1
|
Goates M, Shrestha A, Thapa S, Bettini M, Barrios R, Shivanna B. Blocking IL-17a Signaling Decreases Lung Inflammation and Improves Alveolarization in Experimental Bronchopulmonary Dysplasia. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2023-2035. [PMID: 39117111 DOI: 10.1016/j.ajpath.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/13/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease of preterm infants that is associated with life-long morbidities. Inflammatory insults contribute to BPD pathogenesis. Although the proinflammatory cytokine, IL-17a, plays a role in various neonatal inflammatory disorders, its role in BPD pathogenesis is unclear. To test the hypothesis that blocking IL-17a signaling decreases lipopolysaccharide (LPS)-mediated experimental BPD in neonatal mice, wild-type mice were injected intraperitoneally with phosphate-buffered saline or LPS during the saccular lung developmental phase. Pulmonary IL-17a expression was determined by enzyme-linked immunosorbent assay and by flow cytometry. LPS-injected mice had higher pulmonary IL-17a protein levels and IL-17a+ and IL-22+ cells. γδ T cells, followed by non-T lymphoid cells, were the primary producers of IL-17a. Wild-type mice were then injected intraperitoneally with isotype antibody (Ab) or IL-17a Ab, while they were treated with phosphate-buffered saline or LPS, followed by quantification of lung inflammatory markers, alveolarization, vascularization, cell proliferation, and apoptosis. LPS-mediated alveolar simplification, apoptosis, and cell proliferation inhibition were significantly greater in mice treated with isotype Ab than in those treated with IL-17a Ab. Furthermore, STAT1 activation and IL-6 levels were significantly greater in LPS-exposed mice treated with isotype Ab than in those treated with IL-17a Ab. The study results indicate that blocking IL-17a signaling decreases LPS-mediated experimental BPD.
Collapse
Affiliation(s)
- Meagan Goates
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Amrit Shrestha
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Shyam Thapa
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Matthew Bettini
- Department of Microbiology and Immunology, University of Utah, Salt Lake City, Utah
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Binoy Shivanna
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
2
|
Ma R, Su H, Jiao K, Liu J. Association Between IL-17 and Chronic Obstructive Pulmonary Disease: A Systematic Review and Meta-Analysis. Int J Chron Obstruct Pulmon Dis 2023; 18:1681-1690. [PMID: 37551391 PMCID: PMC10404405 DOI: 10.2147/copd.s412626] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/24/2023] [Indexed: 08/09/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease characterized by neutrophils airway infiltration. It is currently known that Interleukin-17 (IL-17) is an important pro-inflammatory factor. It can promote the accumulation of neutrophils and participate in the chronic inflammatory process of COPD. However, the value of IL-17 levels in the diagnosis and assessment of COPD remains controversial. In view of this, we conducted a systematic review and meta-analysis to assess its relevance. Methods We searched databases such as PubMed, Web of Science, Cochrane Library and Embase to extract original research. Results A total of 10 studies with 2268 participants were included in this meta-analysis. The results showed that the level of serum IL-17 in patients with stable COPD was significantly higher than that in healthy controls (standard mean difference SMD, 1.59, 95% CI 0.84-2.34; p<0.001). Compared with the stable COPD group, the serum IL-17 level in acute exacerbation (AECOPD) was significantly higher (SMD, 1.78, 95% CI 1.22-2.33; p<0.001). The level of IL-17 in sputum of COPD patients was also higher than that of healthy controls (SMD, 2.03, 95% CI 0.74-3.31; p<0.001). Conclusion Our results showed that IL-17 levels were elevated in serum and sputum in COPD patients compared with healthy controls, and IL-17 levels increased with disease progression. IL-17 serves as a potential biomarker to indicate the persistence of neutrophilic inflammation and exacerbation of COPD.
