1
|
Gao X, Jiang J. Exploring the regulatory mechanism of intestinal flora based on PD-1 receptor/ligand targeted cancer immunotherapy. Front Immunol 2024; 15:1359029. [PMID: 38617841 PMCID: PMC11010636 DOI: 10.3389/fimmu.2024.1359029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/18/2024] [Indexed: 04/16/2024] Open
Abstract
Serving as a pivotal immunotherapeutic approach against tumors, anti-PD-1/PD-L1 therapy amplifies the immune cells' capability to eliminate tumors by obstructing the interaction between PD-1 and PD-L1. Research indicates that immune checkpoint inhibitors are effective when a patient's gut harbors unique beneficial bacteria. As such, it has further been revealed that the gut microbiome influences tumor development and the efficacy of cancer treatments, with metabolites produced by the microbiome playing a regulatory role in the antitumor efficacy of Immune checkpoint inhibitors(ICBs). This article discusses the mechanism of anti-PD-1 immunotherapy and the role of intestinal flora in immune regulation. This review focuses on the modulation of intestinal flora in the context of PD-1 immunotherapy, which may offer a new avenue for combination therapy in tumor immunotherapy.
Collapse
Affiliation(s)
- Xinran Gao
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor lmmunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor lmmunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
2
|
Inan S, Meissler JJ, Bessho S, Wiah S, Tukel C, Eisenstein TK, Rawls SM. Blocking IL-17A prevents oxycodone-induced depression-like effects and elevation of IL-6 levels in the ventral tegmental area and reduces oxycodone-derived physical dependence in rats. Brain Behav Immun 2024; 117:100-111. [PMID: 38199516 PMCID: PMC10932873 DOI: 10.1016/j.bbi.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/01/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Oxycodone is the most prescribed opioid for pain management and has been available in clinics for almost a century, but effects of chronic oxycodone have been studied less than morphine in preclinical and clinical studies. Newly developed depression has been coupled with chronic oxycodone use in a few clinical studies, but no preclinical studies have investigated the pathogenesis of oxycodone-induced depression. Gut microbiome changes following oxycodone use is an understudied area, and interleukin-17A (IL-17A) is linked to both the development of mood disorders and regulation of gut microbiome. The present study investigated effects of chronic oxycodone exposure on mood-related behaviors (depression and anxiety), pain hypersensitivity, physical dependence, immune markers, and the gut microbiome and tested the hypothesis that blocking IL-17A with a systemically administered monoclonal antibody reduces oxycodone-derived effects. Oxycodone (using an incremental dosing regimen) or saline was injected twice a day for 12 days. IL-17A Ab (200 µg/100 µl) or saline was administered every 3rd day during the 12-day interval. Chronic oxycodone induced a depression-like effect, but not anxiogenic- or anxiolytic-like effects; promoted hyperalgesia; increased IL-17A and IL-6 levels in the ventral tegmental area (VTA); and induced physical dependence. IL-17A Ab co-administration with oxycodone prevented the depression-like effect and hyperalgesia, reduced naloxone-precipitated withdrawal signs, and normalized the increase in cytokine levels. Chronic oxycodone exposure did not affect gut microbiome and integrity. Our results identify a role for IL-17A in oxycodone-related behavioral and neuroimmune effects and show that IL-17A Ab has potential therapeutic value in blocking these effects. Given that humanized IL-17A Ab is approved for treatment of psoriasis and psoriatic arthritis, our findings point toward studying it for use in the treatment of oxycodone use disorder.
Collapse
Affiliation(s)
- Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| | - Joseph J Meissler
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Shingo Bessho
- Center for Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Sonita Wiah
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Cagla Tukel
- Center for Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA; Department of Microbiology, Immunology, and Inflammation, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Toby K Eisenstein
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA; Department of Microbiology, Immunology, and Inflammation, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Scott M Rawls
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
3
|
Xiao H, Peng L, Jiang D, Liu Y, Zhu L, Li Z, Geng J, Xie B, Huang X, Wang J, Dai H, Wang C. IL‐17A
promotes lung fibrosis through impairing mitochondrial homeostasis in type
II
alveolar epithelial cells. J Cell Mol Med 2022; 26:5728-5741. [DOI: 10.1111/jcmm.17600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/19/2022] [Accepted: 10/04/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Huijuan Xiao
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital, School of Clinical Medicine Peking University Beijing China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical College Beijing China
| | - Liang Peng
- Beijing Key Laboratory for Immune‐Mediated Inflammatory Diseases, Institute of Medical Science China‐Japan Friendship Hospital Beijing China
| | - Dingyuan Jiang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical College Beijing China
| | - Yuan Liu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical College Beijing China
- Department of Respiratory and Critical Care Medicine Zhongnan Hospital of Wuhan University Wuhan China
| | - Lili Zhu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical College Beijing China
| | - Zhen Li
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical College Beijing China
| | - Jing Geng
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical College Beijing China
| | - Bingbing Xie
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical College Beijing China
| | - Xiaoxi Huang
- Medical Research Center Beijing Chaoyang Hospital Affiliated to Capital Medical University Beijing China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences School of Basic Medicine Peking Union Medical College Beijing China
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical College Beijing China
| | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital, School of Clinical Medicine Peking University Beijing China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China‐Japan Friendship Hospital; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical College Beijing China
| |
Collapse
|
4
|
Chen S, Zhang X, Yang C, Wang S, Shen H. Essential role of IL-17 in acute exacerbation of pulmonary fibrosis induced by non-typeable Haemophilus influenzae. Theranostics 2022; 12:5125-5137. [PMID: 35836804 PMCID: PMC9274745 DOI: 10.7150/thno.74809] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/14/2022] [Indexed: 01/12/2023] Open
Abstract
Background: Acute exacerbation (AE) of idiopathic pulmonary fibrosis (IPF) has a poor prognosis and lacks effective therapy. Animal models that mimic AE-IPF can greatly accelerate investigation of its pathogenesis and development of effective therapy. However, there are few reports of animal models of AE-IPF caused by bacteria. Thus, our study aimed to establish a mouse model of bacterium-induced AE-IPF and explore the potential pathogenic mechanism of AE-IPF. Methods: Mice were instilled intranasally with bleomycin (BLM) followed by non-typeable Haemophilus influenzae (NTHi) strain NT127. Murine survival, bacterial load, body weight and pulmonary histopathological changes were evaluated. We analyzed the T cell and inflammatory cell responses in the lungs. Results: Infection with 107 CFU NT127 triggered AE in mice with PF induced by 30 μg BLM. Compared with BLM-instilled mice, the BLM/NT127-treated mice showed more obvious airway inflammation, lower survival rate, higher inflammatory cell response, and increased proportions and numbers of IL-17+CD4+, IL-17+ γδ T, IL-22+CD4+ and regulatory T (Treg) cells in lungs. γδ T cells were the predominant source of IL-17. IL-17 gene knockout mice with AE-IPF had quicker body weight recovery, milder pulmonary inflammation and fibrosis, stronger IL-22+CD4+T, TGF-β+ γδ T and Treg cell responses, and weaker neutrophil and eosinophil responses than wild-type mice with AE-IPF. Conclusions: NTHi infection after BLM-induced IPF can cause AE-IPF in a murine model. This novel model can be used to investigate the pathogenesis of AE-IPF and develop new therapies for AE-IPF caused by bacteria. IL-17 is essential for the development of AE-IPF, and it may be a new therapeutic target for bacteria-induced AE-IPF.
Collapse
Affiliation(s)
- Shengsen Chen
- Department of Endoscopy (the bronchoscope group), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia 19104, USA
| | - Xinyun Zhang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia 19104, USA.,Department of Infectious Diseases, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Cheng Yang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia 19104, USA.,Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shi Wang
- Department of Endoscopy (the bronchoscope group), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China.,✉ Corresponding authors: Shi Wang, Department of Endoscopy (the bronchoscope group), Zhejiang Cancer Hospital, No. 1 Banshandong Road, Hangzhou 310022, China. E-mail: ; Hao Shen, Department of Microbiology, University of Pennsylvania Perelman School of Medicine, 3610 Hamilton Walk, Philadelphia 19104, USA. E-mail:
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia 19104, USA.,✉ Corresponding authors: Shi Wang, Department of Endoscopy (the bronchoscope group), Zhejiang Cancer Hospital, No. 1 Banshandong Road, Hangzhou 310022, China. E-mail: ; Hao Shen, Department of Microbiology, University of Pennsylvania Perelman School of Medicine, 3610 Hamilton Walk, Philadelphia 19104, USA. E-mail:
| |
Collapse
|
5
|
Albrecht M, Halle O, Gaedcke S, Pallenberg ST, Camargo Neumann J, Witt M, Roediger J, Schumacher M, Jirmo AC, Warnecke G, Jonigk D, Braubach P, DeLuca D, Hansen G, Dittrich AM. Interleukin-17A and interleukin-22 production by conventional and non-conventional lymphocytes in three different end-stage lung diseases. Clin Transl Immunology 2022; 11:e1398. [PMID: 35757569 PMCID: PMC9202301 DOI: 10.1002/cti2.1398] [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: 10/22/2021] [Revised: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 11/10/2022] Open
Abstract
Objectives The contribution of adaptive vs. innate lymphocytes to IL-17A and IL-22 secretion at the end stage of chronic lung diseases remains largely unexplored. In order to uncover tissue- and disease-specific secretion patterns, we compared production patterns of IL-17A and IL-22 in three different human end-stage lung disease entities. Methods Production of IL-17A, IL-22 and associated cytokines was assessed in supernatants of re-stimulated lymphocytes by multiplex assays and multicolour flow cytometry of conventional T cells, iNKT cells, γδ T cells and innate lymphoid cells in bronchial lymph node and lung tissue from patients with emphysema (n = 19), idiopathic pulmonary fibrosis (n = 14) and cystic fibrosis (n = 23), as well as lung donors (n = 17). Results We detected secretion of IL-17A and IL-22 by CD4+ T cells, CD8+ T cells, innate lymphoid cells, γδ T cells and iNKT cells in all end-stage lung disease entities. Our analyses revealed disease-specific contributions of individual lymphocyte subpopulations to cytokine secretion patterns. We furthermore found the high levels of microbial detection in CF samples to associate with a more pronounced IL-17A signature upon antigen-specific and unspecific re-stimulation compared to other disease entities and lung donors. Conclusion Our results show that both adaptive and innate lymphocyte populations contribute to IL-17A-dependent pathologies in different end-stage lung disease entities, where they establish an IL-17A-rich microenvironment. Microbial colonisation patterns and cytokine secretion upon microbial re-stimulation suggest that pathogens drive IL-17A secretion patterns in end-stage lung disease.
Collapse
Affiliation(s)
- Melanie Albrecht
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany.,Molecular Allergology Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines Langen Germany.,Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Olga Halle
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany.,Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Svenja Gaedcke
- Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Sophia T Pallenberg
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany
| | - Julia Camargo Neumann
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany
| | - Marius Witt
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany
| | - Johanna Roediger
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany
| | - Marina Schumacher
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany
| | - Adan Chari Jirmo
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany.,Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Gregor Warnecke
- Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany.,Department of Cardiac Surgery Heidelberg Medical School Heidelberg Germany
| | - Danny Jonigk
- Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany.,Institute of Pathology Hannover Medical School Hannover Germany
| | - Peter Braubach
- Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany.,Institute of Pathology Hannover Medical School Hannover Germany
| | - David DeLuca
- Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Gesine Hansen
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany.,Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| | - Anna-Maria Dittrich
- Pediatric Pneumology, Allergology and Neonatology Hannover Medical School Hannover Germany.,Biomedical Research in Endstage and Obstructive Lung Diseases (BREATH), German Center for Lung Research (DZL) Hannover Germany
| |
Collapse
|
6
|
Uzeloto JS, de Toledo-Arruda AC, Silva BSA, Braz AMM, de Lima FF, Grigoletto I, Ramos D, Golim MA, Ramos EMC. Effect of physical training on cytokine expression in CD4+ T lymphocytes in subjects with stable COPD. Ther Adv Respir Dis 2022; 16:17534666221091179. [PMID: 35695009 PMCID: PMC9189509 DOI: 10.1177/17534666221091179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Introduction: Although evidence suggests that physical exercise reduces systemic inflammation, at the plasma level, there are still contradictions in chronic obstructive pulmonary disease (COPD). In this sense, analysis of intracellular cytokines could clear off the effect of physical exercise on the inflammatory profile of these subjects. Aim: The aim was to evaluate the effect of physical training on cytokine expression in CD4+ T lymphocytes from subjects with COPD. Methods: This is a randomized controlled trial. Subjects with stable COPD were grouped into two groups, exercise and control. In total, 23 subjects with stable COPD were evaluated, of which 15 underwent aerobic strength training [physical exercise group (PEG)] and 8 underwent breathing exercises [respiratory physiotherapy group (RPG)]. Intracellular cytokines [interleukin (IL)-8, IL-13, IL-17, IL-6, IL-2, IL-10, and tumor necrosis factor alpha (TNF-α)] from CD4+ T lymphocytes were analyzed from peripheral blood through flow cytometry, before and after 8 weeks of intervention. Results: The PEG and RPG groups had a mean age of 68 ± 5.96 and 72.25 ± 6.86 years and predicted forced expiratory volume in the first second (FEV1) of 58.6 ± 15.99% and 39.75 ± 10.39%, respectively. It was possible to detect a significant reduction in IL-8 (p = 0.0125) and an increase in IL-13 (p = 0.0014) and an increase in TNF-α (p < 0.001) in both groups. Conclusion: Eight weeks of physical training, both peripheral and respiratory, were able to reduce concentrations of IL-8 and to increase IL-13, and TNF-α in CD4+ T lymphocytes in subjects with stable COPD. The findings reinforce the benefits of interventions in subjects with COPD, revealing data not previously investigated.
