: 10.1007/s10753-023-01945-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/18/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
Interleukin-27 receptor (IL-27R) is expressed in a variety of immune cells and structural cells, including dendritic cells. The mechanism of IL-27 in asthma has not been fully elucidated. This study aimed to examine whether IL-27 regulated the CD39/ATP axis of dendritic cells in asthma. Our results showed that in ovalbumin (OVA)-induced asthma mouse model, IL-27Rα-/- asthmatic mice showed increased airway resistance, increased infiltration of inflammatory cells in lung tissue, proliferation of goblet cells, enhanced expression of Muc5 AC around airway epithelium, increased total number of cells and eosinophils, increased levels of total IgE, OVA-IgE, IL-4, IL-5, IL-13 and IL-17 A, and increased expression of transcription factors GATA-3 and RORγt in lung tissue. The expression of CD39 mRNA and protein in the lung tissue of IL-27Rα-/- asthmatic mice decreased, and the expression of NLRP3, ASC and Caspase-1 in NLRP3 inflammasome components increased. The concentration of ATP was significantly increased compared with WT asthmatic mice. In vitro experiments showed that the expression of CD39 in lung dendritic cells of IL-27Rα-/- asthmatic mice decreased, while the expression of NLRP3 inflammasome components NLRP3, ASC and Caspase-1 increased. These findings indicate that IL-27 directly and indirectly regulates immunoinflammatory responses in asthma by acting on dendritic cells CD39/ATP Axis.
Collapse
Affiliation(s)
- Yifei Chen
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Miaojuan Zhu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiahao Hu
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shaojun He
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shuhua Li
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bing Liu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiong Yang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
Zhai Z, Shao L, Lu Z, Yang Y, Wang J, Liu Z, Wang H, Zheng Y, Lu H, Song X, Zhang Y. Characteristics of mucin hypersecretion in different inflammatory patterns based on endotypes of chronic rhinosinusitis. Clin Transl Allergy 2024; 14:e12334. [PMID: 38282195 PMCID: PMC10802810 DOI: 10.1002/clt2.12334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is usually accompanied by mucin hypersecretion that can lead to mucus accumulation and impair nasal mucociliary clearance, thus exacerbating airway inflammation. Abnormal mucin hypersecretion is regulated by different T helper (Th) cytokines, which are associated with different endotype-driven inflammatory responses. Therefore, it is of great significance to understand how these factors regulate mucin hypersecretion to provide precise treatment strategies for different endotypes of CRS. BODY: Thus far, the most common endotypes of CRS are classified as type 1, type 2, or type 3 immune responses based on innate and adaptive cell-mediated effector immunity, and the representative Th cytokines in these immune responses, such as IFN-γ, TNF-α, IL-4, IL-5, IL-13, IL-10, IL-17, and IL-22, play an important regulatory role in mucin secretion. We reviewed all the related literature in the PubMed database to determine the expression of these Th cytokines in CRS and the role they play in the regulation of mucin secretion. CONCLUSION We believe that the main Th cytokines involved in specific endotypes of CRS play a key role in regulating abnormal mucin secretion, which contributes to better understanding of the pathogenesis of CRS and provides therapeutic targets for airway inflammatory diseases associated with mucin hypersecretion.
Collapse
Affiliation(s)
- Zhaoxue Zhai
- Second Clinical Medicine CollegeBinzhou Medical UniversityYantaiChina
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Liting Shao
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Zhaoyang Lu
- Second Clinical Medicine CollegeBinzhou Medical UniversityYantaiChina
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Yujuan Yang
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
- Yantai Key Laboratory of Otorhinolaryngologic DiseasesYantaiChina
| | - Jianwei Wang
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
- Yantai Key Laboratory of Otorhinolaryngologic DiseasesYantaiChina
| | - Zhen Liu
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Huikang Wang
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Yang Zheng
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Haoran Lu
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
| | - Xicheng Song
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
- Yantai Key Laboratory of Otorhinolaryngologic DiseasesYantaiChina
| | - Yu Zhang
- Department of OtolaryngologyHead and Neck Surgery, Yantai Yuhuangding HospitalQingdao UniversityYantaiChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiChina
- Yantai Key Laboratory of Otorhinolaryngologic DiseasesYantaiChina
| |
Collapse
|
5
|
Carroll OR, Pillar AL, Brown AC, Feng M, Chen H, Donovan C. Advances in respiratory physiology in mouse models of experimental asthma. Front Physiol 2023; 14:1099719. [PMID: 37008013 PMCID: PMC10060990 DOI: 10.3389/fphys.2023.1099719] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/07/2023] [Indexed: 03/18/2023] Open
Abstract
Recent advances in mouse models of experimental asthma coupled with vast improvements in systems that assess respiratory physiology have considerably increased the accuracy and human relevance of the outputs from these studies. In fact, these models have become important pre-clinical testing platforms with proven value and their capacity to be rapidly adapted to interrogate emerging clinical concepts, including the recent discovery of different asthma phenotypes and endotypes, has accelerated the discovery of disease-causing mechanisms and increased our understanding of asthma pathogenesis and the associated effects on lung physiology. In this review, we discuss key distinctions in respiratory physiology between asthma and severe asthma, including the magnitude of airway hyperresponsiveness and recently discovered disease drivers that underpin this phenomenon such as structural changes, airway remodeling, airway smooth muscle hypertrophy, altered airway smooth muscle calcium signaling, and inflammation. We also explore state-of-the-art mouse lung function measurement techniques that accurately recapitulate the human scenario as well as recent advances in precision cut lung slices and cell culture systems. Furthermore, we consider how these techniques have been applied to recently developed mouse models of asthma, severe asthma, and asthma-chronic obstructive pulmonary disease overlap, to examine the effects of clinically relevant exposures (including ovalbumin, house dust mite antigen in the absence or presence of cigarette smoke, cockroach allergen, pollen, and respiratory microbes) and to increase our understanding of lung physiology in these diseases and identify new therapeutic targets. Lastly, we focus on recent studies that examine the effects of diet on asthma outcomes, including high fat diet and asthma, low iron diet during pregnancy and predisposition to asthma development in offspring, and environmental exposures on asthma outcomes. We conclude our review with a discussion of new clinical concepts in asthma and severe asthma that warrant investigation and how we could utilize mouse models and advanced lung physiology measurement systems to identify factors and mechanisms with potential for therapeutic targeting.