Collapse
Affiliation(s)
- Ru Ma
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Gansu Provincial People’s Hospital, Lanzhou, Gansu Province, People’s Republic of China
| | - Hongling Su
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Gansu Provincial People’s Hospital, Lanzhou, Gansu Province, People’s Republic of China
| | - Keping Jiao
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Gansu Provincial People’s Hospital, Lanzhou, Gansu Province, People’s Republic of China
| | - Jian Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| |
Collapse
|
3
|
Chen X, Qiu L, Si X, Zhang X, Guo B, Liao Z, Yan X, Qi P. Exploring the Role of a Novel Interleukin-17 Homolog from Invertebrate Marine Mussel Mytilus coruscus in Innate Immune Response: Is Negative Regulation by Mc-Novel_miR_145 the Key? Int J Mol Sci 2023; 24:ijms24065928. [PMID: 36983002 PMCID: PMC10055819 DOI: 10.3390/ijms24065928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/13/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
Interleukin-17 (IL-17) represents a class of proinflammatory cytokines involved in chronic inflammatory and degenerative disorders. Prior to this study, it was predicted that an IL-17 homolog could be targeted by Mc-novel_miR_145 to participate in the immune response of Mytilus coruscus. This study employed a variety of molecular and cell biology research methods to explore the association between Mc-novel_miR_145 and IL-17 homolog and their immunomodulatory effects. The bioinformatics prediction confirmed the affiliation of the IL-17 homolog with the mussel IL-17 family, followed by quantitative real-time PCR assays (qPCR) to demonstrate that McIL-17-3 was highly expressed in immune-associated tissues and responded to bacterial challenges. Results from luciferase reporter assays confirmed the potential of McIL-17-3 to activate downstream NF-κb and its targeting by Mc-novel_miR_145 in HEK293 cells. The study also produced McIL-17-3 antiserum and found that Mc-novel_miR_145 negatively regulates McIL-17-3 via western blotting and qPCR assays. Furthermore, flow cytometry analysis indicated that Mc-novel_miR_145 negatively regulated McIL-17-3 to alleviate LPS-induced apoptosis. Collectively, the current results showed that McIL-17-3 played an important role in molluscan immune defense against bacterial attack. Furthermore, McIL-17-3 was negatively regulated by Mc-novel_miR_145 to participate in LPS-induced apoptosis. Our findings provide new insights into noncoding RNA regulation in invertebrate models.
Collapse
Affiliation(s)
- Xinglu Chen
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316004, China
| | - Longmei Qiu
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316004, China
| | - Xirui Si
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316004, China
| | - Xiaolin Zhang
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316004, China
| | - Baoying Guo
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316004, China
| | - Zhi Liao
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316004, China
| | - Xiaojun Yan
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316004, China
| | - Pengzhi Qi
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan 316004, China
| |
Collapse
|
4
|
Li D, Wang T, Ma Q, Zhou L, Le Y, Rao Y, Jin L, Pei Y, Cheng Y, Huang C, Gai X, Sun Y. IL-17A Promotes Epithelial ADAM9 Expression in Cigarette Smoke-Related COPD. Int J Chron Obstruct Pulmon Dis 2022; 17:2589-2602. [PMID: 36267325 PMCID: PMC9578481 DOI: 10.2147/copd.s375006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