Collapse
Affiliation(s)
- Juliana S Uzeloto
- Department of Physiotherapy, Postgraduate Program in Physiotherapy, Faculty of Science and Technology, São Paulo State University (UNESP), Rua Roberto Simonsen, 305, Presidente Prudente, São Paulo 19060-900, Brazil
| | - Alessandra C de Toledo-Arruda
- Department of Physiology and Pharmacology, Laboratory of Exercise Sciences, Fluminense Federal University, Rio de Janeiro, Brazil
| | - Bruna S A Silva
- Physiotherapy, Universidade Estadual Paulista Julio de Mesquita Filho, Presidente Prudente
| | - Aline M M Braz
- Universidade Estadual Paulista Julio de Mesquita Filho.,Instituto de Biociencias Campus de Botucatu, Medical Biotechnology, Botucatu
| | - Fabiano F de Lima
- Physiotherapy, Universidade Estadual Paulista Julio de Mesquita Filho, Presidente Prudente
| | - Isis Grigoletto
- Physiotherapy, Universidade Estadual Paulista Julio de Mesquita Filho, Presidente Prudente
| | - Dionei Ramos
- Physiotherapy, Universidade Estadual Paulista Julio de Mesquita Filho, Presidente Prudente
| | - Marjorie A Golim
- Botucatu Medical School, Postgraduate Program in Research & Development: Medical Biotechnology, Blood Center, Flow Cytometry Laboratory, São Paulo State University (UNESP), São Paulo, Brazil
| | - Ercy M C Ramos
- Physiotherapy, Universidade Estadual Paulista Julio de Mesquita Filho, Presidente Prudente
| |
Collapse
|
7
|
Fu Z, Dean JW, Xiong L, Dougherty MW, Oliff KN, Chen ZME, Jobin C, Garrett TJ, Zhou L. Mitochondrial transcription factor A in RORγt + lymphocytes regulate small intestine homeostasis and metabolism. Nat Commun 2021; 12:4462. [PMID: 34294718 PMCID: PMC8298438 DOI: 10.1038/s41467-021-24755-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
RORγt+ lymphocytes, including interleukin 17 (IL-17)-producing gamma delta T (γδT17) cells, T helper 17 (Th17) cells, and group 3 innate lymphoid cells (ILC3s), are important immune regulators. Compared to Th17 cells and ILC3s, γδT17 cell metabolism and its role in tissue homeostasis remains poorly understood. Here, we report that the tissue milieu shapes splenic and intestinal γδT17 cell gene signatures. Conditional deletion of mitochondrial transcription factor A (Tfam) in RORγt+ lymphocytes significantly affects systemic γδT17 cell maintenance and reduces ILC3s without affecting Th17 cells in the gut. In vivo deletion of Tfam in RORγt+ lymphocytes, especially in γδT17 cells, results in small intestine tissue remodeling and increases small intestine length by enhancing the type 2 immune responses in mice. Moreover, these mice show dysregulation of the small intestine transcriptome and metabolism with less body weight but enhanced anti-helminth immunity. IL-22, a cytokine produced by RORγt+ lymphocytes inhibits IL-13-induced tuft cell differentiation in vitro, and suppresses the tuft cell-type 2 immune circuit and small intestine lengthening in vivo, highlighting its key role in gut tissue remodeling.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Joseph W Dean
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Lifeng Xiong
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | | | - Kristen N Oliff
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Zong-Ming E Chen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Christian Jobin
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32608, USA
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
8
|
Hagner M, Albrecht M, Guerra M, Braubach P, Halle O, Zhou-Suckow Z, Butz S, Jonigk D, Hansen G, Schultz C, Dittrich AM, Mall MA. IL-17A from innate and adaptive lymphocytes contributes to inflammation and damage in cystic fibrosis lung disease. Eur Respir J 2021; 57:13993003.00716-2019. [PMID: 33303549 DOI: 10.1183/13993003.00716-2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 11/15/2020] [Indexed: 11/05/2022]
Abstract
BACKGROUND Elevated levels of interleukin (IL)-17A were detected in the airways of patients with cystic fibrosis (CF), but its cellular sources and role in the pathogenesis of CF lung disease remain poorly understood. The aim of this study was to determine the sources of IL-17A and its role in airway inflammation and lung damage in CF. METHODS We performed flow cytometry to identify IL-17A-producing cells in lungs and peripheral blood from CF patients and β-epithelial Na+ channel transgenic (Scnn1b-Tg) mice with CF-like lung disease, and determined the effects of genetic deletion of Il17a and Rag1 on the pulmonary phenotype of Scnn1b-Tg mice. RESULTS T-helper 17 cells, CD3+CD8+ T-cells, γδ T-cells, invariant natural killer T-cells and innate lymphoid cells contribute to IL-17A secretion in lung tissue, lymph nodes and peripheral blood of patients with CF. Scnn1b-Tg mice displayed increased pulmonary expression of Il17a and elevated IL-17A-producing innate and adaptive lymphocytes with a major contribution by γδ T-cells. Lack of IL-17A, but not the recombination activating protein RAG1, reduced neutrophilic airway inflammation in Scnn1b-Tg mice. Genetic deletion of Il17a or Rag1 had no effect on mucus obstruction, but reduced structural lung damage and revealed an IL-17A-dependent macrophage activation in Scnn1b-Tg mice. CONCLUSIONS We identify innate and adaptive sources of IL-17A in CF lung disease. Our data demonstrate that IL-17A contributes to airway neutrophilia, macrophage activation and structural lung damage in CF-like lung disease in mice. These results suggest IL-17A as a novel target for anti-inflammatory therapy of CF lung disease.
Collapse
Affiliation(s)
- Matthias Hagner
- Dept of Translational Pulmonology, Translational Lung Research Center (TLRC), University of Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Germany.,These authors contributed equally to the study
| | - Melanie Albrecht
- German Center for Lung Research (DZL), Germany.,Clinic for Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,These authors contributed equally to the study
| | - Matteo Guerra
- Dept of Translational Pulmonology, Translational Lung Research Center (TLRC), University of Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Germany.,Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory, Heidelberg, Germany.,Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Peter Braubach
- German Center for Lung Research (DZL), Germany.,Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Olga Halle
- German Center for Lung Research (DZL), Germany.,Clinic for Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Zhe Zhou-Suckow
- Dept of Translational Pulmonology, Translational Lung Research Center (TLRC), University of Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Germany
| | - Simone Butz
- Dept of Translational Pulmonology, Translational Lung Research Center (TLRC), University of Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Germany
| | - Danny Jonigk
- German Center for Lung Research (DZL), Germany.,Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Gesine Hansen
- German Center for Lung Research (DZL), Germany.,Clinic for Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Carsten Schultz
- German Center for Lung Research (DZL), Germany.,Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory, Heidelberg, Germany.,Dept of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Anna-Maria Dittrich
- German Center for Lung Research (DZL), Germany.,Clinic for Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,These authors contributed equally as senior authors
| | - Marcus A Mall
- Dept of Translational Pulmonology, Translational Lung Research Center (TLRC), University of Heidelberg, Heidelberg, Germany .,German Center for Lung Research (DZL), Germany.,Dept of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,These authors contributed equally as senior authors
| |
Collapse
|
9
|
Yadav M, Sardana I, Sharma A, Sharma N, Nagpal K, Malik P. Emerging Pathophysiological Targets of Psoriasis for Future Therapeutic Strategies. Infect Disord Drug Targets 2021; 20:409-422. [PMID: 31288731 DOI: 10.2174/1871526519666190617162701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/04/2019] [Accepted: 04/13/2019] [Indexed: 12/28/2022]
Abstract
Psoriasis is a chronic autoimmune skin disorder which involves complex interactions between genes, keratinocytes, T-cells and inflammatory cells. It affects 2-3% population worldwide. Molecular biology and cellular immunology of psoriasis, when linked with biotechnology and genetic studies can help researchers to understand the pathophysiology of psoriasis. T-cells activation, keratinocyte hyperproliferation, and angiogenesis are the core mechanisms entailed in the development of psoriasis lesion. Investigators are trying to overcome the challenges of complex pathophysiology pathways involved in this disorder. The different possible hypotheses for its pathophysiology such as growth factors, enzymes, inflammation, and genetic factors mediated pathophysiology have been described in the present review paper in detail. Clinically available drugs only control the symptoms of psoriasis but are not effective for the treatment of the disorder completely and are also associated with some side effects such as itching, renal disorders, hematologic, nonmelanoma skin cancer, pulmonary, gastrointestinal toxicity, etc. This paper made an effort to understand the pathophysiological targets, discuss the research done so far and the treatments available for the effective management of psoriasis.
Collapse
Affiliation(s)
- Monu Yadav
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani-127021, Haryana, India
| | - Ishu Sardana
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani-127021, Haryana, India
| | - Amarjeet Sharma
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani-127021, Haryana, India
| | - Nidhi Sharma
- Shri Baba Mastnath Institute of Pharmaceutical Science and Research, Rohtak -124001, Haryana, India
| | - Kalpana Nagpal
- Amity Institute of Pharmacy, Amity University Noida- 201313, Uttar Pradesh, India
| | - Paramjeet Malik
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani-127021, Haryana, India
| |
Collapse
|
10
|
The role of genomic profiling in identifying molecular phenotypes in obstructive lung diseases. Curr Opin Pulm Med 2021; 26:84-89. [PMID: 31714272 DOI: 10.1097/mcp.0000000000000646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW The biology underlying asthma and chronic obstructive pulmonary disease (COPD) is heterogeneous. Targeting therapies to patient subgroups, or 'molecular phenotypes', based on their underlying biology is emerging as an efficacious treatment strategy. This review summarizes the role of airway sample gene expression profiling in understanding molecular phenotypes in obstructive lung disease. RECENT FINDINGS Recent gene expression studies have reinforced the importance of Type two (T2) inflammation in asthma and COPD subgroups. Studies in asthma also suggest that the molecular phenotype with enhanced T2 inflammation is itself heterogeneous with a subgroup that has steroid-refractory inflammation. Other inflammatory pathways are also emerging as implicated in asthma and COPD molecular phenotypes, including Type one and Type 17 adaptive immune responses and proinflammatory cytokines, such as interleukin-6. SUMMARY Genomic profiling studies are advancing our understanding of the complex biology contributing to asthma and COPD molecular phenotypes. Recent studies suggest that asthma and COPD subgroups may benefit from different treatment strategies than those currently in practice.
Collapse
|
11
|
Zhou JS, Li ZY, Xu XC, Zhao Y, Wang Y, Chen HP, Zhang M, Wu YF, Lai TW, Di CH, Dong LL, Liu J, Xuan NX, Zhu C, Wu YP, Huang HQ, Yan FG, Hua W, Wang Y, Xiong WN, Qiu H, Chen T, Weng D, Li HP, Zhou X, Wang L, Liu F, Lin X, Ying SM, Li W, Imamura M, Choi ME, Stampfli MR, Choi AMK, Chen ZH, Shen HH. Cigarette smoke-initiated autoimmunity facilitates sensitisation to elastin-induced COPD-like pathologies in mice. Eur Respir J 2020; 56:13993003.00404-2020. [PMID: 32366484 DOI: 10.1183/13993003.00404-2020] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 04/10/2020] [Indexed: 12/11/2022]
Abstract
It is currently not understood whether cigarette smoke exposure facilitates sensitisation to self-antigens and whether ensuing auto-reactive T cells drive chronic obstructive pulmonary disease (COPD)-associated pathologies.To address this question, mice were exposed to cigarette smoke for 2 weeks. Following a 2-week period of rest, mice were challenged intratracheally with elastin for 3 days or 1 month. Rag1-/- , Mmp12-/- , and Il17a-/- mice and neutralising antibodies against active elastin fragments were used for mechanistic investigations. Human GVAPGVGVAPGV/HLA-A*02:01 tetramer was synthesised to assess the presence of elastin-specific T cells in patients with COPD.We observed that 2 weeks of cigarette smoke exposure induced an elastin-specific T cell response that led to neutrophilic airway inflammation and mucus hyperproduction following elastin recall challenge. Repeated elastin challenge for 1 month resulted in airway remodelling, lung function decline and airspace enlargement. Elastin-specific T cell recall responses were dose dependent and memory lasted for over 6 months. Adoptive T cell transfer and studies in T cells deficient Rag1-/- mice conclusively implicated T cells in these processes. Mechanistically, cigarette smoke exposure-induced elastin-specific T cell responses were matrix metalloproteinase (MMP)12-dependent, while the ensuing immune inflammatory processes were interleukin 17A-driven. Anti-elastin antibodies and T cells specific for elastin peptides were increased in patients with COPD.These data demonstrate that MMP12-generated elastin fragments serve as a self-antigen and drive the cigarette smoke-induced autoimmune processes in mice that result in a bronchitis-like phenotype and airspace enlargement. The study provides proof of concept of cigarette smoke-induced autoimmune processes and may serve as a novel mouse model of COPD.
Collapse
Affiliation(s)
- Jie-Sen Zhou
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,These authors contribute equally to this work
| | - Zhou-Yang Li
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,These authors contribute equally to this work
| | - Xu-Chen Xu
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Zhao
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong Wang
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Pin Chen
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Zhang
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yin-Fang Wu
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian-Wen Lai
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chun-Hong Di
- Dept of Clinical Laboratory, the Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Ling-Ling Dong
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Liu
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nan-Xia Xuan
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Zhu
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan-Ping Wu
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hua-Qiong Huang
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fu-Gui Yan
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Hua
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Wang
- Dept of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Wei-Ning Xiong
- Dept of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Hui Qiu
- Dept of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tao Chen
- Dept of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dong Weng
- Dept of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui-Ping Li
- Dept of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaobo Zhou
- Channing Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lie Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Liu
- Institute for Immunology, Tsinghua University School of Medicine, Tsinghua University-Peking University Jointed Center for Life Sciences, Beijing, China
| | - Xin Lin
- Institute for Immunology, Tsinghua University School of Medicine, Tsinghua University-Peking University Jointed Center for Life Sciences, Beijing, China
| | - Song-Min Ying
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Li
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mitsuru Imamura
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Dept of Medicine, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, NY, USA
| | - Martin R Stampfli
- Dept of Pathology and Molecular Medicine, McMaster Immunology Research Centre, and Dept of Medicine, Firestone Institute for Respiratory Health at St Joseph's Healthcare, McMaster University, Hamilton, ON, Canada.,State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, New York, NY, USA.,These authors contribute equally to this work
| | - Zhi-Hua Chen
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,These authors contribute equally to this work
| | - Hua-Hao Shen
- Key Lab of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China .,State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou, China.,These authors contribute equally to this work
| |
Collapse
|
12
|
High salt diet may promote progression of breast tumor through eliciting immune response. Int Immunopharmacol 2020; 87:106816. [PMID: 32721893 DOI: 10.1016/j.intimp.2020.106816] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/05/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Dietary patterns are believed to regulate tumor progression by altering the tumor microenvironment. Of note, a high salt diet is a risk factor for various diseases. However, the role of high salt intake in the progression of cancers remains unknown. METHODS We constructed an in vivo high salt diet model in MMTV-PyVT mice with spontaneous tumor-forming properties to explore the role of a high salt diet in the progression of breast cancer as well as the modulation of the tumor microenvironment. Also, in vitro experiments were performed to understand the mechanism. RESULTS High salt diet accelerated the development (P < 0.05) and lung metastasis (P < 0.05) of breast cancer in MMTV-PyVT mice, compared to the normal diet model. Moreover, higher frequency of Th17 cells in circulation, tumor tissue and draining lymph node tissue were observed in the high salt diet model (P < 0.05 for all). In vitro, co-culture with Th17 cells facilitated the proliferation, migration and invasion of MCF-7 human breast cancer cells, while these enhanced aggressive behaviors could be reversed by application of 1,25-vitamin D3 which could inhibit the differentiation of Th17 cells (P < 0.001 for all). In vitro, co-culture with Th17 cells activated MAPK signaling in MCF-7 cells (P < 0.001 for all). Consistently, activated MAPK/ERK signaling was observed by immunohistochemistry in breast cancer cell nodes in the high salt diet model (P < 0.05 for all). Mechanistically, higher level of IL-17F could be detected in breast tumors and serum from the high salt diet model through qRT-PCR and ELISA (P < 0.05 for all). IL-17F treatment facilitated the proliferation, migration and invasion of MCF-7 human breast cancer cells and activated MAPK/ERK signaling in MCF-7 cells (P < 0.001 for all). Moreover, the tumor-promoting function induced by Th17 cells and IL-17F could be inhibited by the administration of ERK inhibitor (sch772894) (P < 0.001 for all). Lastly, high concentration NaCl-induced Th17 cells promoted the proliferation, migration and invasion of MCF-7 human breast cancer cells and activated MAPK/ERK signaling in MCF-7 cells which could be inhibited by neutralizing anti-IL-17F (P < 0.001 for all). CONCLUSION High salt intake accelerates the growth of breast cancer and facilitates lung metastasis, as well as increases the level of Th17 cells. Increased Th17 cells might promote the growth of breast cancer via the secretion of IL-17F to activate the MAPK signaling pathway in breast cancer cells.