Collapse
Affiliation(s)
- Olivia R. Carroll
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Amber L. Pillar
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Alexandra C. Brown
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Min Feng
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Hui Chen
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Chantal Donovan
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
- *Correspondence: Chantal Donovan,
| |
Collapse
|
6
|
CTNNAL1 enhances glucocorticoid sensitivity in HDM-induced asthma mouse model through deactivating hsp90 signaling pathway. Life Sci 2023; 313:121304. [PMID: 36535402 DOI: 10.1016/j.lfs.2022.121304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
AIMS Adhesion molecules play vital roles in the induction of airway hyperresponsiveness (AHR) or airway inflammation. The down-regulation of catenin alpha-like 1 (CTNNAL1) in the bronchial epithelial cells of asthma patients and mice models has been noted in our previous study. In this work, we further explore the underlying mechanism of CTNNAL1 in asthma. MAIN METHODS We constructed a house dust mite (HDM)-induced asthma animal model on control mice and applied CTNNAL1-siRNA transfection to create CTNNAL1-deficient mice. KEY FINDINGS We documented much more severe airway inflammation and increased leukocyte infiltration in the lungs of the CTNNAL1-deficient mice comparing to control mice, along with elevated expression of inflammatory cytokines. Dexamethasone (DEX) treatment led to less reduced inflammation in CTNNAL1-deficient mice compared with control mice. Immunoprecipitation confirmed the interaction between heat shock protein90 (hsp90) and CTNNAL1. The expression of hsp90 was upregulated after CTNNAL1 silencing. Meanwhile, the use of hsp90 inhibitor geldanamycin significantly decreased the expression of NR3C1, ICAM-1 and the ratio of p-p65/p65 in CTNNAL1-silenced 16HBE14o- cells. Both geldanamycin and DEX could function to suppress the expression of ICAM-1 and the phosphorylation level of p65. Nevertheless, the anti-inflammatory effect of DEX proved less potent than geldanamycin in the CTNNAL1-silenced group. The combined therapy of geldanamycin and DEX significantly decreased the inflammatory responses in CTNNAL1-deficient HBE cells than DEX monotherapy. SIGNIFICANCE Our study corroborates that CTNNAL1 deficiency induced aggravated airway inflammation and rendered insensitivity to glucocorticoids via triggering hsp90 signaling pathway.
Collapse
|
7
|
Bryant N, Muehling LM. T-cell responses in asthma exacerbations. Ann Allergy Asthma Immunol 2022; 129:709-718. [PMID: 35918022 PMCID: PMC9987567 DOI: 10.1016/j.anai.2022.07.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Asthma is a chronic lung disease comprising multiple endotypes and characterized by periodic exacerbations. A diverse array of T cells has been found to contribute to all endotypes of asthma in pathogenic and regulatory roles. Here, we review the contributions of CD4+, CD8+, and unconventional T cells in allergic and nonallergic asthma. DATA SOURCES Review of published literature pertaining to conventional and unconventional T-cell types in asthma. STUDY SELECTIONS Recent peer-reviewed articles pertaining to T cells in asthma, with additional peer-reviewed studies for context. RESULTS Much research in asthma has focused on the roles of CD4+ TH cells. Roles for TH2 cells in promoting allergic asthma pathogenesis have been well-described, and the recent description of pathogenic TH2A cells provides additional insight into these responses. Other TH types, notably TH1 and TH17, have been linked to neutrophilic and steroid-resistant asthma phenotypes. Beyond CD4+ T cells, CD8+ Tc2 cells are also strongly associated with allergic asthma. An emerging area for study is unconventional T-cell types, including γδT, invariant natural killer T, and mucosal-associated invariant T cells. Although data in asthma remain limited for these cells, their ability to bridge innate and adaptive responses likely makes them key players in asthma. A number of asthma therapies target T-cell responses, and, although data are limited, they seem to modulate T-cell populations. CONCLUSION Given the diversity and heterogeneity of asthma and T-cell responses, there remain many rich avenues for research to better understand the pathogenesis of asthma. Despite the breadth of T cells in asthma, approved therapeutics remain limited to TH2 networks.