Background It has been reported that a disintegrin and metalloproteinase 9 (ADAM9) is involved in the pathogenesis of cigarette smoke (CS)-associated chronic obstructive pulmonary disease (COPD). But how CS exposure leads to upregulation of ADAM9 remains unknown. Methods Patients who underwent lobectomy for a solitary pulmonary nodule were enrolled and divided into three groups: non-smokers with normal lung function, smokers without COPD and smoker patients with COPD. Immunoreactivity of interleukin (IL)-17A and ADAM9 in small airways and alveolar walls was measured by immunohistochemistry. Wild-type and Il17a−/− C57BL/6 mice were exposed to CS for six months, and ADAM9 expression in the airway epithelia was measured by immunoreactivity. In addition, the protein and mRNA expression levels of IL-17A and ADAM9 were assessed in CS extract (CSE) and/or IL-17A-treated human bronchial epithelial (HBE) cells. Results The immunoreactivity of ADAM9 was increased in the airway epithelia and alveolar walls of patients with COPD compared to that of the controls. The expression of IL-17A was also upregulated in airway epithelial cells of patients with COPD and correlated positively with the level of ADAM9. The results from the animal model showed that Il17a−/− mice were protected from emphysema induced by CS exposure, together with a reduced level of ADAM9 expression in the airway epithelia, suggesting a possible link between ADAM9 and IL-17A. Consistently, our in vitro cell model showed that CSE stimulated the expression of ADAM9 and IL-17A in HBE cells in a dose- and time-dependent manner. Recombinant IL-17A induced ADAM9 upregulation in HBE cells and had a synergistic effect with CSE, whereas blocking IL-17A inhibited CSE-induced ADAM9 expression. Further analysis revealed that IL-17A induced c-Jun N-terminal kinase (JNK) phosphorylation, thereby increasing ADAM9 expression. Conclusion Our results revealed a novel role of IL-17A in CS-related COPD, where IL-17A contributes to ADAM9 expression by activating JNK signaling.
Collapse
Affiliation(s)
- Danyang Li
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Tong Wang
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Qianli Ma
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Lu Zhou
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yanqing Le
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yafei Rao
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Liang Jin
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yuqiang Pei
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yaning Cheng
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, People’s Republic of China
| | - Chen Huang
- Center of Basic Medical Research, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China,Correspondence: Xiaoyan Gai; Yongchang Sun, Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, People’s Republic of China, Email ;
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| |
Collapse
|
5
|
Ritzmann F, Lunding LP, Bals R, Wegmann M, Beisswenger C. IL-17 Cytokines and Chronic Lung Diseases. Cells 2022; 11:2132. [PMID: 35883573 PMCID: PMC9318387 DOI: 10.3390/cells11142132] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/27/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
IL-17 cytokines are expressed by numerous cells (e.g., gamma delta (γδ) T, innate lymphoid (ILC), Th17, epithelial cells). They contribute to the elimination of bacteria through the induction of cytokines and chemokines which mediate the recruitment of inflammatory cells to the site of infection. However, IL-17-driven inflammation also likely promotes the progression of chronic lung diseases, such as chronic obstructive pulmonary disease (COPD), lung cancer, cystic fibrosis, and asthma. In this review, we highlight the role of IL-17 cytokines in chronic lung diseases.
Collapse
Affiliation(s)
- Felix Ritzmann
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
- Helmholtz Institute for Pharmaceutical Research, 66123 Saarbrücken, Germany
| | - Lars Peter Lunding
- Division of Lung Immunology, Priority Area Asthma and Allergy, Research Center Borstel—Leibniz Lung Center, 23845 Borstel, Germany; (L.P.L.); (M.W.)
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Robert Bals
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
- Helmholtz Institute for Pharmaceutical Research, 66123 Saarbrücken, Germany
| | - Michael Wegmann
- Division of Lung Immunology, Priority Area Asthma and Allergy, Research Center Borstel—Leibniz Lung Center, 23845 Borstel, Germany; (L.P.L.); (M.W.)