Collapse
|
13
|
Tejwani V, Yun X, Sikka G, Shimoda L, Suresh K. Airway Epithelial Genomic Signatures in Steroid-Resistant COPD; Role for SMAD3 in Vascular Remodeling in Pulmonary Hypertension; Regulation of Lung Endothelial Cell Function by VEGFR3. Am J Respir Cell Mol Biol 2020; 61:392-394. [PMID: 31038982 DOI: 10.1165/rcmb.2019-0075ro] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Vickram Tejwani
- Division of Pulmonary/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xin Yun
- Division of Pulmonary/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gautam Sikka
- Division of Pulmonary/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Larissa Shimoda
- Division of Pulmonary/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Karthik Suresh
- Division of Pulmonary/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
14
|
Borger JG, Lau M, Hibbs ML. The Influence of Innate Lymphoid Cells and Unconventional T Cells in Chronic Inflammatory Lung Disease. Front Immunol 2019; 10:1597. [PMID: 31354734 PMCID: PMC6637857 DOI: 10.3389/fimmu.2019.01597] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/26/2019] [Indexed: 12/11/2022] Open
Abstract
The lungs are continuously subjected to environmental insults making them susceptible to infection and injury. They are protected by the respiratory epithelium, which not only serves as a physical barrier but also a reactive one that can release cytokines, chemokines, and other defense proteins in response to danger signals, and can undergo conversion to protective mucus-producing goblet cells. The lungs are also guarded by a complex network of highly specialized immune cells and their mediators to support tissue homeostasis and resolve integrity deviation. This review focuses on specialized innate-like lymphocytes present in the lung that act as key sensors of lung insults and direct the pulmonary immune response. Included amongst these tissue-resident lymphocytes are innate lymphoid cells (ILCs), which are classified into five distinct subsets (natural killer, ILC1, ILC2, ILC3, lymphoid tissue-inducer cells), and unconventional T cells including natural killer T (NKT) cells, mucosal-associated invariant T (MAIT) cells, and γδ-T cells. While ILCs and unconventional T cells together comprise only a small proportion of the total immune cells in the lung, they have been found to promote lung homeostasis and are emerging as contributors to a variety of chronic lung diseases including pulmonary fibrosis, allergic airway inflammation, and chronic obstructive pulmonary disease (COPD). A particularly intriguing trait of ILCs that has recently emerged is their plasticity and ability to alter their gene expression profiles and adapt their function in response to environmental cues. The malleable nature of these cells may aid in rapid responses to pathogen but may also have downstream pathological consequences. The role of ILC2s in Th2 allergic airway responses is becoming apparent but the contribution of other ILCs and unconventional T cells during chronic lung inflammation is poorly described. This review presents an overview of our current understanding of the involvement of ILCs and unconventional T cells in chronic pulmonary diseases.
Collapse
Affiliation(s)
- Jessica G Borger
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Maverick Lau
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, Lung Health Research Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Margaret L Hibbs
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
15
|
Klasen C, Meyer A, Wittekind PS, Waqué I, Nabhani S, Kofler DM. Prostaglandin receptor EP4 expression by Th17 cells is associated with high disease activity in ankylosing spondylitis. Arthritis Res Ther 2019; 21:159. [PMID: 31253169 PMCID: PMC6599260 DOI: 10.1186/s13075-019-1948-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/18/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Th17 cells are involved in the pathogenesis of ankylosing spondylitis (AS). However, the mechanism underlying enhanced Th17 cell accumulation in AS remains unknown. The prostaglandin E2 receptor EP2/EP4 signaling pathway plays a critical role in the development of autoimmune Th17 cells. Interestingly, recent genome-wide association studies (GWAS) have identified five risk alleles for AS in PTGER4, the gene encoding for EP4. The aim of this study was to reveal a possible link between EP4 and disease activity in patients with AS. METHODS Th17 cells from patients with AS were analyzed for the transcriptional expression of prostaglandin receptor genes by quantitative RT-PCR. Th17 cells from patients with rheumatoid arthritis (RA) and from healthy individuals served as controls. EP4 receptor expression in Th17 cells was assessed ex vivo by flow cytometry and by western blot. Functional analysis using EP4-specific agonists was performed to reveal how EP4 regulates Th17 cells. RESULTS EP4 is significantly overexpressed in Th17 cells from patients with AS compared to Th17 cells from healthy individuals or patients with RA or psoriatic arthritis (PsA). EP4 upregulation is unique to Th17 cells and is not found in other CD4+ T cell subsets. Specific activation of EP4 drives Th17 cell development and promotes EP4 expression in a positive feedback loop in AS but not in RA or PsA. Mechanistically, EP4 acts via upregulation of the interleukin-23 receptor (IL-23R), by suppressing the RORγt inhibitor FoxO1 and by enhancing STAT3 phosphorylation. Increased EP4 expression levels in Th17 cells from AS patients correlate with high disease activity as defined by a Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) score ≥ 4 (r = 0.7591, p = 0.0016). CONCLUSIONS EP4 is a potential marker of disease activity in patients with AS. Aberrant EP4 expression might contribute to pathogenic Th17 cell accumulation and represent a new target for the treatment of AS.
Collapse
Affiliation(s)
- Charlotte Klasen
- Division of Clinical Immunology and Rheumatology, Department I of Internal Medicine, University of Cologne, Kerpenerstr. 62, 50937, Cologne, Germany
| | - Anja Meyer
- Division of Clinical Immunology and Rheumatology, Department I of Internal Medicine, University of Cologne, Kerpenerstr. 62, 50937, Cologne, Germany
| | - Paula S Wittekind
- Division of Clinical Immunology and Rheumatology, Department I of Internal Medicine, University of Cologne, Kerpenerstr. 62, 50937, Cologne, Germany
| | - Iris Waqué
- Division of Clinical Immunology and Rheumatology, Department I of Internal Medicine, University of Cologne, Kerpenerstr. 62, 50937, Cologne, Germany
| | - Schafiq Nabhani
- Division of Clinical Immunology and Rheumatology, Department I of Internal Medicine, University of Cologne, Kerpenerstr. 62, 50937, Cologne, Germany
| | - David M Kofler
- Division of Clinical Immunology and Rheumatology, Department I of Internal Medicine, University of Cologne, Kerpenerstr. 62, 50937, Cologne, Germany.
| |
Collapse
|
16
|
Wu JH, Zhou M, Jin Y, Meng ZJ, Xiong XZ, Sun SW, Miao SY, Han HL, Tao XN. Generation and Immune Regulation of CD4 +CD25 -Foxp3 + T Cells in Chronic Obstructive Pulmonary Disease. Front Immunol 2019; 10:220. [PMID: 30842769 PMCID: PMC6392103 DOI: 10.3389/fimmu.2019.00220] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 01/25/2019] [Indexed: 11/13/2022] Open
Abstract
The imbalance of CD4+Foxp3+ T cell subsets is reportedly involved in abnormal inflammatory immune responses in patients with chronic obstructive pulmonary disease (COPD). However, the possible role of CD4+CD25-Foxp3+ T cells in immune regulation in COPD remains to be investigated. In the current study, distribution and phenotypic characteristics of CD4+CD25-Foxp3+ T cells from peripheral blood were determined by flow cytometry; the origin, immune function and ultimate fate of CD4+CD25-Foxp3+ T cells were further explored in vitro. It was observed that circulating CD4+CD25-Foxp3+ T cells were significantly increased in stable COPD patients (SCOPD) and resembled central memory or effector memory T cells. Compared with peripheral CD4+CD25+Foxp3+ T cells, peripheral CD4+CD25-Foxp3+ T cells showed a lower expression of Foxp3, CTLA-4, HELIOS, and TIGIT, but a higher expression of CD127 and KI-67, suggesting that CD4+CD25-Foxp3+ T cells lost the expression of Tregs-associated molecules following the reduction in CD25. Unexpectedly, our study found that transforming growth factor-β1 (TGFβ1) decreased CD25 expression and played a critical role in the generation of CD4+CD25-Foxp3+ T cells from CD4+CD25+Foxp3+ T cells. Phenotypic analysis further revealed that both inducible and peripheral CD4+CD25-Foxp3+ T cells exhibited the features of activated conventional T cells. Importantly, memory CD4+CD25-Foxp3+ T cells facilitated the proliferation and differentiation of naïve CD4+ T cells into Th17 cells in the presence of IL-1β, IL-6, IL-23, and TGFβ1. Finally, a fraction of CD4+CD25-Foxp3+ T cells, exhibiting instability and plasticity, were converted to Th17 cells when subjected to Th17 cell-polarizing condition. Taken together, we propose that TGFβ1 is responsible for the generation of CD4+CD25-Foxp3+ T cells, and these cells functionally exert an auxiliary effect on Th17 cells generation and might perpetuate chronic inflammation in COPD.
Collapse
Affiliation(s)
- Jiang-Hua Wu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mei Zhou
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Ji Meng
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian-Zhi Xiong
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng-Wen Sun
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai-Ying Miao
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Li Han
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Nan Tao
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Christenson SA, van den Berge M, Faiz A, Inkamp K, Bhakta N, Bonser LR, Zlock LT, Barjaktarevic IZ, Barr RG, Bleecker ER, Boucher RC, Bowler RP, Comellas AP, Curtis JL, Han MK, Hansel NN, Hiemstra PS, Kaner RJ, Krishnanm JA, Martinez FJ, O’Neal WK, Paine R, Timens W, Wells JM, Spira A, Erle DJ, Woodruff PG. An airway epithelial IL-17A response signature identifies a steroid-unresponsive COPD patient subgroup. J Clin Invest 2019; 129:169-181. [PMID: 30383540 PMCID: PMC6307967 DOI: 10.1172/jci121087] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 10/19/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a heterogeneous smoking-related disease characterized by airway obstruction and inflammation. This inflammation may persist even after smoking cessation and responds variably to corticosteroids. Personalizing treatment to biologically similar "molecular phenotypes" may improve therapeutic efficacy in COPD. IL-17A is involved in neutrophilic inflammation and corticosteroid resistance, and thus may be particularly important in a COPD molecular phenotype. METHODS We generated a gene expression signature of IL-17A response in bronchial airway epithelial brushings from smokers with and without COPD (n = 238), and validated it using data from 2 randomized trials of IL-17 blockade in psoriasis. This IL-17 signature was related to clinical and pathologic characteristics in 2 additional human studies of COPD: (a) SPIROMICS (n = 47), which included former and current smokers with COPD, and (b) GLUCOLD (n = 79), in which COPD participants were randomized to placebo or corticosteroids. RESULTS The IL-17 signature was associated with an inflammatory profile characteristic of an IL-17 response, including increased airway neutrophils and macrophages. In SPIROMICS the signature was associated with increased airway obstruction and functional small airways disease on quantitative chest CT. In GLUCOLD the signature was associated with decreased response to corticosteroids, irrespective of airway eosinophilic or type 2 inflammation. CONCLUSION These data suggest that a gene signature of IL-17 airway epithelial response distinguishes a biologically, radiographically, and clinically distinct COPD subgroup that may benefit from personalized therapy. TRIAL REGISTRATION ClinicalTrials.gov NCT01969344. FUNDING Primary support from the NIH, grants K23HL123778, K12HL11999, U19AI077439, DK072517, U01HL137880, K24HL137013 and R01HL121774 and contracts HHSN268200900013C, HHSN268200900014C, HHSN268200900015C, HHSN268200900016C, HHSN268200900017C, HHSN268200900018C, HHSN268200900019C and HHSN268200900020C.