Collapse
Affiliation(s)
- Naomi Bryant
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Lyndsey M Muehling
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia.
| |
Collapse
|
8
|
Liu X, Li X, Chen L, Hsu ACY, Asquith KL, Liu C, Laurie K, Barr I, Foster PS, Yang M. Proteomic Analysis Reveals a Novel Therapeutic Strategy Using Fludarabine for Steroid-Resistant Asthma Exacerbation. Front Immunol 2022; 13:805558. [PMID: 35280986 PMCID: PMC8913936 DOI: 10.3389/fimmu.2022.805558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/04/2022] [Indexed: 11/30/2022] Open
Abstract
Virus-induced asthma exacerbation is a health burden worldwide and lacks effective treatment. To better understand the disease pathogenesis and find novel therapeutic targets, we established a mouse model of steroid (dexamethasone (DEX)) resistant asthma exacerbation using ovalbumin (OVA) and influenza virus (FLU) infection. Using liquid chromatography-tandem mass spectrometry (LC-MC/MS), we performed a shotgun proteomics assay coupled with label-free quantification to define all dysregulated proteins in the lung proteome of asthmatic mice. Compared to control, 71, 89, and 30 proteins were found significantly upregulated by at least two-fold (p-value ≤ 0.05) in OVA-, OVA/FLU-, and OVA/FLU/DEX-treated mice, respectively. We then applied a Z-score transformed hierarchical clustering analysis and Ingenuity Pathway Analysis (IPA) to highlight the key inflammation pathways underlying the disease. Within all these upregulated proteins, 64 proteins were uniquely highly expressed in OVA/FLU mice compared to OVA mice; and 11 proteins were DEX-refractory. IPA assay revealed two of the most enriched pathways associated with these over-expressed protein clusters were those associated with MHC class I (MHC-I) antigen-presentation and interferon (IFN) signaling. Within these pathways, signal-transducer-and-activator-of-transcription-1 (STAT1) protein was identified as the most significantly changed protein contributing to the pathogenesis of exacerbation and the underlying steroid resistance based on the label-free quantification; and this was further confirmed by both Parallel Reaction Monitoring (PRM) proteomics assay and western blots. Further, the pharmacological drug Fludarabine decreased STAT1 expression, restored the responsiveness of OVA/FLU mice to DEX and markedly suppressed disease severity. Taken together, this study describes the proteomic profile underpinning molecular mechanisms of FLU-induced asthma exacerbation and identifies STAT1 as a potential therapeutic target, more importantly, we provided a novel therapeutic strategy that may be clinically translated into practice.
Collapse
Affiliation(s)
- Xiaoming Liu
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
| | - Xiang Li
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
| | - Ling Chen
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
| | - Alan Chen-Yu Hsu
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Kelly L. Asquith
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Karen Laurie
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ian Barr
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Paul S. Foster
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
- *Correspondence: Ming Yang, ; Paul S. Foster,
| | - Ming Yang
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Health Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
- *Correspondence: Ming Yang, ; Paul S. Foster,
| |
Collapse
|
9
|
Wu D, Jiang W, Liu C, Liu L, Li F, Ma X, Pan L, Liu C, Qu X, Liu H, Qin X, Xiang Y. CTNNAL1 participates in the regulation of mucus overproduction in HDM‐induced asthma mouse model through the YAP‐ROCK2 pathway. J Cell Mol Med 2022; 26:1656-1671. [PMID: 35092120 PMCID: PMC8899158 DOI: 10.1111/jcmm.17206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/24/2021] [Accepted: 01/12/2022] [Indexed: 01/26/2023] Open
Abstract
Our previous study indicated that adhesion molecule catenin alpha‐like 1(CTNNAL1) is downregulated in airway epithelial cells of asthma patients and asthma animal model but little is known about how the CTNNAL1 affects asthma pathogenesis. To reveal the direct relationship between asthma and CTNNAL1, CTNNAL1‐deficient mouse model in bronchopulmonary tissue was constructed by introducing CTNNAL1‐siRNA sequence using adeno‐associated virus (AAV) as vector. The mouse model of asthma was established by stimulation of house dust mite (HDM). After HDM‐challenged, there was marked airway inflammation, especially mucus hypersecretion in the CTNNAL1‐deficient mice. In addition, the CTNNAL1‐deficient mice exhibited an increase of lung IL‐4 and IL‐13 levels, as well as a significant increase of goblet cell hyperplasia and MUC5AC after HDM exposure. The expression of Yes‐associated protein (YAP), protein that interacted with α‐catenin, was downregulated after CTNNAL1 silencing and was upregulated due to its overexpression. In addition, the interaction between CTNNAL1 and YAP was confirmed by CO‐IP. Besides, inhibition of YAP could decrease the secretion of MUC5AC, IL‐4 and IL‐13 in CTNNAL1‐deficient 16HBE14o‐cells. Above results indicated us that CTNNAL1 regulated mucus hypersecretion through YAP pathway. In addition, the expression of ROCK2 increased when CTNNAL1 was silenced and decreased after YAP silencing, and inhibition of YAP decreased the expression of ROCK2 in CTNNAL1‐deficient HBE cells. Inhibition of ROCK2 decreased MUC5AC expression and IL‐13 secretion. In all, our study demonstrates that CTNNAL1 plays an important role in HDM‐induced asthma, mediating mucus secretion through the YAP‐ROCK2 pathway.
Collapse
Affiliation(s)
- Di Wu
- Department of Physiology School of Basic Medical Science Central South University Changsha China
| | - Wang Jiang
- Department of Medical Microbiology and Parasitology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Caixia Liu
- School of Integrated Chinese and Western Medicine Hunan University of Chinese Medicine Changsha China
| | - Lexin Liu
- Department of Physiology School of Basic Medical Science Central South University Changsha China
| | - Furong Li
- Department of Physiology School of Basic Medical Science Central South University Changsha China
| | - Xiaodi Ma
- Department of Physiology School of Basic Medical Science Central South University Changsha China
| | - Lang Pan
- Department of Physiology School of Basic Medical Science Central South University Changsha China
| | - Chi Liu
- Department of Physiology School of Basic Medical Science Central South University Changsha China
| | - Xiangping Qu
- Department of Physiology School of Basic Medical Science Central South University Changsha China
| | - Huijun Liu
- Department of Physiology School of Basic Medical Science Central South University Changsha China
| | - Xiaoqun Qin
- Department of Physiology School of Basic Medical Science Central South University Changsha China
| | - Yang Xiang
- Department of Physiology School of Basic Medical Science Central South University Changsha China
| |
Collapse
|
10
|
Bergeron HC, Tripp RA. Immunopathology of RSV: An Updated Review. Viruses 2021; 13:2478. [PMID: 34960746 PMCID: PMC8703574 DOI: 10.3390/v13122478] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
RSV is a leading cause of respiratory tract disease in infants and the elderly. RSV has limited therapeutic interventions and no FDA-approved vaccine. Gaps in our understanding of virus-host interactions and immunity contribute to the lack of biological countermeasures. This review updates the current understanding of RSV immunity and immunopathology with a focus on interferon responses, animal modeling, and correlates of protection.