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
| |
Collapse
|
6
|
Chen XY, Chen YY, Lin W, Chen CH, Wen YC, Hsiao TC, Chou HC, Chung KF, Chuang HC. Therapeutic Potential of Human Umbilical Cord-Derived Mesenchymal Stem Cells in Recovering From Murine Pulmonary Emphysema Under Cigarette Smoke Exposure. Front Med (Lausanne) 2021; 8:713824. [PMID: 34646841 PMCID: PMC8502916 DOI: 10.3389/fmed.2021.713824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/31/2021] [Indexed: 01/08/2023] Open
Abstract
Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) were shown to have potential for immunoregulation and tissue repair. The objective of this study was to investigate the effects of hUC-MSCs on emphysema in chronic obstructive pulmonary disease (COPD). The C57BL/6JNarl mice were exposed to cigarette smoke (CS) for 4 months followed by administration of hUC-MSCs at 3 × 106 (low dose), 1 × 107 (medium dose), and 3 × 107 cells/kg body weight (high dose). The hUC-MSCs caused significant decreases in emphysema severity by measuring the mean linear intercept (MLI) and destructive index (DI). A decrease in neutrophils (%) and an increase in lymphocytes (%) in bronchoalveolar lavage fluid (BALF) were observed in emphysematous mice after hUC-MSC treatment. Lung levels of interleukin (IL)-1β, C-X-C motif chemokine ligand 1 (CXCL1)/keratinocyte chemoattractant (KC), and matrix metalloproteinase (MMP)-12 significantly decreased after hUC-MSC administration. Significant reductions in tumor necrosis factor (TNF)-α, IL-1β, and IL-17A in serum occurred after hUC-MSC administration. Notably, the cell viability of lung fibroblasts improved with hUC-MSCs after being treated with CS extract (CSE). Furthermore, the hUC-MSCs-conditioned medium (hUC-MSCs-CM) restored the contractile force, and increased messenger RNA expressions of elastin and fibronectin by lung fibroblasts. In conclusion, hUC-MSCs reduced inflammatory responses and emphysema severity in CS-induced emphysematous mice.
Collapse
Affiliation(s)
- Xiao-Yue Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Ying Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Willie Lin
- Meridigen Biotech Co., Ltd., Taipei, Taiwan
| | | | | | - Ta-Chih Hsiao
- Graduate Institute of Environmental Engineering, National Taiwan University, Taipei, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
7
|
Wu M, Lai T, Jing D, Yang S, Wu Y, Li Z, Wu Y, Zhao Y, Zhou L, Chen H, Shen J, Li W, Ying S, Chen Z, Wu X, Shen H. Epithelium-derived IL17A Promotes Cigarette Smoke-induced Inflammation and Mucus Hyperproduction. Am J Respir Cell Mol Biol 2021; 65:581-592. [PMID: 34186014 DOI: 10.1165/rcmb.2020-0424oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Airway epithelium is a central modulator of innate and adaptive immunity in the lung. Interleukin (IL)17A expression was found to be increased in airway epithelium; however, the role of epithelial-derived IL17A in chronic obstructive pulmonary disease (COPD) remains unclear. In this study, we aim to determine whether epithelial-derived IL17A regulates inflammation and mucus hyperproduction in COPD using a cultured human bronchial epithelial (HBE) cell line in vitro and airway epithelium IL17A-specific knockout mouse in vivo. Increased IL17A expression was observed in mouse airway epithelium upon cigarette smoke (CS) exposure or in a COPD mouse model that was induced by CS and elastin. CS extract (CSE) also triggered IL17A expression in HBE cells. Blocking IL17A or IL17RA effectively attenuated CSE-induced MUC5AC and the inflammatory cytokines IL6, tumor necrosis factor (TNF)-α, and IL1β in HBE cells, suggesting that IL17A mediates CSE-induced inflammation and mucin production in an autocrine manner. CSE activated p-JUN and p-JNK, which were also reduced by IL17RA-siRNA, and JUN-siRNA attenuated CSE-induced IL6 and MUC5AC. In vivo, selective knockout of IL17A in airway epithelium markedly reduced the neutrophilic infiltration in Bronchoalveolar Lavage Fluid (BALF), peribronchial inflammation, pro-inflammatory mediators (CXCL1 and CXCL2), and mucus production in a COPD mouse model. We showed a novel function of airway epithelium-derived IL17A, which can act locally in an autocrine manner to amplify inflammation and increase mucus production in COPD pathogenesis.