Collapse
Affiliation(s)
| | - Maarten van den Berge
- University Medical Center Groningen, Department of Pulmonary Diseases and Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Alen Faiz
- University Medical Center Groningen, Department of Pulmonary Diseases and Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Kai Inkamp
- University Medical Center Groningen, Department of Pulmonary Diseases and Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Nirav Bhakta
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Luke R. Bonser
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Lorna T. Zlock
- Department of Pathology, UCSF, San Francisco, California, USA
| | | | - R. Graham Barr
- Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | | | - Richard C. Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | - Jeffrey L. Curtis
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - MeiLan K. Han
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nadia N. Hansel
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Pieter S. Hiemstra
- Department of Pulmonology, University Medical Center, Leiden, Netherlands
| | - Robert J. Kaner
- Department of Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Jerry A. Krishnanm
- Breathe Chicago Center, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Wanda K. O’Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Robert Paine
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Wim Timens
- University Medical Center Groningen, Department of Pathology and Medical Biology and Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - J. Michael Wells
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Avrum Spira
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - David J. Erle
- Department of Medicine, UCSF, San Francisco, California, USA
| | | |
Collapse
|
18
|
Chen T, Qiu H, Zhao MM, Chen SS, Wu Q, Zhou NY, Lu LQ, Song JC, Tang DL, Weng D, Li HP. IL-17A contributes to HSV1 infection-induced acute lung injury in a mouse model of pulmonary fibrosis. J Cell Mol Med 2018; 23:908-919. [PMID: 30378252 PMCID: PMC6349191 DOI: 10.1111/jcmm.13992] [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: 11/15/2017] [Revised: 09/09/2018] [Accepted: 10/07/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Patients with idiopathic pulmonary fibrosis (IPF) often experience acute exacerbation (AE) after an episode of common cold. AIMS To establish a mouse model of virus infection-induced AE-IPF and investigate the mechanism underlying the AE-IPF. METHODS Herpes simplex virus 1 (HSV1) was inoculated intranasally to wild-type (WT) and IL-17A gene knockout (IL-17A-/- ) mice 21 days after intratracheal administration of bleomycin (BLM). RESULTS HSV1 infection caused acute exacerbation in mice with BLM-induced fibrosis. Compared with the BLM+Saline mice, the mice with BLM+HSV1 showed significantly higher acute lung injury (ALI) score (P < 0.0001), lower survival rate (100% vs 21.4%, P < 0.0001), poorer lung function and higher inflammatory response representing by increased total inflammatory cells in bronchoalveolar lavage fluid (BALF) (P = 0.0323), increased proportion of Th17 cells in peripheral blood (P = 0.0004) and higher inflammatory factors in BALF. In addition, HSV1 infection increased the expression of endoplasmic reticulum stress (ERS)-related proteins in mice with BLM-induced fibrosis. The inhibition of ERS by tauroursodeoxycholic acid (TUDCA, an ERS inhibitor) significantly reduced the IL-17A levels in BALF (P = 0.0140) and TH17 cells in the peripheral blood (P = 0.0084) of mice with BLM+HSV1, suggesting that suppression of ERS may reduce TH17 response in mice with AE-IPF. Compared with WT mice with BLM+HSV1, IL-17A-/- mice with BLM+HSV1 had lower ALI score (P = 0.0119), higher survival rate (78.6% vs 21.4%, P = 0.004), improved lung function, and milder inflammatory response. CONCLUSIONS HSV1 infection in addition to BLM-induced IPF can successfully establish AE-IPF in mice. IL-17A and ERS promote lung inflammation in AE-IPF development.
Collapse
Affiliation(s)
- Tao Chen
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Hui Qiu
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Meng-Meng Zhao
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Shan-Shan Chen
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Soochow University, Suzhou, China
| | - Qin Wu
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Nian-Yu Zhou
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Li-Qin Lu
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Jia-Cui Song
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Soochow University, Suzhou, China
| | - Dan-Li Tang
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Dong Weng
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Hui-Ping Li
- Department of Respiratory Medicine, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| |
Collapse
|
19
|
Kim HY, Kim MS, Kim SH, Joen D, Lee K. Protective Effects of Nintedanib against Polyhexamethylene Guanidine Phosphate-Induced Lung Fibrosis in Mice. Molecules 2018; 23:molecules23081974. [PMID: 30087305 PMCID: PMC6222351 DOI: 10.3390/molecules23081974] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 12/18/2022] Open
Abstract
Nintedanib (NDN), a tyrosine kinase inhibitor, has been shown to have anti-tumor, anti-inflammatory, and anti-fibrotic effects in several reports. We investigated the protective effects of NDN against polyhexamethylene guanidine phosphate (PHMG)-induced lung fibrosis in mice. The following three experimental groups were evaluated: (1) vehicle control; (2) PHMG (1.1 mg/kg); and (3) PHMG & NDN (60 mg/kg). PHMG induced pulmonary inflammation and fibrosis by intratracheal instillation in mice. In contrast, NDN treatment effectively alleviated the PHMG induced lung injury, and attenuated the number of total cells and inflammatory cells in the bronchoalveolar lavage fluid, including the fibrotic histopathological changes, and also reduced the hydroxyproline content. NDN also significantly decreased the expression of inflammatory cytokines and fibrotic factors, and the activation of the NLRP3 inflammasome in lung tissues. These results suggest that NDN may mitigate the inflammatory response and development of pulmonary fibrosis in the lungs of mice treated with PHMG.
Collapse
Affiliation(s)
- Hyeon-Young Kim
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, 30, Baekhak 1-gil, Jeongeup-si 56212, Korea.
- Department of Toxicology Evaluation, Graduate School of Pre-Clinical Laboratory Science, Konyang University, Daejeon 35365, Korea.
| | - Min-Seok Kim
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, 30, Baekhak 1-gil, Jeongeup-si 56212, Korea.
| | - Sung-Hwan Kim
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, 30, Baekhak 1-gil, Jeongeup-si 56212, Korea.
- Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon 34113, Korea.
| | - Doin Joen
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, 30, Baekhak 1-gil, Jeongeup-si 56212, Korea.
| | - Kyuhong Lee
- National Center for Efficacy Evaluation of Respiratory Disease Product, Korea Institute of Toxicology, 30, Baekhak 1-gil, Jeongeup-si 56212, Korea.
- Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon 34113, Korea.
| |
Collapse
|
20
|
Fei X, Zhang PY, Zhang X, Zhang GQ, Bao WP, Zhang YY, Zhang M, Zhou X. IL-17A Monoclonal Antibody Partly Reverses the Glucocorticoids Insensitivity in Mice Exposed to Ozonec. Inflammation 2018; 40:788-797. [PMID: 28194607 PMCID: PMC5429348 DOI: 10.1007/s10753-017-0523-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Exposure to ozone has been associated with airway inflammation and glucocorticoid insensitivity. This study aimed to observe the capacity of anti-murine interleukin-17A monoclonal antibody (IL-17mAb) to reverse ozone-induced glucocorticoid insensitivity and to detect its effects with glucocorticoids in protecting against airway inflammation. After C57/BL6 mice were exposed to ozone (2.5 ppm; 3 h) for 12 times over 6 weeks, PBS, IL-17mAb (50 ug/ml), dexamethasone (2 mg/kg), and combination administration of IL-17mAb (50 ug/ml) and dexamethasone (2 mg/kg) were intraperitoneally injected into mice at a dose of 0.1 ml, respectively, for 10 times over 5 weeks. At sacrifice, lung histology, airway inflammatory cells, levels of related cytokines in bronchoalveolar lavage fluid (BALF), and serum were analyzed, airway inflammatory cell infiltration density and mean linear intercept (Lm) were measured, the expression of IL-17A mRNA, glucocorticoid receptors (GR), NF-κB, and p38 mitogen-activated protein kinase (MAPK) phosphorylation were determined. We found that combination administration markedly reduced ozone-induced total inflammatory cells, especially neutrophils; inhibited levels of cytokines, including IL-8, IL-17A, and TNF-α in BALF; and suppressed airway inflammatory cell infiltration density and Lm. Additionally, combination administration significantly elevated levels of IFN-γ in BALF, decreased the dexamethasone-induced increase of IL-17A mRNA, and increased the expression of GR and decrement of NF-κB and p38MAPK phosphorylation, which are also related to glucocorticoids insensitivity. Collectively, combination administration shows profound efficacy in inhibiting certain cytokines, and IL-17 mAb partly improved the glucocorticoids insensitivity via modulating the enhanced production rate and improving expression of IL-17A induced by glucocorticoids administration and p38MAPK, NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xia Fei
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Peng-Yu Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Xue Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Guo-Qing Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Wu-Ping Bao
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Ying-Ying Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Min Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China.
| | - Xin Zhou
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China.
| |
Collapse
|
21
|
JAK/STAT inhibitors and other small molecule cytokine antagonists for the treatment of allergic disease. Ann Allergy Asthma Immunol 2018; 120:367-375. [PMID: 29454096 DOI: 10.1016/j.anai.2018.02.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/08/2018] [Accepted: 02/12/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To provide an overview of janus kinase (JAK), chemoattractant receptor homologous molecule expressed on TH2 cells (CRTH2), and phosphodiesterase 4 (PDE4) inhibitors in allergic disorders. DATA SOURCES PubMed literature review. STUDY SELECTIONS Articles included in this review discuss the emerging mechanism of action of small molecule inhibitors and their use in the treatment of atopic dermatitis (AD), asthma, and allergic rhinitis (AR). RESULTS Allergic diseases represent a spectrum of diseases, including AD, asthma, and AR. For decades, these diseases have been primarily characterized by increased TH2 signaling and downstream inflammation. In recent years, additional research has identified disease phenotypes and subsets of patients with non-Th2 mediated inflammation. The increasing heterogeneity of disease has prompted investigators to move away from wide-ranging treatment approaches with immunosuppressive agents, such as corticosteroids, to consider more targeted immunomodulatory approaches focused on specific pathways. In the past decade, inhibitors that target JAK signaling, PDE4, and CRTH2 have been explored for their potential activity in models of allergic disease and therapeutic benefit in clinical trials. Interestingly, although JAK inhibitors provide an opportunity to interfere with cytokine signaling and could be beneficial in a broad range of allergic diseases, current clinical trials are focused on the treatment of AD. Conversely, both PDE4 and CRTH2 inhibitors have been evaluated in a spectrum of allergic diseases. This review summarizes the varying degrees of success that these small molecules have demonstrated across allergic diseases. CONCLUSION Emerging therapies currently in development may provide more consistent benefit to patients with allergic diseases by specifically targeting inflammatory pathways important for disease pathogenesis.
Collapse
|
22
|
Bullone M, Lavoie JP. The Contribution of Oxidative Stress and Inflamm-Aging in Human and Equine Asthma. Int J Mol Sci 2017; 18:ijms18122612. [PMID: 29206130 PMCID: PMC5751215 DOI: 10.3390/ijms18122612] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
Aging is associated with a dysregulation of the immune system, leading to a general pro-inflammatory state of the organism, a process that has been named inflamm-aging. Oxidative stress has an important role in aging and in the regulation of immune responses, probably playing a role in the development of age-related diseases. The respiratory system function physiologically declines with the advancement of age. In elderly asthmatic patients, this may contribute to disease expression. In this review, we will focus on age-related changes affecting the immune system and in respiratory structure and function that could contribute to asthma occurrence, and/or clinical presentation in the elderly. Also, naturally occurring equine asthma will be discussed as a possible model for studying the importance of oxidative stress and immun-aging/inflamm-aging in humans.
Collapse
Affiliation(s)
- Michela Bullone
- Department of Clinical and Biological Sciences, University of Turin, AUO San Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano, Italy.
| | - Jean-Pierre Lavoie
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, 3200 Rue Sicotte, St-Hyacinthe, QC J2S 2M2, Canada.
| |
Collapse
|
23
|
Yang X, Huo B, Zhong X, Su W, Liu W, Li Y, He Z, Bai J. Imbalance between Subpopulations of Regulatory T Cells in Patients with Acute Exacerbation of COPD. COPD 2017; 14:618-625. [PMID: 29166179 DOI: 10.1080/15412555.2017.1385055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Human regulatory T cells (Tregs) have been reported to be not significantly different in the peripheral blood of patients with chronic obstructive pulmonary disease (COPD) and healthy controls. Recent research has identified some new markers for Tregs and indicated that Tregs are composed of distinct subpopulations. The aim of the study was to describe the changing patterns of circulating Treg subpopulations in patients with acute exacerbation of COPD (AECOPD) and healthy controls, and to explore their potential roles in AECOPD pathogenesis. Blood samples were obtained from 30 never-smokers with normal lung function and 30 patients with COPD before and after they had an exacerbation. The proportions of Treg subpopulations were evaluated using flow cytometry. In the peripheral blood, decreased proportions of CD4+CD25+CD127low Tregs, CD4+CD25+CD45RA+ Tregs, and CD4+CD25+CD62L+ Tregs and an increased proportion of CD4+CD25+CD45RO+ Tregs were found in patients with stable COPD compared with non-smokers with normal lung function. The patients showed further changes in Treg subpopulations when they had an AECOPD, with an overall decrease in a suppressive subset, indicating that the immune negative regulatory population of Tregs did not play an effective role. Immune homeostasis favored inflammation, and a negative correlation between the circulating tumor necrosis factor-alpha and the proportions of CD4+CD25+CD62L+ cells (r = -0.698, p < 0.05) in patients with AECOPD was found. The imbalance between the suppressive subsets and the proinflammatory subset of Tregs and the decline of Treg subpopulations with immunosuppressive activity may play important roles in AECOPD progression.
Collapse
Affiliation(s)
- Xia Yang
- a Department of Respiratory Medicine , First Affiliated Hospital of Guangxi Medical University , Nanning , China
| | - Bo Huo
- a Department of Respiratory Medicine , First Affiliated Hospital of Guangxi Medical University , Nanning , China
| | - Xiaoning Zhong
- a Department of Respiratory Medicine , First Affiliated Hospital of Guangxi Medical University , Nanning , China
| | - Wenyan Su
- a Department of Respiratory Medicine , First Affiliated Hospital of Guangxi Medical University , Nanning , China
| | - Wenting Liu
- a Department of Respiratory Medicine , First Affiliated Hospital of Guangxi Medical University , Nanning , China
| | - Yumei Li
- a Department of Respiratory Medicine , First Affiliated Hospital of Guangxi Medical University , Nanning , China
| | - Zhiyi He
- a Department of Respiratory Medicine , First Affiliated Hospital of Guangxi Medical University , Nanning , China
| | - Jing Bai
- a Department of Respiratory Medicine , First Affiliated Hospital of Guangxi Medical University , Nanning , China
| |
Collapse
|
24
|
Gan PY, Fujita T, Ooi JD, Alikhan MA, Dick J, Shim R, Odobasic D, O'Sullivan KM, Kitching AR, Holdsworth SR. Pathogenic Role for γδ T Cells in Autoimmune Anti-Myeloperoxidase Glomerulonephritis. THE JOURNAL OF IMMUNOLOGY 2017; 199:3042-3050. [PMID: 28954887 DOI: 10.4049/jimmunol.1602025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 08/24/2017] [Indexed: 01/13/2023]
Abstract
Myeloperoxidase (MPO) anti-neutrophil cytoplasmic Ab (ANCA)-associated vasculitis results from autoimmunity to MPO. IL-17A plays a critical role in generating this form of autoimmune injury but its cell of origin is uncertain. We addressed the hypothesis that IL-17A-producing γδ T cells are a nonredundant requisite in the development of MPO autoimmunity and glomerulonephritis (GN). We studied MPO-ANCA GN in wild type, αβ, or γδ T cell-deficient (C57BL/6, βTCR-/- , and δTCR-/- respectively) mice. Both T cell populations played important roles in the generation of autoimmunity to MPO and GN. Humoral autoimmunity was dependent on intact αβ T cells but was unaffected by γδ T cell deletion. Following MPO immunization, activated γδ T cells migrate to draining lymph nodes. Studies in δTCR-/- and transfer of γδ T cells to δTCR-/- mice show that γδ T cells facilitate the generation of anti-MPO autoimmunity and GN. δTCR-/- mice that received IL-17A-/- γδ T cells demonstrate that the development of anti-MPO autoimmunity and GN are dependent on γδ T cell IL-17A production. Finally, transfer of anti-MPO CD4+ T cell clones to naive δTCR-/- and wild type mice with planted glomerular MPO shows that γδ T cells are also necessary for recruitment of anti-MPO αβ CD4+ effector T cells. This study demonstrates that IL-17A produced by γδ T cells plays a critical role in the pathogenesis of MPO-ANCA GN by promoting the development of MPO-specific αβ T cells.