Collapse
Affiliation(s)
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
11
|
Shimba A, Ejima A, Ikuta K. Pleiotropic Effects of Glucocorticoids on the Immune System in Circadian Rhythm and Stress. Front Immunol 2021; 12:706951. [PMID: 34691020 PMCID: PMC8531522 DOI: 10.3389/fimmu.2021.706951] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 09/22/2021] [Indexed: 12/16/2022] Open
Abstract
Glucocorticoids (GCs) are a class of steroid hormones secreted from the adrenal cortex. Their production is controlled by circadian rhythm and stress, the latter of which includes physical restraint, hunger, and inflammation. Importantly, GCs have various effects on immunity, metabolism, and cognition, including pleiotropic effects on the immune system. In general, GCs have strong anti-inflammatory and immunosuppressive effects. Indeed, they suppress inflammatory cytokine expression and cell-mediated immunity, leading to increased risks of some infections. However, recent studies have shown that endogenous GCs induced by the diurnal cycle and dietary restriction enhance immune responses against some infections by promoting the survival, redistribution, and response of T and B cells via cytokine and chemokine receptors. Furthermore, although GCs are reported to reduce expression of Th2 cytokines, GCs enhance type 2 immunity and IL-17-associated immunity in some stress conditions. Taken together, GCs have both immunoenhancing and immunosuppressive effects on the immune system.
Collapse
Affiliation(s)
- Akihiro Shimba
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aki Ejima
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Koichi Ikuta
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
12
|
Kwon Y, Kim M, Kim Y, Jeong MS, Jung HS, Jeoung D. EGR3-HDAC6-IL-27 Axis Mediates Allergic Inflammation and Is Necessary for Tumorigenic Potential of Cancer Cells Enhanced by Allergic Inflammation-Promoted Cellular Interactions. Front Immunol 2021; 12:680441. [PMID: 34234781 PMCID: PMC8257050 DOI: 10.3389/fimmu.2021.680441] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/07/2021] [Indexed: 11/18/2022] Open
Abstract
The objective of this study was to investigate mechanisms of allergic inflammation both in vitro and in vivo in details. For this, RNA sequencing was performed. Early growth response 3 gene (Egr3) was one of the most highly upregulated genes in rat basophilic leukemia (RBL2H3) cells stimulated by antigen. The role of Egr3 in allergic inflammation has not been studied extensively. Egr3 was necessary for passive cutaneous anaphylaxis (PCA) and passive systemic anaphylaxis (PSA). Egr3 promoter sequences contained potential binding site for NF-κB p65. NF-κB p65 directly regulated Egr3 expression and mediated allergic inflammation in vitro. Histone deacetylases (HDACs) is known to be involved in allergic airway inflammation. HDAC6 promoter sequences contained potential binding site for EGR3. EGR3 showed binding to promoter sequences of HDAC6. EGR3 was necessary for increased expression of histone deacetylase 6 (HDAC6) in antigen-stimulated RBL2H3 cells. HDAC6 mediated allergic inflammation in vitro and PSA. TargetScan analysis predicted that miR-182-5p was a negative regulator of EGR3. Luciferase activity assay confirmed that miR-182-5p was a direct regulator of EGR3. MiR-182-5p mimic inhibited allergic inflammation both in vitro and in vivo. Cytokine array showed that HDAC6 was necessary for increased interleukin-27 (IL-27) expression in BALB/C mouse model of PSA. Antigen stimulation did not affect expression of EBI3, another subunit of IL-27 in RBL2H3 cells or BALB/C mouse model of PCA or PSA. IL-27 receptor alpha was shown to be able to bind to HDAC6. IL-27 p28 mediated allergic inflammation in vitro, PCA, and PSA. Mouse recombinant IL-27 protein promoted features of allergic inflammation in an antigen-independent manner. HDAC6 was necessary for tumorigenic and metastatic potential enhanced by PSA. PSA enhanced the metastatic potential of mouse melanoma B16F1 cells in an IL-27-dependent manner. Experiments employing culture medium and mouse recombinant IL-27 protein showed that IL-27 mediated and promoted cellular interactions involving B16F1 cells, lung macrophages, and mast cells during allergic inflammation. IL-27 was present in exosomes of antigen-stimulated RBL2H3 cells. Exosomes from antigen-stimulated RBL2H3 cells enhanced invasion of B16F1 melanoma cells in an IL-27-dependemt manner. These results present evidence that EGR3-HDAC6-IL-27 axis can regulate allergic inflammation by mediating cellular interactions.
Collapse
Affiliation(s)
- Yoojung Kwon
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Misun Kim
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Youngmi Kim
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Myeong Seon Jeong
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea.,Chuncheon Center, Korea Basic Science Institute, Chuncheon, South Korea
| | - Hyun Suk Jung
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Dooil Jeoung
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
13
|
Li H, Wang H, Sokulsky L, Liu S, Yang R, Liu X, Zhou L, Li J, Huang C, Li F, Lei X, Jia H, Cheng J, Li F, Yang M, Zhang G. Single-cell transcriptomic analysis reveals key immune cell phenotypes in the lungs of patients with asthma exacerbation. J Allergy Clin Immunol 2021; 147:941-954. [PMID: 33039479 DOI: 10.1016/j.jaci.2020.09.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Asthma exacerbations are associated with heightened asthma symptoms, which can result in hospitalization in severe cases. However, the molecular immunologic processes that determine the course of an exacerbation remain poorly understood, impeding the progression of development of effective therapies. OBJECTIVE Our aim was to identify candidate genes that are strongly associated with asthma exacerbation at a cellular level. METHODS Subjects with asthma exacerbation and healthy control subjects were recruited, and bronchoalveolar lavage fluid was isolated from these subjects via bronchoscopy. Cells were isolated through fluorescence-activated cell sorting, and single-cell RNA sequencing was performed on enriched cell populations. RESULTS We showed that the levels of monocytes, CD8+ T cells, and macrophages are significantly elevated in the bronchoalveolar lavage fluid of patients. A set of cytokines and intracellular transduction regulators are associated with asthma exacerbations and are shared across multiple cell clusters, forming a complicated molecular framework. An additional group of core exacerbation-associated modules is activated, including eukaryotic initiation factor 2 signaling, ephrin receptor signaling, and C-X-C chemokine receptor type 4 signaling in the subpopulations of CD8+ T cells (C1-a) and monocyte clusters (C7 clusters), which are associated with infection. CONCLUSION Our study identified a significant number of severe asthma-associated genes that are differentially expressed by multiple cell clusters.
Collapse
Affiliation(s)
- Hui Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huaqi Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Leon Sokulsky
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| | - Shaoxia Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Yang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojie Liu
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lujia Zhou
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Juan Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chun Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangfang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu Lei
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongxia Jia
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiuling Cheng
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fuguang Li
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia; Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| | - Guojun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
14
|
Tu X, Donovan C, Kim RY, Wark PAB, Horvat JC, Hansbro PM. Asthma-COPD overlap: current understanding and the utility of experimental models. Eur Respir Rev 2021; 30:30/159/190185. [PMID: 33597123 PMCID: PMC9488725 DOI: 10.1183/16000617.0185-2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 11/03/2020] [Indexed: 12/21/2022] Open
Abstract
Pathological features of both asthma and COPD coexist in some patients and this is termed asthma-COPD overlap (ACO). ACO is heterogeneous and patients exhibit various combinations of asthma and COPD features, making it difficult to characterise the underlying pathogenic mechanisms. There are no controlled studies that define effective therapies for ACO, which arises from the lack of international consensus on the definition and diagnostic criteria for ACO, as well as scant in vitro and in vivo data. There remain unmet needs for experimental models of ACO that accurately recapitulate the hallmark features of ACO in patients. The development and interrogation of such models will identify underlying disease-causing mechanisms, as well as enabling the identification of novel therapeutic targets and providing a platform for assessing new ACO therapies. Here, we review the current understanding of the clinical features of ACO and highlight the approaches that are best suited for developing representative experimental models of ACO. Understanding the pathogenesis of asthma-COPD overlap is critical for improving therapeutic approaches. We present current knowledge on asthma-COPD overlap and the requirements for developing an optimal animal model of disease.https://bit.ly/3lsjyvm
Collapse
Affiliation(s)
- Xiaofan Tu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Both authors contributed equally
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, Australia.,University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia.,Both authors contributed equally
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, Australia.,University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia .,Centre for Inflammation, Centenary Institute, Camperdown, Australia.,University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| |
Collapse
|
15
|
Yuan F, Jiang L, Li Q, Sokulsky L, Wanyan Y, Wang L, Liu X, Zhou L, Tay HL, Zhang G, Yang M, Li F. A Selective α7 Nicotinic Acetylcholine Receptor Agonist, PNU-282987, Attenuates ILC2s Activation and Alternaria-Induced Airway Inflammation. Front Immunol 2021; 11:598165. [PMID: 33597946 PMCID: PMC7883686 DOI: 10.3389/fimmu.2020.598165] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 12/16/2020] [Indexed: 01/02/2023] Open
Abstract
Background The anti-inflammatory effect of an α7nAChR agonist, PNU-282987, has previously been explored in the context of inflammatory disease. However, the effects of PNU-282987 on type 2 innate lymphoid cells (ILC2s)-mediated allergic airway inflammation has not yet been established. Aims To determine the effects of PNU-282987 on the function of ILC2s in the context of IL-33– or Alternaria Alternata (AA)– induced airway inflammation. Methods PNU-282987 was administered to mice that received recombinant IL-33 or AA intranasal challenges. Lung histological analysis and flow cytometry were performed to determine airway inflammation and the infiltration and activation of ILC2s. The previously published α7nAChR agonist GTS-21 was employed as a comparable reagent. ILC2s were isolated from murine lung tissue and cultured in vitro in the presence of IL-33, IL-2, and IL-7 with/without either PNU-282987 or GTS-21. The expression of the transcription factors GATA3, IKK, and NF-κB were also determined. Results PNU-282987 and GTS-21 significantly reduced goblet cell hyperplasia in the airway, eosinophil infiltration, and ILC2s numbers in BALF, following IL-33 or AA challenge. In vitro IL-33 stimulation of isolated lung ILC2s showed a reduction of GATA3 and Ki67 in response to PNU-282987 or GTS-21 treatments. There was a significant reduction in IKK and NF-κB phosphorylation in the PNU-282987–treated group when compared to the GTS-21–treated ILC2s. Conclusion PNU-282987 inhibits ILC2-associated airway inflammation, where its effects were comparable to that of GTS-21.