Collapse
Affiliation(s)
- Poh-Yi Gan
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia; .,Department of Immunology, Monash Health, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Takeshi Fujita
- Department of Cardiology, Respiratory Medicine and Nephrology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan; and
| | - Joshua Daniel Ooi
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Maliha Asghar Alikhan
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Jonathan Dick
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia.,Department of Nephrology, Monash Health, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Raymond Shim
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Dragana Odobasic
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Kim Maree O'Sullivan
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Arthur Richard Kitching
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia.,Department of Nephrology, Monash Health, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Stephen Roger Holdsworth
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia.,Department of Immunology, Monash Health, Monash Medical Centre, Clayton, Victoria 3168, Australia.,Department of Nephrology, Monash Health, Monash Medical Centre, Clayton, Victoria 3168, Australia
| |
Collapse
|
25
|
Okasha EF, Bayomy NA, Abdelaziz EZ. Effect of Topical Application of Black Seed Oil on Imiquimod-Induced Psoriasis-like Lesions in the Thin Skin of Adult Male Albino Rats. Anat Rec (Hoboken) 2017; 301:166-174. [PMID: 28926201 DOI: 10.1002/ar.23690] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 08/07/2017] [Accepted: 08/09/2017] [Indexed: 12/15/2022]
Abstract
Psoriasis is a chronic inflammatory skin disease that affects about 1%-3% of the world's population. Black seed oil, i.e., the oil extracted from black seeds (Nigella sativa seeds), possesses a broad spectrum of pharmacological actions including anti-inflammatory, immunostimulatory, and antioxidant properties. This study aimed to investigate the effect of black seed oil on imiquimod (IMQ) induced psoriasis-like skin lesions. To this end, 30 male albino rats were divided into three groups: group I, control group; group II, psoriasis-induced group receiving daily topical applications of IMQ cream (5%) on the shaved back skin for 10 consecutive days; and group III, black seed oil group receiving a daily topical dose of black seed oil 5 mg/kg body weight for 10 days after induction of psoriasis. Animals of all groups were sacrificed and specimens obtained from the skin of the central part of the back were processed for histological and immunohistochemical staining with proliferating cell nuclear antigen (PCNA). IMQ application led to epidermal inflammation, hyperplasia and alterations in the normal appearance of keratinocytes with degenerative changes observed at both light and electron microscopic levels. Collagenous fibers were abundant in the dermis and PCNA-positive cells were detected in all layers of the epidermis. However, topical use of black seed oil strongly inhibited IMQ-induced psoriasis-like inflammation and alleviated all epidermal and dermal changes observed after IMQ application, allowing us to conclude that black seed oil can be used as an adjuvant topical therapy for treating psoriasis. Anat Rec, 2017. © 2017 Wiley Periodicals, Inc. Anat Rec, 301:166-174, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ebtsam F Okasha
- Histology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Naglaa A Bayomy
- Histology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Eman Z Abdelaziz
- Pharmacology department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
26
|
Cova E, Inghilleri S, Pandolfi L, Morosini M, Magni S, Colombo M, Piloni D, Finetti C, Ceccarelli G, Benedetti L, Cusella MG, Agozzino M, Corsi F, Allevi R, Mrakic-Sposta S, Moretti S, De Gregori S, Prosperi D, Meloni F. Bioengineered gold nanoparticles targeted to mesenchymal cells from patients with bronchiolitis obliterans syndrome does not rise the inflammatory response and can be safely inhaled by rodents. Nanotoxicology 2017; 11:534-545. [PMID: 28415888 DOI: 10.1080/17435390.2017.1317862] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The use of gold nanoparticles (GNPs) as drug delivery system represents a promising issue for diseases without effective pharmacological treatment due to insufficient local drug accumulation and excessive systemic toxicity. Bronchiolitis obliterans syndrome (BOS) represents about 70% of cases of chronic lung allograft dysfunction, the main challenge to long-term lung transplantation. It is believed that due to repeated insults to epithelial bronchiolar cells local inflammatory response creates a milieu that favors epithelial-mesenchymal transition and activation of local mesenchymal cells (MCs) leading to airway fibro-obliteration. In a previous work, we engineered GNPs loaded with the mammalian target of rapamycin inhibitor everolimus, specifically decorated with an antibody against CD44, a surface receptor expressed by primary MCs isolated from bronchoalveolar lavage of BOS patients. We proved in vitro that these GNPs (GNP-HCe) were able to specifically inhibit primary MCs without affecting the bronchial epithelial cell. In the present work, we investigated the effect of these bioengineered nanoconstructs on inflammatory cells, given that a stimulating effect on macrophages, neutrophils or lymphocytes is strongly unwanted in graft airways since it would foster fibrogenesis. In addition, we administered GNP-HCe by the inhalatory route to normal mice for a preliminary assessment of their pulmonary and peripheral (liver, spleen and kidney) uptake. By these experiments, an evaluation of tissue toxicity was also performed. The present study proves that our bioengineered nanotools do not rise an inflammatory response and, under the tested inhalatory conditions that were used, are non-toxic.
Collapse
Affiliation(s)
- Emanuela Cova
- a Clinica di Malattie dell'Apparato Respiratorio , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Simona Inghilleri
- a Clinica di Malattie dell'Apparato Respiratorio , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Laura Pandolfi
- b Dipartimento di Biotecnologie e Bioscienze , Università di Milano-Bicocca , Milano , Italy
| | - Monica Morosini
- a Clinica di Malattie dell'Apparato Respiratorio , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Sara Magni
- a Clinica di Malattie dell'Apparato Respiratorio , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Miriam Colombo
- b Dipartimento di Biotecnologie e Bioscienze , Università di Milano-Bicocca , Milano , Italy
| | - Davide Piloni
- c Dipartimento di Medicina Interna, Unità di Pneumologia , Università degli Studi di Pavia , Pavia , Italy
| | - Chiara Finetti
- b Dipartimento di Biotecnologie e Bioscienze , Università di Milano-Bicocca , Milano , Italy
| | - Gabriele Ceccarelli
- d Istituto di Anatomia Umana, Dipartimento di Salute Pubblica, Medicina Sperimentale e Forense , Università degli Studi di Pavia , Pavia , Italy
| | - Laura Benedetti
- d Istituto di Anatomia Umana, Dipartimento di Salute Pubblica, Medicina Sperimentale e Forense , Università degli Studi di Pavia , Pavia , Italy
| | - Maria Gabriella Cusella
- d Istituto di Anatomia Umana, Dipartimento di Salute Pubblica, Medicina Sperimentale e Forense , Università degli Studi di Pavia , Pavia , Italy
| | - Manuela Agozzino
- e Centro per le Malattie Cardiovascolari Ereditarie , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Fabio Corsi
- f Dipartimento di Scienze Biomediche e Cliniche L. Sacco , Università degli Studi di Milano , Pavia , Italy.,g Chirurgia Senologica , ICS Maugeri S.p.A. SB , Pavia , Italy
| | - Raffaele Allevi
- f Dipartimento di Scienze Biomediche e Cliniche L. Sacco , Università degli Studi di Milano , Pavia , Italy
| | - Simona Mrakic-Sposta
- h Istituto di Bioimmagini e Fisiologia Molecolare , Consiglio Nazionale delle Ricerche (CNR) , Segrate , Milano , Italia
| | - Sarah Moretti
- h Istituto di Bioimmagini e Fisiologia Molecolare , Consiglio Nazionale delle Ricerche (CNR) , Segrate , Milano , Italia
| | - Simona De Gregori
- i S.S.di Farmacocinetica Clinica e Sperimentale , IRCCS Fondazione Policlinico San Matteo , Pavia , Italy
| | - Davide Prosperi
- b Dipartimento di Biotecnologie e Bioscienze , Università di Milano-Bicocca , Milano , Italy
| | - Federica Meloni
- c Dipartimento di Medicina Interna, Unità di Pneumologia , Università degli Studi di Pavia , Pavia , Italy
| |
Collapse
|
27
|
Zenobia C, Hajishengallis G. Basic biology and role of interleukin-17 in immunity and inflammation. Periodontol 2000 2017; 69:142-59. [PMID: 26252407 DOI: 10.1111/prd.12083] [Citation(s) in RCA: 289] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2014] [Indexed: 02/06/2023]
Abstract
Interleukin-17 (also known as interleukin-17A) is a key cytokine that links T-cell activation to neutrophil mobilization and activation. As such, interleukin-17 can mediate protective innate immunity to pathogens or contribute to the pathogenesis of inflammatory diseases, such as psoriasis and rheumatoid arthritis. This review summarizes the basic biology of interleukin-17 and discusses its emerging role in periodontal disease. The current burden of evidence from human and animal model studies suggests that the net effect of interleukin-17 signaling promotes disease development. In addition to promoting neutrophilic inflammation, interleukin-17 has potent pro-osteoclastogenic effects that are likely to contribute to the pathogenesis of periodontitis, rheumatoid arthritis and other diseases involving bone immunopathology. Systemic treatments with anti-interleukin-17 biologics have shown promising results in clinical trials for psoriasis and rheumatoid arthritis; however, their impact on the highly prevalent periodontal disease has not been investigated or reported. Future clinical trials, preferably using locally administered interleukin-17 blockers, are required to implicate conclusivelyinterleukin-17 in periodontitis and, more importantly, to establish an effective adjunctive treatment for this oral inflammatory disease.
Collapse
|
28
|
Vandermeulen E, Lammertyn E, Verleden SE, Ruttens D, Bellon H, Ricciardi M, Somers J, Bracke KR, Van Den Eynde K, Tousseyn T, Brusselle GG, Verbeken EK, Verschakelen J, Emonds MP, Van Raemdonck DE, Verleden GM, Vos R, Vanaudenaerde BM. Immunological diversity in phenotypes of chronic lung allograft dysfunction: a comprehensive immunohistochemical analysis. Transpl Int 2016; 30:134-143. [PMID: 27933655 DOI: 10.1111/tri.12882] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/10/2016] [Accepted: 10/28/2016] [Indexed: 11/30/2022]
Abstract
Chronic rejection after organ transplantation is defined as a humoral- and cell-mediated immune response directed against the allograft. In lung transplantation, chronic rejection is nowadays clinically defined as a cause of chronic lung allograft dysfunction (CLAD), consisting of different clinical phenotypes including restrictive allograft syndrome (RAS) and bronchiolitis obliterans syndrome (BOS). However, the differential role of humoral and cellular immunity is not investigated up to now. Explant lungs of patients with end-stage BOS (n = 19) and RAS (n = 18) were assessed for the presence of lymphoid (B and T cells) and myeloid cells (dendritic cells, eosinophils, mast cells, neutrophils, and macrophages) and compared to nontransplant control lung biopsies (n = 21). All myeloid cells, with exception of dendritic cells, were increased in RAS versus control (neutrophils, eosinophils, and mast cells: all P < 0.05, macrophages: P < 0.001). Regarding lymphoid cells, B cells and cytotoxic T cells were increased remarkably in RAS versus control (P < 0.001) and in BOS versus control (P < 0.01). Interestingly, lymphoid follicles were restricted to RAS (P < 0.001 versus control and P < 0.05 versus BOS). Our data suggest an immunological diversity between BOS and RAS, with a more pronounced involvement of the B-cell response in RAS characterized by a structural organization of lymphoid follicles. This may impact future therapeutic approaches.
Collapse
Affiliation(s)
- Elly Vandermeulen
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Elise Lammertyn
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Stijn E Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - David Ruttens
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Hannelore Bellon
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Mario Ricciardi
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Jana Somers
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Ken R Bracke
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Kathleen Van Den Eynde
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KULeuven, Leuven, Belgium
| | - Thomas Tousseyn
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KULeuven, Leuven, Belgium
| | - Guy G Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Erik K Verbeken
- Translational Cell & Tissue Research Unit, Department of Imaging & Pathology, KULeuven, Leuven, Belgium
| | - Johny Verschakelen
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | | | - Dirk E Van Raemdonck
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Geert M Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Robin Vos
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Lung Transplant Unit, Division of Respiratory Disease, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium
| |
Collapse
|
29
|
Ostadkarampour M, Müller M, Öckinger J, Kullberg S, Lindén A, Eklund A, Grunewald J, Wahlström J. Distinctive Regulatory T Cells and Altered Cytokine Profile Locally in the Airways of Young Smokers with Normal Lung Function. PLoS One 2016; 11:e0164751. [PMID: 27798682 PMCID: PMC5087844 DOI: 10.1371/journal.pone.0164751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/02/2016] [Indexed: 12/31/2022] Open
Abstract
Smoking influences the immune system in different ways and, hypothetically, effects on pulmonary effector and regulatory T cells emerge as potentially detrimental. Therefore, we characterized the frequencies and characteristics of CD4+ and CD8+ T cell subsets in the blood and lungs of young tobacco smokers. Bronchoalveolar lavage (BAL) and peripheral blood were obtained from healthy moderate smokers (n = 18; 2–24 pack-years) and never-smokers (n = 15), all with normal lung function. Cells were stimulated ex vivo and key intracellular cytokines (IFNγ, IL-17, IL-10 and TNFα) and transcription factors (Foxp3, T-bet and Helios) were analyzed using flow cytometry. Our results indicate that smoking is associated with a decline in lung IL-17+ CD4+ T cells, increased IFNγ+ CD8+ T cells and these alterations relate to the history of daily cigarette consumption. There is an increased fraction of Foxp3+ regulatory T cells being Helios- in the lungs of smokers. Cytokine production is mainly confined to the Helios- T cells, both in regulatory and effector subsets. Moreover, we detected a decline of Helios+Foxp3- postulated regulatory CD8+ T cells in smokers. These alterations in the immune system are likely to increase risk for infection and may have implications for autoimmune processes initiated in the lungs among tobacco smokers.