Collapse
Affiliation(s)
- Fang Yuan
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.,Department of Medical Laboratory, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lili Jiang
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qianyang Li
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Leon Sokulsky
- Priority Research Centre for Healthy Lungs, Faculty of Health and Hunter Medical Research Institute, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Yuanyuan Wanyan
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lingli Wang
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaojie Liu
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lujia Zhou
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hock L Tay
- Priority Research Centre for Healthy Lungs, Faculty of Health and Hunter Medical Research Institute, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Guojun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ming Yang
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.,Priority Research Centre for Healthy Lungs, Faculty of Health and Hunter Medical Research Institute, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Fuguang Li
- Academy of Medical Sciences and Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Airway epithelial integrin β4 suppresses allergic inflammation by decreasing CCL17 production. Clin Sci (Lond) 2021; 134:1735-1749. [PMID: 32608482 DOI: 10.1042/cs20191188] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022]
Abstract
Airway epithelial cells (AECs) play a key role in asthma susceptibility and severity. Integrin β4 (ITGB4) is a structural adhesion molecule that is down-regulated in the airway epithelium of asthma patients. Although a few studies hint toward the role of ITGB4 in asthmatic inflammation pathogenesis, their specific resultant effects remain unexplored. In the present study, we determined the role of ITGB4 of AECs in the regulation of Th2 response and identified the underpinning molecular mechanisms. We found that ITGB4 deficiency led to exaggerated lung inflammation and AHR with higher production of CCL17 in house dust mite (HDM)-treated mice. ITGB4 regulated CCL17 production in AECs through EGFR, ERK and NF-κB pathways. EFGR-antagonist treatment or the neutralization of CCL17 both inhibited exaggerated pathological marks in HDM-challenged ITGB4-deficient mice. Together, these results demonstrated the involvement of ITGB4 deficiency in the development of Th2 responses of allergic asthma by down-regulation of EGFR and CCL17 pathway in AECs.
Collapse
|
17
|
Sokulsky LA, Garcia-Netto K, Nguyen TH, Girkin JLN, Collison A, Mattes J, Kaiko G, Liu C, Bartlett NW, Yang M, Foster PS. A Critical Role for the CXCL3/CXCL5/CXCR2 Neutrophilic Chemotactic Axis in the Regulation of Type 2 Responses in a Model of Rhinoviral-Induced Asthma Exacerbation. THE JOURNAL OF IMMUNOLOGY 2020; 205:2468-2478. [PMID: 32948685 DOI: 10.4049/jimmunol.1901350] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 08/22/2020] [Indexed: 11/19/2022]
Abstract
Rhinovirus (RV) infections in asthmatic patients are often associated with asthma exacerbation, characterized by worsened airways hyperreactivity and increased immune cell infiltration to the airways. The C-X-C chemokines, CXCL3 and CXCL5, regulate neutrophil trafficking to the lung via CXCR2, and their expression in the asthmatic lung is associated with steroid-insensitive type 2 inflammatory signatures. Currently, the role of CXCL3 and CXCL5 in regulating neutrophilic and type 2 responses in viral-induced asthma exacerbation is unknown. Inhibition of CXCL3 or CXCL5 with silencing RNAs in a mouse model of RV-induced exacerbation of asthma attenuated the accumulation of CXCR2+ neutrophils, eosinophils, and innate lymphoid cells in the lung and decreased production of type 2 regulatory factors IL-25, IL-33, IL-5, IL-13, CCL11, and CCL24. Suppression of inflammation was associated with decreased airways hyperreactivity, mucus hypersecretion, and collagen deposition. Similar results were obtained by employing RC-3095, which has been shown to bind to CXCR2, or by depletion of neutrophils. Our data demonstrate that CXCL3 and CXCL5 may be critical in the perpetuation of RV-induced exacerbation of asthma through the recruitment of CXCR2-positive neutrophils and by promoting type 2 inflammation. Targeting the CXCL3/CXCL5/CXCR2 axis may provide a new therapeutic approach to attenuating RV-induced exacerbations of asthma.
Collapse
Affiliation(s)
- Leon A Sokulsky
- Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan 2308, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton Heights 2305, New South Wales, Australia
| | - Keilah Garcia-Netto
- Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan 2308, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton Heights 2305, New South Wales, Australia
| | - Thi Hiep Nguyen
- Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan 2308, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton Heights 2305, New South Wales, Australia
| | - Jason L N Girkin
- Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan 2308, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton Heights 2305, New South Wales, Australia
| | - Adam Collison
- Hunter Medical Research Institute, New Lambton Heights 2305, New South Wales, Australia.,Priority Research Centre GrowUpWell, Experimental and Translational Respiratory Medicine Group, The University of Newcastle, Callaghan 2308, Australia
| | - Joerg Mattes
- Hunter Medical Research Institute, New Lambton Heights 2305, New South Wales, Australia.,Priority Research Centre GrowUpWell, Experimental and Translational Respiratory Medicine Group, The University of Newcastle, Callaghan 2308, Australia.,Paediatric Respiratory and Sleep Medicine Department, Newcastle Children's Hospital, Kaleidoscope, New Lambton Heights 2305, Australia; and
| | - Gerard Kaiko
- Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan 2308, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton Heights 2305, New South Wales, Australia
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha 410083, Hunan, China
| | - Nathan W Bartlett
- Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan 2308, New South Wales, Australia.,Hunter Medical Research Institute, New Lambton Heights 2305, New South Wales, Australia
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan 2308, New South Wales, Australia; .,Hunter Medical Research Institute, New Lambton Heights 2305, New South Wales, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, University of Newcastle, Callaghan 2308, New South Wales, Australia; .,Hunter Medical Research Institute, New Lambton Heights 2305, New South Wales, Australia
| |
Collapse
|
18
|
LeMessurier KS, Tiwary M, Morin NP, Samarasinghe AE. Respiratory Barrier as a Safeguard and Regulator of Defense Against Influenza A Virus and Streptococcus pneumoniae. Front Immunol 2020; 11:3. [PMID: 32117216 PMCID: PMC7011736 DOI: 10.3389/fimmu.2020.00003] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/03/2020] [Indexed: 12/27/2022] Open
Abstract
The primary function of the respiratory system of gas exchange renders it vulnerable to environmental pathogens that circulate in the air. Physical and cellular barriers of the respiratory tract mucosal surface utilize a variety of strategies to obstruct microbe entry. Physical barrier defenses including the surface fluid replete with antimicrobials, neutralizing immunoglobulins, mucus, and the epithelial cell layer with rapidly beating cilia form a near impenetrable wall that separates the external environment from the internal soft tissue of the host. Resident leukocytes, primarily of the innate immune branch, also maintain airway integrity by constant surveillance and the maintenance of homeostasis through the release of cytokines and growth factors. Unfortunately, pathogens such as influenza virus and Streptococcus pneumoniae require hosts for their replication and dissemination, and prey on the respiratory tract as an ideal environment causing severe damage to the host during their invasion. In this review, we outline the host-pathogen interactions during influenza and post-influenza bacterial pneumonia with a focus on inter- and intra-cellular crosstalk important in pulmonary immune responses.
Collapse
Affiliation(s)
- Kim S LeMessurier
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| | - Meenakshi Tiwary
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| | - Nicholas P Morin
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Critical Care Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Amali E Samarasinghe
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Division of Pulmonology, Allergy-Immunology, and Sleep, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, United States
| |
Collapse
|
19
|
Tang S, Du X, Yuan L, Xiao G, Wu M, Wang L, Wu S, Duan Z, Xiang Y, Qu X, Liu H, Zou Y, Qin X, Qin L, Liu C. Airway epithelial ITGB4 deficiency in early life mediates pulmonary spontaneous inflammation and enhanced allergic immune response. J Cell Mol Med 2020; 24:2761-2771. [PMID: 31970850 PMCID: PMC7077534 DOI: 10.1111/jcmm.15000] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 12/13/2019] [Accepted: 12/27/2019] [Indexed: 12/22/2022] Open
Abstract
Lung immune responses to respiratory pathogens and allergens are initiated in early life which will further influence the later onset of asthma. The airway epithelia form the first mechanical physical barrier to allergic stimuli and environmental pollutants, which is also the key regulator in the initiation and development of lung immune response. However, the epithelial regulation mechanisms of early-life lung immune responses are far from clear. Our previous study found that integrin β4 (ITGB4) is decreased in the airway epithelium of asthma patients with specific variant site. ITGB4 deficiency in adult mice aggravated the lung Th2 immune responses and enhanced airway hyper-responsiveness (AHR) with a house dust mite (HDM)-induced asthma model. However, the contribution of ITGB4 to the postnatal lung immune response is still obscure. Here, we further demonstrated that ITGB4 deficiency following birth mediates spontaneous lung inflammation with ILC2 activation and increased infiltration of eosinophils and lymphocytes. Moreover, ITGB4 deficiency regulated thymic stromal lymphopoietin (TSLP) production in airway epithelial cells through EGFR pathways. Neutralization of TSLP inhibited the spontaneous inflammation significantly in ITGB4-deficient mice. Furthermore, we also found that ITGB4 deficiency led to exaggerated lung allergic inflammation response to HDM stress. In all, these findings indicate that ITGB4 deficiency in early life causes spontaneous lung inflammation and induces exaggerated lung inflammation response to HDM aeroallergen.
Collapse
Affiliation(s)
- Sha Tang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,Reproductive and Genetic Hospital of Citic-Xiangya, Changsha, China
| | - Xizi Du
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Lin Yuan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Mengping Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Leyuan Wang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - ShuangYan Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Zhen Duan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yizhou Zou
- Department of Immunology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,Research Center of China-Africa Infectious Diseases, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
20
|
van Erp EA, Feyaerts D, Duijst M, Mulder HL, Wicht O, Luytjes W, Ferwerda G, van Kasteren PB. Respiratory Syncytial Virus Infects Primary Neonatal and Adult Natural Killer Cells and Affects Their Antiviral Effector Function. J Infect Dis 2019; 219:723-733. [PMID: 30252097 PMCID: PMC6376914 DOI: 10.1093/infdis/jiy566] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/18/2018] [Indexed: 01/01/2023] Open
Abstract
Background Respiratory syncytial virus (RSV) is a major cause of severe acute lower respiratory tract infections in infants. Natural killer (NK) cells are important antiviral effector cells that likely encounter RSV in the presence of virus-specific (maternal) antibodies. As NK cells potentially contribute to immunopathology, we investigated whether RSV affects their antiviral effector functions. Methods We assessed the phenotype and functionality of primary neonatal and adult NK cells by flow cytometry after stimulation with RSV or RSV-antibody complexes. Results We demonstrate for the first time that RSV infects neonatal and adult NK cells in vitro. Preincubation of virus with subneutralizing concentrations of RSV-specific antibodies significantly increased the percentage of infected NK cells. Upon infection, NK cells were significantly more prone to produce interferon-γ, while secretion of the cytotoxicity molecule perforin was not enhanced. Conclusions Our findings suggest that (antibody-enhanced) RSV infection of NK cells induces a proinflammatory rather than a cytotoxic response, which may contribute to immunopathology. Considering that most RSV vaccines currently being developed aim at inducing (maternal) antibodies, these results highlight the importance of understanding the interactions between innate effector cells and virus-specific antibodies.