Collapse
Affiliation(s)
- Mahyar Ostadkarampour
- Respiratory Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Malin Müller
- Rheumatology Research Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| | - Johan Öckinger
- Respiratory Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| | - Susanna Kullberg
- Respiratory Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Anders Lindén
- Unit for Lung and Airway Research, Institute of Environmental Medicine (IMM), Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Anders Eklund
- Respiratory Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Johan Grunewald
- Respiratory Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Jan Wahlström
- Respiratory Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
He Y, Wen Q, Yao F, Xu D, Huang Y, Wang J. Gut-lung axis: The microbial contributions and clinical implications. Crit Rev Microbiol 2016; 43:81-95. [PMID: 27781554 DOI: 10.1080/1040841x.2016.1176988] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Gut microbiota interacts with host immune system in ways that influence the development of disease. Advances in respiratory immune system also broaden our knowledge of the interaction between host and microbiome in the lung. Increasing evidence indicated the intimate relationship between the gastrointestinal tract and respiratory tract. Exacerbations of chronic gut and lung disease have been shown to share key conceptual features with the disorder and dysregulation of the microbial ecosystem. In this review, we discuss the impact of gut and lung microbiota on disease exacerbation and progression, and the recent understanding of the immunological link between the gut and the lung, the gut-lung axis.
Collapse
Affiliation(s)
- Yang He
- a Department of Cancer Center, Union Hospital , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Qu Wen
- a Department of Cancer Center, Union Hospital , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Fangfang Yao
- a Department of Cancer Center, Union Hospital , Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Dong Xu
- b Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Yuancheng Huang
- b Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | - Junshuai Wang
- c Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| |
Collapse
|
31
|
|
32
|
Yamada Y, Vandermeulen E, Heigl T, Somers J, Vaneylen A, Verleden SE, Bellon H, De Vleeschauwer S, Verbeken EK, Van Raemdonck DE, Vos R, Verleden GM, Jungraithmayr W, Vanaudenaerde BM. The role of recipient derived interleukin-17A in a murine orthotopic lung transplant model of restrictive chronic lung allograft dysfunction. Transpl Immunol 2016; 39:10-17. [PMID: 27737799 DOI: 10.1016/j.trim.2016.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/10/2016] [Accepted: 10/10/2016] [Indexed: 02/06/2023]
Abstract
The single most important cause of late mortality after lung transplantation is chronic lung allograft dysfunction (CLAD). However, the pathological development of CLAD was not as simple as previously presumed and subclassification phenotypes, bronchiolitis obliterans syndrome (BOS) and restrictive CLAD (rCLAD), have been introduced. We want to re-investigate how CLAD manifests in the murine orthotopic lung transplant model and investigate the role of interleukin 17A (IL-17A) within this model. Orthotopic LTx was performed in CB57BL/6, IL-17 WT and IL-17 KO mice. In a first experiment, CB57BL/6 mice receiving an isograft (CB57BL/6) or allograft (BALB/C) were compared. In a second experiment IL-17 WT and IL-17 KO mice (both CB57BL/6 background) received an allograft (BALB/C). Mice received daily immunosuppression with steroids and cyclosporine and were sacrificed 10weeks after transplantation for histopathological analysis by an experienced lung pathologist. After murine orthotopic lung transplantation, the allograft histopathologically presented features of human rCLAD (i.e. overt inflammation, pleural/parenchymal fibrosis and obliterative bronchiolitis). In the IL-17A KO group, less inflammation in the bronchovascular axis (p=0.03) was observed and a non-significant trend towards less bronchovascular fibrosis, pleural/septal inflammation and fibrosis, and parenchymal inflammation and fibrosis when compared to WT mice. The major mismatch orthotopic lung transplant model resembles features of human rCLAD. IL-17A mediated immunity is involved in the inflammatory component, but had little influence on the degree of fibrosis. Further mechanistic and therapeutic studies in this mouse model are needed to fully understand the mechanisms in rCLAD.
Collapse
Affiliation(s)
- Y Yamada
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland; Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - E Vandermeulen
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - T Heigl
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - J Somers
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - A Vaneylen
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - S E Verleden
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - H Bellon
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - S De Vleeschauwer
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - E K Verbeken
- Department of Pathology, UZ Leuven, Leuven, Belgium
| | - D E Van Raemdonck
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - R Vos
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - G M Verleden
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - W Jungraithmayr
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - B M Vanaudenaerde
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium.
| |
Collapse
|
33
|
Southworth T, Plumb J, Gupta V, Pearson J, Ramis I, Lehner MD, Miralpeix M, Singh D. Anti-inflammatory potential of PI3Kδ and JAK inhibitors in asthma patients. Respir Res 2016; 17:124. [PMID: 27716212 PMCID: PMC5051065 DOI: 10.1186/s12931-016-0436-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/17/2016] [Indexed: 01/20/2023] Open
Abstract
Background Phosphatidylinositol 3-kinase delta (PI3Kδ) and Janus-activated kinases (JAK) are both novel anti-inflammatory targets in asthma that affect lymphocyte activation. We have investigated the anti-inflammatory effects of PI3Kδ and JAK inhibition on cytokine release from asthma bronchoalveolar lavage (BAL) cells and T-cell activation, and measured lung PI3Kδ and JAK signalling pathway expression. Method Cells isolated from asthma patients and healthy subjects were treated with PI3Kδ or JAK inhibitors, and/or dexamethasone, before T-cell receptor stimulation. Levels of IFNγ, IL-13 and IL-17 were measured by ELISA and flow cytometry was used to assess T-cell activation. PI3Kδ, PI3Kγ, phosphorylated protein kinase B (pAKT) and Signal Transducer and Activator of Transcription (STAT) protein expression were assessed by immunohistochemistry in bronchial biopsy tissue from asthma patients and healthy subjects. PI3Kδ expression in BAL CD3 cells was measured by flow cytometry. Results JAK and PI3Kδ inhibitors reduced cytokine levels from both asthma and healthy BAL cells. Combining dexamethasone with either a JAK or PI3Kδ inhibitor showed an additive anti-inflammatory effect. JAK and PI3Kδ inhibitors were shown to have direct effects on T-cell activation. Immunohistochemistry showed increased numbers of PI3Kδ expressing cells in asthma bronchial tissue compared to controls. Asthma CD3 cells in BAL expressed higher levels of PI3Kδ protein compared to healthy cells. Conclusions Targeting PI3Kδ or JAK may prove effective in reducing T-cell activation and the resulting cytokine production in asthma. Electronic supplementary material The online version of this article (doi:10.1186/s12931-016-0436-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Thomas Southworth
- The University of Manchester; Division of Infection, Immunity & Respiratory Medicine; Manchester Academic Health Science Centre; University Hospital South Manchester NHS Foundation Trust, Southmoor Road, Manchester, M23 9LT, UK.
| | - Jonathan Plumb
- The University of Manchester; Division of Infection, Immunity & Respiratory Medicine; Manchester Academic Health Science Centre; University Hospital South Manchester NHS Foundation Trust, Southmoor Road, Manchester, M23 9LT, UK
| | - Vandana Gupta
- The University of Manchester; Division of Infection, Immunity & Respiratory Medicine; Manchester Academic Health Science Centre; University Hospital South Manchester NHS Foundation Trust, Southmoor Road, Manchester, M23 9LT, UK
| | - James Pearson
- The University of Manchester; Division of Infection, Immunity & Respiratory Medicine; Manchester Academic Health Science Centre; University Hospital South Manchester NHS Foundation Trust, Southmoor Road, Manchester, M23 9LT, UK
| | - Isabel Ramis
- Almirall R&D Center, Sant Feliu de Llobregat, Barcelona, Spain
| | - Martin D Lehner
- Almirall R&D Center, Sant Feliu de Llobregat, Barcelona, Spain
| | | | - Dave Singh
- The University of Manchester; Division of Infection, Immunity & Respiratory Medicine; Manchester Academic Health Science Centre; University Hospital South Manchester NHS Foundation Trust, Southmoor Road, Manchester, M23 9LT, UK
| |
Collapse
|
34
|
Peters M, Köhler-Bachmann S, Lenz-Habijan T, Bufe A. Influence of an Allergen-Specific Th17 Response on Remodeling of the Airways. Am J Respir Cell Mol Biol 2016. [PMID: 26222011 DOI: 10.1165/rcmb.2014-0429oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We showed previously that sensitization of mice with dendritic cells (DCs) via the airways depends on activation of these cells with LPS. Allergen-pulsed DCs that were stimulated with low doses of LPS induce a strong Th2 response in vivo. Our objective was to investigate whether airway sensitization of mice by the application of DCs with a phenotype that is able to induce Th17 cells results in increased remodeling of the airways. We generated DCs from the bone marrow of mice and pulsed them with LPS-free ovalbumin. Subsequently, cells were activated with LPS with or without ATP for inflammasome activation. The activated cells were used to sensitize mice via the airways. Intranasal instillation of DCs that were activated with 0.1 ng/ml LPS induced a Th2 response with airway eosinophilia. High doses of LPS, particularly when given in combination with ATP, led to induction of a mixed Th2/Th17 response. Interestingly, we found a correlation between IL-17A production and the remodeling of the airways. Stimulation of mouse fibroblasts with purified IL-17A protein in vitro resulted in transforming growth factor-β1 secretion and collagen transcription. Interestingly, we found enhanced secretion of transforming growth factor-β1 by fibroblasts after costimulation with IL-17A and the profibrotic factor wingless-type MMTV integration site family, member 5A (Wnt5a). We showed that an allergen-specific Th17 response in the airway is accompanied by increased airway remodeling. Furthermore, we revealed that increased remodeling is not only based on neutrophilic inflammation, but also on the direct impact of IL-17A on airway structural cells.
Collapse
Affiliation(s)
- Marcus Peters
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany
| | | | - Tim Lenz-Habijan
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany
| | - Albrecht Bufe
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
35
|
Caramori G, Casolari P, Barczyk A, Durham AL, Di Stefano A, Adcock I. COPD immunopathology. Semin Immunopathol 2016; 38:497-515. [PMID: 27178410 PMCID: PMC4897000 DOI: 10.1007/s00281-016-0561-5] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/20/2016] [Indexed: 02/06/2023]
Abstract
The immunopathology of chronic obstructive pulmonary disease (COPD) is based on the innate and adaptive inflammatory immune responses to the chronic inhalation of cigarette smoking. In the last quarter of the century, the analysis of specimens obtained from the lower airways of COPD patients compared with those from a control group of age-matched smokers with normal lung function has provided novel insights on the potential pathogenetic role of the different cells of the innate and acquired immune responses and their pro/anti-inflammatory mediators and intracellular signalling pathways, contributing to a better knowledge of the immunopathology of COPD both during its stable phase and during its exacerbations. This also has provided a scientific rationale for new drugs discovery and targeting to the lower airways. This review summarises and discusses the immunopathology of COPD patients, of different severity, compared with control smokers with normal lung function.
Collapse
Affiliation(s)
- Gaetano Caramori
- Centro Interdipartimentale per lo Studio delle Malattie Infiammatorie delle Vie Aeree e Patologie Fumo-correlate (CEMICEF; formerly named Centro di Ricerca su Asma e BPCO), Sezione di Medicina Interna e Cardiorespiratoria, Università di Ferrara, Via Savonarola 9, 44121, Ferrara, Italy.
| | - Paolo Casolari
- Centro Interdipartimentale per lo Studio delle Malattie Infiammatorie delle Vie Aeree e Patologie Fumo-correlate (CEMICEF; formerly named Centro di Ricerca su Asma e BPCO), Sezione di Medicina Interna e Cardiorespiratoria, Università di Ferrara, Via Savonarola 9, 44121, Ferrara, Italy
| | - Adam Barczyk
- Katedra i Klinika Pneumonologii, Slaski Uniwersytet Medyczny w Katowicach, Katowice, Poland
| | - Andrew L Durham
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Antonino Di Stefano
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell'Apparato Cardio Respiratorio, Salvatore Maugeri Foundation, IRCCS, Veruno, NO, Italy
| | - Ian Adcock
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
36
|
Autocrine Acetylcholine, Induced by IL-17A via NFκB and ERK1/2 Pathway Activation, Promotes MUC5AC and IL-8 Synthesis in Bronchial Epithelial Cells. Mediators Inflamm 2016; 2016:9063842. [PMID: 27298519 PMCID: PMC4889862 DOI: 10.1155/2016/9063842] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/07/2016] [Accepted: 04/18/2016] [Indexed: 02/07/2023] Open
Abstract
IL-17A is overexpressed in the lung during acute neutrophilic inflammation. Acetylcholine (ACh) increases IL-8 and Muc5AC production in airway epithelial cells. We aimed to characterize the involvement of nonneuronal components of cholinergic system on IL-8 and Muc5AC production in bronchial epithelial cells stimulated with IL-17A. Bronchial epithelial cells were stimulated with recombinant human IL-17A (rhIL-17A) to evaluate the ChAT expression, the ACh binding and production, the IL-8 release, and the Muc5AC production. Furthermore, the effectiveness of PD098,059 (inhibitor of MAPKK activation), Bay11-7082 (inhibitor of IkBα phosphorylation), Hemicholinium-3 (HCh-3) (choline uptake blocker), and Tiotropium bromide (Spiriva®) (anticholinergic drug) was tested in our in vitro model. We showed that rhIL-17A increased the expression of ChAT, the levels of ACh binding and production, and the IL-8 and Muc5AC production in stimulated bronchial epithelial cells compared with untreated cells. The pretreatment of the cells with PD098,059 and Bay11-7082 decreased the ChAT expression and the ACh production/binding, while HCh-3 and Tiotropium decreased the IL-8 and Muc5AC synthesis in bronchial epithelial cells stimulated with rhIL-17A. IL-17A is involved in the IL-8 and Muc5AC production promoting, via NFκB and ERK1/2 pathway activation, the synthesis of ChAT, and the related activity of autocrine ACh in bronchial epithelial cells.