Collapse
Affiliation(s)
- Elisabeth A van Erp
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven
- Section of Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Dorien Feyaerts
- Section of Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Maxime Duijst
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven
| | - H Lie Mulder
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven
| | - Oliver Wicht
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven
| | - Willem Luytjes
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven
| | - Gerben Ferwerda
- Section of Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Puck B van Kasteren
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven
- Correspondence: P. B. van Kasteren, PhD, National Institute for Public Health and the Environment, Center for Infectious Diseases Control, Postbus 1, 3720 BA Bilthoven, The Netherlands ()
| |
Collapse
|
21
|
Hadjigol S, Netto KG, Maltby S, Tay HL, Nguyen TH, Hansbro NG, Eyers F, Hansbro PM, Yang M, Foster PS. Lipopolysaccharide induces steroid-resistant exacerbations in a mouse model of allergic airway disease collectively through IL-13 and pulmonary macrophage activation. Clin Exp Allergy 2019; 50:82-94. [PMID: 31579973 DOI: 10.1111/cea.13505] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/26/2019] [Accepted: 09/15/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Acute exacerbations of asthma represent a major burden of disease and are often caused by respiratory infections. Viral infections are recognized as significant triggers of exacerbations; however, less is understood about the how microbial bioproducts such as the endotoxin (lipopolysaccharide (LPS)) trigger episodes. Indeed, increased levels of LPS have been linked to asthma onset, severity and steroid resistance. OBJECTIVE The goal of this study was to identify mechanisms underlying bacterial-induced exacerbations by employing LPS as a surrogate for infection. METHODS We developed a mouse model of LPS-induced exacerbation on the background of pre-existing type-2 allergic airway disease (AAD). RESULTS LPS-induced exacerbation was characterized by steroid-resistant airway hyperresponsiveness (AHR) and an exaggerated inflammatory response distinguished by increased numbers of infiltrating neutrophils/macrophages and elevated production of lung inflammatory cytokines, including TNFα, IFNγ, IL-27 and MCP-1. Expression of the type-2 associated inflammatory factors such as IL-5 and IL-13 were elevated in AAD but not altered by LPS exposure. Furthermore, AHR and airway inflammation were no longer suppressed by corticosteroid (dexamethasone) treatment after LPS exposure. Depletion of pulmonary macrophages by administration of 2-chloroadenosine into the lungs suppressed AHR and reduced IL-13, TNFα and IFNγ expression. Blocking IL-13 function, through either IL-13-deficiency or administration of specific blocking antibodies, also suppressed AHR and airway inflammation. CONCLUSIONS & CLINICAL RELEVANCE We present evidence that IL-13 and innate immune pathways (in particular pulmonary macrophages) contribute to LPS-induced exacerbation of pre-existing AAD and provide insight into the complex molecular processes potentially underlying microbial-induced exacerbations.
Collapse
Affiliation(s)
- Sara Hadjigol
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Keilah G Netto
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Steven Maltby
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Hock L Tay
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Thi H Nguyen
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Fiona Eyers
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Department of Microbiology and Immunology, School of Biomedical Sciences & Pharmacy, Faculty of Health and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
22
|
Tait Wojno ED, Hunter CA, Stumhofer JS. The Immunobiology of the Interleukin-12 Family: Room for Discovery. Immunity 2019; 50:851-870. [PMID: 30995503 PMCID: PMC6472917 DOI: 10.1016/j.immuni.2019.03.011] [Citation(s) in RCA: 291] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 12/12/2022]
Abstract
The discovery of interleukin (IL)-6 and its receptor subunits provided a foundation to understand the biology of a group of related cytokines: IL-12, IL-23, and IL-27. These family members utilize shared receptors and cytokine subunits and influence the outcome of cancer, infection, and inflammatory diseases. Consequently, many facets of their biology are being therapeutically targeted. Here, we review the landmark discoveries in this field, the combinatorial biology inherent to this family, and how patient datasets have underscored the critical role of these pathways in human disease. We present significant knowledge gaps, including how similar signals from these cytokines can mediate distinct outcomes, and discuss how a better understanding of the biology of the IL-12 family provides new therapeutic opportunities.
Collapse
Affiliation(s)
- Elia D Tait Wojno
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, 235 Hungerford Hill Rd., Ithaca, NY 14853, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Ave., Philadelphia, PA 19104-4539, USA.
| | - Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, 4301 West Markham St., Little Rock, AR 72205, USA.
| |
Collapse
|
23
|
Regulatory cytokine function in the respiratory tract. Mucosal Immunol 2019; 12:589-600. [PMID: 30874596 PMCID: PMC7051906 DOI: 10.1038/s41385-019-0158-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/22/2019] [Accepted: 02/27/2019] [Indexed: 02/04/2023]
Abstract
The respiratory tract is an important site of immune regulation; required to allow protective immunity against pathogens, while minimizing tissue damage and avoiding aberrant inflammatory responses to inhaled allergens. Several cell types work in concert to control pulmonary immune responses and maintain tolerance in the respiratory tract, including regulatory and effector T cells, airway and interstitial macrophages, dendritic cells and the airway epithelium. The cytokines transforming growth factor β, interleukin (IL-) 10, IL-27, and IL-35 are key coordinators of immune regulation in tissues such as the lung. Here, we discuss the role of these cytokines during respiratory infection and allergic airway disease, highlighting the critical importance of cellular source and immunological context for the effects of these cytokines in vivo.
Collapse
|