Collapse
|
37
|
Qian X, Chen H, Wu X, Hu L, Huang Q, Jin Y. Interleukin-17 acts as double-edged sword in anti-tumor immunity and tumorigenesis. Cytokine 2016; 89:34-44. [PMID: 26883678 DOI: 10.1016/j.cyto.2015.09.011] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 09/13/2015] [Accepted: 09/14/2015] [Indexed: 12/13/2022]
Abstract
Interleukin-17 (IL-17), a proinflammatory cytokine, mainly produced by Th17 cells, participates in both innate and adaptive immune responses and is involved in various diseases, including infectious diseases, autoimmune disorders and cancer. Emerging evidence indicates that IL-17 not only has an oncogenic role in tumorigenesis by regulating tumor angiogenesis and enhancing tumor immune evasion but also exerts anti-tumor functions by enhancing natural killer (NK) cells and cytotoxic T lymphocytes (CTLs) activation and through the recruitment of neutrophils, NK cells and CD4+ and CD8+ T cells to tumor tissue. In this review, we provide an overview on the basic biology of IL-17 and recent findings regarding its enigmatic double-edged features in tumorigenesis, with special attention to the roles of IL-17 produced by tumor cells interacting with other factors in the tumor microenvironment.
Collapse
Affiliation(s)
- Xin Qian
- Department of Respiratory Medicine, Taihe Hospital, Hubei University of Medicine, No. 32, South Renmin Road, Shiyan 44200, Hubei Province, China.
| | - Hankui Chen
- Rush University Cancer Center, Chicago, IL 60612, USA.
| | - Xiaofeng Wu
- Department of Respiratory Medicine, Taihe Hospital, Hubei University of Medicine, No. 32, South Renmin Road, Shiyan 44200, Hubei Province, China.
| | - Ling Hu
- Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430064, China.
| | - Qi Huang
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, PR China.
| | - Yang Jin
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, PR China.
| |
Collapse
|
38
|
Barnes PJ. Therapeutic approaches to asthma-chronic obstructive pulmonary disease overlap syndromes. J Allergy Clin Immunol 2015; 136:531-45. [PMID: 26343937 DOI: 10.1016/j.jaci.2015.05.052] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 05/21/2015] [Accepted: 05/22/2015] [Indexed: 12/14/2022]
Abstract
The recognition that there are some patients with features of asthma and chronic obstructive pulmonary disease (COPD) has highlighted the need to develop more specific treatments for these clinical phenotypes. Some patients with COPD have predominantly eosinophilic inflammation and might respond to high doses of inhaled corticosteroids and newly developed specific antieosinophil therapies, including blocking antibodies against IL-5, IL-13, IL-33, and thymic stromal lymphopoietin, as well as oral chemoattractant receptor-homologous molecule expressed on TH2 cells antagonists. Other patients have severe asthma or are asthmatic patients who smoke with features of COPD-induced inflammation and might benefit from treatments targeting neutrophils, including macrolides, CXCR2 antagonists, phosphodiesterase 4 inhibitors, p38 mitogen-activating protein kinase inhibitors, and antibodies against IL-1 and IL-17. Other patients appear to have largely fixed obstruction with little inflammation and might respond to long-acting bronchodilators, including long-acting muscarinic antagonists, to reduce hyperinflation. Highly selected patients with severe asthma might benefit from bronchial thermoplasty. Some patients with overlap syndromes can be conveniently treated with triple fixed-dose combination inhaler therapy with an inhaled corticosteroid, long-acting β2-agonist, and long-acting muscarinic antagonist, several of which are now in development. Corticosteroid resistance is a feature of asthma-COPD overlap syndrome, and understanding the various molecular mechanisms of this resistance has identified novel therapeutic targets and presented the prospect of therapies that can restore corticosteroid responsiveness.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College, London, United Kingdom.
| |
Collapse
|
39
|
Yamada Y, Jang JH, De Meester I, Baerts L, Vliegen G, Inci I, Yoshino I, Weder W, Jungraithmayr W. CD26 costimulatory blockade improves lung allograft rejection and is associated with enhanced interleukin-10 expression. J Heart Lung Transplant 2015; 35:508-17. [PMID: 26755203 DOI: 10.1016/j.healun.2015.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 10/15/2015] [Accepted: 11/19/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The ectoenzyme CD26/dipeptidyl peptidase 4 (DPP4) has costimulatory activity that contributes to T cell activation and proliferation. Here, we aimed to target this costimulatory activity for the attenuation of the alloreactive Th17-cell response during acute rejection after mouse lung transplantation. METHODS To test the CD26-costimulatory blockade in vitro, mixed lymphocyte reaction was performed between major histocompatibility complex class I and II fully mismatched cells (CD4(+) splenocytes, C57BL/6, responders, and antigen-presenting cells, BALB/c, stimulators) by adding the CD26 inhibitor vildagliptin (0-15 μg). Lung transplantation between BALB/c (donor) and C57BL/6 (recipient) mice was performed, including controls, CD26-inhibited (CD26-I, daily administration of vildagliptin [GLSynthesis, Worcester, MA], 10 mg/kg subcutaneous), and CD26 knockout (CD26KO) mice was performed. Analysis on Day 1 and 5 after transplant included immunohistochemistry, fluorescence-activated cell sorting, and enzyme-linked immunosorbent assay (ELISA) for immune cell detection and their key cytokines. RESULTS In vitro, there was a significant reduction of the Th17 cytokines interleukin (IL)-17 and IL-21. In vivo, CD26-I-treated and CD26KO mice showed significantly preserved macroscopic and histologic characteristics on Day 5 (p < 0.01), a higher partial pressure of arterial oxygen/fraction of inspired oxygen ratio (p ≤ 0.05), fewer infiltrating CD3(+) T cells (p < 0.01), but more interstitial macrophages on Day 1 (p < 0.01) compared with control. Fewer IL-17(+) cells were found in CD26-I allografts on Day 1 (p = 0.05). Higher levels of IL-10 in CD26-I and CD26KO allografts on day 5 were seen (p < 0.05). IL-10/CD206 double-staining (alternative macrophages) revealed more positive cells in CD26-I and CD26KO on Day 1 and 5 (p < 0.01). CONCLUSIONS CD26 costimulatory blockade promotes lung allograft acceptance via reduced T cell infiltration, less expression of IL-17, and increased expression of IL-10, likely to be derived from alternatively activated macrophages.
Collapse
Affiliation(s)
- Yoshito Yamada
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland; Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Jae-Hwi Jang
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Ingrid De Meester
- Department of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Lesley Baerts
- Department of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Gwendolyn Vliegen
- Department of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Ilhan Inci
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Ichiro Yoshino
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Walter Weder
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
40
|
Baba S, Kagoya R, Kondo K, Suzukawa M, Ohta K, Yamasoba T. T-cell phenotypes in chronic rhinosinusitis with nasal polyps in Japanese patients. Allergy Asthma Clin Immunol 2015; 11:33. [PMID: 26594227 PMCID: PMC4653844 DOI: 10.1186/s13223-015-0100-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 10/19/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyps is characterized by local inflammation and is categorized into two subtypes in Japan: eosinophilic chronic rhinosinusitis, and non-eosinophilic chronic rhinosinusitis. The objective of this study was to investigate the expression of key transcription factors for Treg and Th1/Th2/Th17 cells, in relation to the mRNA expression of representative cytokines in these two subtypes of chronic rhinosinusitis with nasal polyps. METHODS The expression of forkhead box P3 (FOXP3), T-box transcription factor (T-bet), GATA3, retinoid acid-related orphan receptor C (RORc), the suppressive cytokines TGF-β1 and IL-10, and Th1/Th2/Th17 cytokines (IFN-γ, IL-4, IL-5, IL-13, IL-17) were analyzed by means of RT-PCR in eosinophilic polyps. Eosinophilic polyps were defined as having an eosinophil count of more than 50 per microscopic field (×400 magnification) using five fields located in the subepithelial area of the polyps, while the non-eosinophilic polyps and controls did not fulfill this criteria. The numbers of T cells, CD4+ T cells, CD8+ T cells and Treg were histologically counted using sections that were immunostained for CD3, CD4, CD8, and FOXP3, respectively. RESULTS In eosinophilic polyps, we observed significantly fewer CD4+ T cells and CD8+ T cells, and lower GATA3, RORc and IL-10 mRNA expression, but a significantly higher IL-5, and IL-13 mRNA expression compared with controls, whereas FOXP3 and T-bet mRNA expression were not significantly different compared with controls. In non-eosinophilic polyps, FOXP3, IL-10, IL-17A, TGFβ1 and IFNγ mRNA expression was significantly higher compared with controls, whereas IL-4, 5 and 13 expression was not significantly different from controls. CONCLUSION We showed a reduction of GATA3 and RORc mRNA, low Treg-related cytokines and elevated Th2 cytokine levels in eosinophilic chronic rhinosinusitis, whereas we demonstrated the upregulation of Treg cells and increases of Th1 and Th17 cytokines in non-eosinophilic chronic rhinosinusitis in the Japanese population. The different mRNA expression profiles of Treg and Th1/Th2/Th17 signature transcription factors and cytokines between eosinophilic chronic rhinosinusitis and non-eosinophilic chronic rhinosinusitis suggests heterogeneity in the pathogenesis of chronic rhinosinusitis with nasal polyps.
Collapse
Affiliation(s)
- Shintaro Baba
- Department of Otolaryngology, Faculty of Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan ; Department of Otolaryngology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Ryoji Kagoya
- Department of Otolaryngology, Faculty of Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Kenji Kondo
- Department of Otolaryngology, Faculty of Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Maho Suzukawa
- Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Itabashi-ku, Tokyo, Japan ; National Hospital Organization Tokyo National Hospital, Kiyose, Tokyo Japan
| | - Ken Ohta
- Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Itabashi-ku, Tokyo, Japan ; National Hospital Organization Tokyo National Hospital, Kiyose, Tokyo Japan
| | - Tatsuya Yamasoba
- Department of Otolaryngology, Faculty of Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| |
Collapse
|
41
|
Vandermeulen E, Verleden SE, Ruttens D, Moelants E, Mortier A, Somers J, Bellon H, Piloni D, Dupont LJ, Van Raemdonck DE, Proost P, Schols D, Vos R, Verleden GM, Vanaudenaerde BM. BAL neutrophilia in azithromycin-treated lung transplant recipients: Clinical significance. Transpl Immunol 2015; 33:37-44. [DOI: 10.1016/j.trim.2015.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/02/2015] [Accepted: 07/03/2015] [Indexed: 11/25/2022]
|
42
|
Nitric oxide sustains IL-1β expression in human dendritic cells enhancing their capacity to induce IL-17-producing T-cells. PLoS One 2015; 10:e0120134. [PMID: 25853810 PMCID: PMC4390375 DOI: 10.1371/journal.pone.0120134] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 02/04/2015] [Indexed: 01/09/2023] Open
Abstract
The role played by lung dendritic cells (DCs) which are influenced by external antigens and by their redox state in controlling inflammation is unclear. We studied the role played by nitric oxide (NO) in DC maturation and function. Human DCs were stimulated with a long-acting NO donor, DPTA NONOate, prior to exposure to lipopolysaccharide (LPS). Dose-and time-dependent experiments were performed with DCs with the aim of measuring the release and gene expression of inflammatory cytokines capable of modifying T-cell differentiation, towardsTh1, Th2 and Th17 cells. NO changed the pattern of cytokine release by LPS-matured DCs, dependent on the concentration of NO, as well as on the timing of its addition to the cells during maturation. Addition of NO before LPS-induced maturation strongly inhibited the release of IL-12, while increasing the expression and release of IL-23, IL-1β and IL-6, which are all involved in Th17 polarization. Indeed, DCs treated with NO efficiently induced the release of IL-17 by T-cells through IL-1β. Our work highlights the important role that NO may play in sustaining inflammation during an infection through the preferential differentiation of the Th17 lineage.
Collapse
|
43
|
Chen ACH, Martin ML, Lourie R, Rogers GB, Burr LD, Hasnain SZ, Bowler SD, McGuckin MA, Serisier DJ. Adult non-cystic fibrosis bronchiectasis is characterised by airway luminal Th17 pathway activation. PLoS One 2015; 10:e0119325. [PMID: 25822228 PMCID: PMC4379018 DOI: 10.1371/journal.pone.0119325] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 01/28/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Non-cystic fibrosis (CF) bronchiectasis is characterised by chronic airway infection and neutrophilic inflammation, which we hypothesised would be associated with Th17 pathway activation. METHODS Th17 pathway cytokines were quantified in bronchoalveolar lavage fluid (BALF), and gene expression of IL-17A, IL-1β, IL-8 and IL-23 determined from endobronchial biopsies (EBx) in 41 stable bronchiectasis subjects and 20 healthy controls. Relationships between IL-17A levels and infection status, important clinical measures and subsequent Pseudomonas aeruginosa infection were determined. RESULTS BALF levels of all Th17 cytokines (median (IQR) pg/mL) were significantly higher in bronchiectasis than control subjects, including IL-17A (1.73 (1.19, 3.23) vs. 0.27 (0.24, 0.35), 95% CI 1.05 to 2.21, p<0.0001) and IL-23 (9.48 (4.79, 15.75) vs. 0.70 (0.43, 1.79), 95% CI 4.68 to 11.21, p<0.0001). However, BALF IL-17A levels were not associated with clinical measures or airway microbiology, nor predictive of subsequent P. aeruginosa infection. Furthermore, gene expression of IL-17A in bronchiectasis EBx did not differ from control. In contrast, gene expression (relative to medians of controls) in bronchiectasis EBx was significantly higher than control for IL1β (4.12 (1.24, 8.05) vs 1 (0.13, 2.95), 95% CI 0.05 to 4.07, p = 0.04) and IL-8 (3.75 (1.64, 11.27) vs 1 (0.54, 3.89), 95% CI 0.32 to 4.87, p = 0.02) and BALF IL-8 and IL-1α levels showed significant relationships with clinical measures and airway microbiology. P. aeruginosa infection was associated with increased levels of IL-8 while Haemophilus influenzae was associated with increased IL-1α. CONCLUSIONS AND CLINICAL RELEVANCE Established adult non-CF bronchiectasis is characterised by luminal Th17 pathway activation, however this pathway may be relatively less important than activation of non-antigen-specific innate neutrophilic immunity.
Collapse
Affiliation(s)
- Alice C.-H. Chen
- Immunity, Infection and Inflammation Program, Mater Research—University of Qld, Translational Research Institute, Woolloongabba, Qld, Australia
| | - Megan L. Martin
- Department of Respiratory Medicine, Mater Adult Hospital, South Brisbane, Qld, Australia
| | - Rohan Lourie
- Immunity, Infection and Inflammation Program, Mater Research—University of Qld, Translational Research Institute, Woolloongabba, Qld, Australia
- Department of Anatomical Pathology, Mater Health Services, South Brisbane, Qld, Australia
| | - Geraint B. Rogers
- Infection and Immunity Theme, South Australia Health and Medical Research Institute, North Terrace, Adelaide, Australia
- School of Medicine, Flinders University, Bedford Park, Adelaide, Australia
| | - Lucy D. Burr
- Immunity, Infection and Inflammation Program, Mater Research—University of Qld, Translational Research Institute, Woolloongabba, Qld, Australia
- Department of Respiratory Medicine, Mater Adult Hospital, South Brisbane, Qld, Australia
| | - Sumaira Z. Hasnain
- Immunity, Infection and Inflammation Program, Mater Research—University of Qld, Translational Research Institute, Woolloongabba, Qld, Australia
| | - Simon D. Bowler
- Department of Respiratory Medicine, Mater Adult Hospital, South Brisbane, Qld, Australia
| | - Michael A. McGuckin
- Immunity, Infection and Inflammation Program, Mater Research—University of Qld, Translational Research Institute, Woolloongabba, Qld, Australia
- School of Biomedical Science, The University of Queensland, Qld, Australia
| | - David J. Serisier
- Immunity, Infection and Inflammation Program, Mater Research—University of Qld, Translational Research Institute, Woolloongabba, Qld, Australia
- Department of Respiratory Medicine, Mater Adult Hospital, South Brisbane, Qld, Australia
- * E-mail:
| |
Collapse
|
44
|
Somers J, Ruttens D, Verleden SE, Vandermeulen E, Piloni D, Wauters E, Lambrechts D, Vos R, Verleden GM, Vanaudenaerde B, van Raemdonck DE. Interleukin-17 receptor polymorphism predisposes to primary graft dysfunction after lung transplantation. J Heart Lung Transplant 2015; 34:941-9. [PMID: 25935436 DOI: 10.1016/j.healun.2015.03.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 02/24/2015] [Accepted: 03/16/2015] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Primary graft dysfunction (PGD), with an incidence of 11% to 57%, is a major cause of morbidity and mortality within the first 30 days after lung transplantation (LTx). In this study, we postulate that recipient genetic variants in interleukin-17 and -23 receptor genes (IL-17R and IL-23R, respectively) may predispose LTx recipients to an increased risk for developing PGD. METHODS Seven genetic variants of IL-17R and IL-23R were successfully genotyped in 431 lung transplant recipients. Our primary end-point was PGD and secondary end-points were time to extubation, intensive care unit (ICU) stay, bronchoalveolar lavage neutrophilia and serum C-reactive protein. RESULTS The AA genotype of the rs882643 genetic variant of IL-17R was associated with higher PGD grades at 0 hour (adjusted p = 0.042), 12 hours (adjusted p = 0.013) and 48 hours (adjusted p = 0.0092) after LTx. The GG genotype of the rs2241049 genetic variant of IL-17R was associated with higher PGD grades at 48 hours (adjusted p = 0.0067) after LTx. For both genetic variants, no association was found with extubation time, ICU stay, post-operative BAL neutrophilia, serum CRP, chronic lung allograft dysfunction (CLAD) or graft loss. CONCLUSION Both genetic variants of IL-17R (rs882643 and rs2241049) were associated with PGD. This confirms a genetic predisposition toward PGD and suggests a role of IL-17 in driving neutrophilia in PGD.
Collapse
Affiliation(s)
- Jana Somers
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine
| | - David Ruttens
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine
| | - Stijn E Verleden
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine
| | - Elly Vandermeulen
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine
| | - Davide Piloni
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine
| | - Els Wauters
- Laboratory of Translational Genetics, Vesalius Research Center; Vesalius Reseach Centrum, VIB, Vlaams Instituut voor Biotechnologie, KU Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory of Translational Genetics, Vesalius Research Center; Vesalius Reseach Centrum, VIB, Vlaams Instituut voor Biotechnologie, KU Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine
| | - Geert M Verleden
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine
| | - Bart Vanaudenaerde
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine
| | - Dirk E van Raemdonck
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine.
| |
Collapse
|
45
|
Krenn K, Gmeiner M, Paulus P, Sela N, Torres L, Zins K, Dekan G, Aharinejad S. Effects of azithromycin and tanomastat on experimental bronchiolitis obliterans. J Thorac Cardiovasc Surg 2014; 149:1194-202. [PMID: 25595376 DOI: 10.1016/j.jtcvs.2014.11.088] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 11/16/2014] [Accepted: 11/29/2014] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Azithromycin has become a standard of care in therapy of bronchiolitis obliterans following lung transplantation. Matrix metalloprotease-9 broncho-alveolar lavage levels increase in airway neutrophilia and bronchiolitis obliterans. Interleukin-17 may play a role in lung allograft rejection, and interleukin-12 is downregulated in bronchiolitis obliterans. Whether these mechanisms can be targeted by azithromycin remains unclear. METHODS Bronchiolitis obliterans was induced by transplantation of Fischer F344 rat left lungs to Wistar Kyoto rats. Allografts with azithromycin therapy from day 1 to 28 or 56 and mono- or combination therapy with the broad-spectrum matrix metalloprotease inhibitor tanomastat from day 1 to 56 were compared to control allografts and isografts. Graft histology was assessed, and tissue cytokine expression studied using Western blotting and immunofluorescence. RESULTS The chronic airway rejection score in the azithromycin group did not change between 4 and 8 weeks after transplantation, whereas it significantly worsened in control allografts (P = .041). Azithromycin+tanomastat prevented complete allograft fibrosis, which occurred in 40% of control allografts. Azithromycin reduced interleukin-17 expression (P = .049) and the number of IL-17(+)/CD8(+) lymphocytes at 4 weeks, and active matrix metalloprotease-9 at 8 weeks (P = .017), and increased interleukin-12 expression (P = .025) at 8 weeks following transplantation versus control allografts. CONCLUSIONS The expression of interleukin-17 and matrix metalloprotease-9 in bronchiolitis obliterans may be attenuated by azithromycin, and the decrease in interleukin-12 expression was prevented by azithromycin. Combination of azithromycin with a matrix metalloprotease inhibitor is worth studying further because it prevented complete allograft fibrosis in this study.
Collapse
Affiliation(s)
- Katharina Krenn
- Department of Anesthesia and General Intensive Care, Medical University of Vienna, Vienna, Austria
| | - Matthias Gmeiner
- Department of Cardiovascular Research, Medical University of Vienna, Vienna, Austria
| | - Patrick Paulus
- Department of Cardiovascular Research, Medical University of Vienna, Vienna, Austria; Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe University Hospital, Frankfurt, Germany
| | - Nezir Sela
- Department of Cardiovascular Research, Medical University of Vienna, Vienna, Austria
| | - Linda Torres
- Department of Cardiovascular Research, Medical University of Vienna, Vienna, Austria
| | - Karin Zins
- Department of Cardiovascular Research, Medical University of Vienna, Vienna, Austria
| | - Gerhard Dekan
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Seyedhossein Aharinejad
- Department of Cardiovascular Research, Medical University of Vienna, Vienna, Austria; Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
46
|
Vos R, Verleden SE, Ruttens D, Vandermeulen E, Bellon H, Neyrinck A, Van Raemdonck DE, Yserbyt J, Dupont LJ, Verbeken EK, Moelants E, Mortier A, Proost P, Schols D, Cox B, Verleden GM, Vanaudenaerde BM. Azithromycin and the treatment of lymphocytic airway inflammation after lung transplantation. Am J Transplant 2014; 14:2736-48. [PMID: 25394537 DOI: 10.1111/ajt.12942] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/19/2014] [Accepted: 06/22/2014] [Indexed: 01/25/2023]
Abstract
Lymphocytic airway inflammation is a major risk factor for chronic lung allograft dysfunction, for which there is no established treatment. We investigated whether azithromycin could control lymphocytic airway inflammation and improve allograft function. Fifteen lung transplant recipients demonstrating acute allograft dysfunction due to isolated lymphocytic airway inflammation were prospectively treated with azithromycin for at least 6 months (NCT01109160). Spirometry (FVC, FEV1 , FEF25-75 , Tiffeneau index) and FeNO were assessed before and up to 12 months after initiation of azithromycin. Radiologic features, local inflammation assessed on airway biopsy (rejection score, IL-17(+) cells/mm(2) lamina propria) and broncho-alveolar lavage fluid (total and differential cell counts, chemokine and cytokine levels); as well as systemic C-reactive protein levels were compared between baseline and after 3 months of treatment. Airflow improved and FeNO decreased to baseline levels after 1 month of azithromycin and were sustained thereafter. After 3 months of treatment, radiologic abnormalities, submucosal cellular inflammation, lavage protein levels of IL-1β, IL-8/CXCL-8, IP-10/CXCL-10, RANTES/CCL5, MIP1-α/CCL3, MIP-1β/CCL4, Eotaxin, PDGF-BB, total cell count, neutrophils and eosinophils, as well as plasma C-reactive protein levels all significantly decreased compared to baseline (p < 0.05). Administration of azithromycin was associated with suppression of posttransplant lymphocytic airway inflammation and clinical improvement in lung allograft function.
Collapse
Affiliation(s)
- R Vos
- Department of Clinical and Experimental Medicine, Lab of Pneumology, Katholieke Universiteit Leuven and University Hospital Gasthuisberg, Leuven, Belgium; Lung Transplant Unit, Katholieke Universiteit Leuven and University Hospital Gasthuisberg, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ostadkarampour M, Eklund A, Moller D, Glader P, Olgart Höglund C, Lindén A, Grunewald J, Wahlström J. Higher levels of interleukin IL-17 and antigen-specific IL-17 responses in pulmonary sarcoidosis patients with Löfgren's syndrome. Clin Exp Immunol 2014; 178:342-52. [PMID: 24962673 DOI: 10.1111/cei.12403] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2014] [Indexed: 01/26/2023] Open
Abstract
Sarcoidosis is a granulomatous disorder of unknown aetiology. The presence of Mycobacterium tuberculosis catalase-peroxidase (mKatG) in sarcoidosis tissue has been reported. T helper type 1 (Th1) responses against mKatG have previously been observed. However, little is known about interleukin (IL)-17 and Th17 responses in sarcoidosis. Here, we investigated the levels of IL-17 and frequencies of IL-17-producing cells responding to mKatG in sarcoidosis patients with different prognosis. Peripheral blood and bronchoalveolar lavage (BAL) cells were obtained from sarcoidosis patients with or without Löfgren's syndrome (often associated with spontaneous recovery), and also stratified according to human leucocyte antigen (HLA) type. Cells producing IL-17 and interferon (IFN)-γ after stimulation with mKatG were enumerated by enzyme-linked immunospot (ELISPOT). The level of IL-17 in the BAL fluid of sarcoidosis patients and healthy controls was measured by quantitative immuno-polymerase chain reaction (qIPCR). We also performed flow cytometry and immunohistochemistry for further characterization of IL-17 expression. Patients with Löfgren's syndrome had a higher frequency of IL-17-producing cells responding to mKatG in BAL fluid compared to patients without Löfgren's syndrome (P < 0·05). The HLA-DR3(+) sarcoidosis patients with Löfgren's syndrome (known to have a particularly good prognosis) also had a clearly higher level of IL-17 in BAL fluid compared to healthy controls and sarcoidosis patients without Löfgren's syndrome (P < 0·01) and (P < 0·05), respectively. No such difference between patient groups was observed with regard to IFN-γ and not with regard to either cytokine in peripheral blood. These findings suggest that IL-17-producing cells may be a useful biomarker for the prognosis of sarcoidosis and play a role in the spontaneous recovery typical of patients with Löfgren's syndrome.
Collapse
Affiliation(s)
- M Ostadkarampour
- Respiratory Medicine Unit, Department of Medicine Solna and CMM, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Meyer KC, Raghu G, Verleden GM, Corris PA, Aurora P, Wilson KC, Brozek J, Glanville AR. An international ISHLT/ATS/ERS clinical practice guideline: diagnosis and management of bronchiolitis obliterans syndrome. Eur Respir J 2014; 44:1479-503. [PMID: 25359357 DOI: 10.1183/09031936.00107514] [Citation(s) in RCA: 395] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bronchiolitis obliterans syndrome (BOS) is a major complication of lung transplantation that is associated with poor survival. The International Society for Heart and Lung Transplantation, American Thoracic Society, and European Respiratory Society convened a committee of international experts to describe and/or provide recommendations for 1) the definition of BOS, 2) the risk factors for developing BOS, 3) the diagnosis of BOS, and 4) the management and prevention of BOS. A pragmatic evidence synthesis was performed to identify all unique citations related to BOS published from 1980 through to March, 2013. The expert committee discussed the available research evidence upon which the updated definition of BOS, identified risk factors and recommendations are based. The committee followed the GRADE (Grading of Recommendation, Assessment, Development and Evaluation) approach to develop specific clinical recommendations. The term BOS should be used to describe a delayed allograft dysfunction with persistent decline in forced expiratory volume in 1 s that is not caused by other known and potentially reversible causes of post-transplant loss of lung function. The committee formulated specific recommendations about the use of systemic corticosteroids, cyclosporine, tacrolimus, azithromycin and about re-transplantation in patients with suspected and confirmed BOS. The diagnosis of BOS requires the careful exclusion of other post-transplant complications that can cause delayed lung allograft dysfunction, and several risk factors have been identified that have a significant association with the onset of BOS. Currently available therapies have not been proven to result in significant benefit in the prevention or treatment of BOS. Adequately designed and executed randomised controlled trials that properly measure and report all patient-important outcomes are needed to identify optimal therapies for established BOS and effective strategies for its prevention.
Collapse
Affiliation(s)
- Keith C Meyer
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ganesh Raghu
- School of Medicine, University of Washington, Seattle, WA, USA
| | | | | | - Paul Aurora
- Great Ormond Street Hospital for Children, London, UK
| | | | - Jan Brozek
- McMaster University, Hamilton, ON, Canada
| | | | | | | |
Collapse
|
49
|
Vandermeulen E, Ruttens D, Verleden SE, Vos R, Van Raemdonck DE, Kastelijn EA, Wauters E, Lambrechts D, Nawrot TS, Cox B, Verleden GM, Vanaudenaerde BM. Genetic Variation in Caveolin-1 Affects Survival After Lung Transplantation. Transplantation 2014; 98:354-9. [DOI: 10.1097/tp.0000000000000058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Fouka E, Lamprianidou E, Arvanitidis K, Filidou E, Kolios G, Miltiades P, Paraskakis E, Antoniadis A, Kotsianidis I, Bouros D. Low-Dose Clarithromycin Therapy Modulates Th17 Response In Non-Cystic Fibrosis Bronchiectasis Patients. Lung 2014; 192:849-55. [DOI: 10.1007/s00408-014-9619-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 06/22/2014] [Indexed: 12/27/2022]
|