Collapse
Affiliation(s)
- Mindan Wu
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Tianwen Lai
- Zhejiang University School of Medicine, 26441, Hangzhou, China
| | - Du Jing
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Shiyi Yang
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Yanping Wu
- Zhejiang University School of Medicine, 26441, Respiratory and Critical Care Medicine, Hangzhou, China
| | - Zhouyang Li
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Yinfang Wu
- Zhejiang University School of Medicine, 26441, Respiratory and Critical Care Medicine, Hangzhou, China
| | - Yun Zhao
- Zhejiang University School of Medicine, 26441, Respiratory and Critical Care Medicine, Hangzhou, China
| | - Lingren Zhou
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Haipin Chen
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, China
| | - Jiaxin Shen
- Zhejiang University School of Medicine, 26441, Respiratory and Critical Care Medicine, Hangzhou, China
| | - Wen Li
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Respiratory and Critical Care Midicine, Hangzhou, China
| | - Songmin Ying
- Zhejiang University School of Medicine, 26441, Respiratory and Critical Care Medicine, Hangzhou, China
| | - Zhihua Chen
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Respiratory and Critical Care Midicine, Hangzhou, China
| | - Xiaohong Wu
- Zhejiang University School of Medicine Sir Run Run Shaw Hospital, 56660, Hangzhou, China
| | - Huahao Shen
- Zhejiang University School of Medicine, 26441, Respiratory Medicine, Hangzhou, China;
| |
Collapse
|
8
|
Dual interleukin-17A/F deficiency protects against acute and chronic response to cigarette smoke exposure in mice. Sci Rep 2021; 11:11508. [PMID: 34075087 PMCID: PMC8169846 DOI: 10.1038/s41598-021-90853-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/18/2021] [Indexed: 12/19/2022] Open
Abstract
IL-17A and IL-17F are both involved in the pathogenesis of neutrophilic inflammation observed in COPD and severe asthma. To explore this, mice deficient in both Il17a and Il17f and wild type (WT) mice were exposed to cigarette smoke or environmental air for 5 to 28 days and changes in inflammatory cells in bronchoalveolar lavage (BAL) fluid were determined. We also measured the mRNA expression of keratinocyte derived chemokine (Kc), macrophage inflammatory protein-2 (Mip2), granulocyte–macrophage colony stimulating factor (Gmcsf) and matrix metalloproteinase-9 (Mmp9 ) in lung tissue after 8 days, and lung morphometric changes after 24 weeks of exposure to cigarette smoke compared to air-exposed control animals. Macrophage counts in BAL fluid initially peaked at day 8 and again on day 28, while neutrophil counts peaked between day 8 and 12 in WT mice. Mice dual deficient with Il17a and 1l17f showed similar kinetics with macrophages and neutrophils, but cell numbers at day 8 and mRNA expression of Kc, Gmcsf and Mmp9 were significantly reduced. Furthermore, airspaces in WT mice became larger after cigarette smoke exposure for 24 weeks, whereas this was not seen dual Il17a and 1l17f deficient mice. Combined Il17a and Il17f deficiency resulted in significant attenuation of neutrophilic inflammatory response and protection against structural lung changes after long term cigarette smoke exposure compared with WT mice. Dual IL-17A/F signalling plays an important role in pro-inflammatory responses associated with histological changes induced by cigarette smoke exposure.
Collapse
|
9
|
Ritzmann F, Beisswenger C. Preclinical studies and the function of IL-17 cytokines in COPD. Ann Anat 2021; 237:151729. [PMID: 33798693 DOI: 10.1016/j.aanat.2021.151729] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is among the leading causes of death worldwide and imposes a high economic burden to the health systems. COPD is characterized by chronic inflammation of the lung leading to airflow limitation, alveolar tissue destruction, and emphysema. Therefore, anti-inflammatory therapies for the treatment of COPD are of interest. In this review, we focus on the function of the IL-17 cytokines IL-17A and IL-17C, both known to mediate the recruitment of inflammatory cells, in the pathogenesis of COPD. We highlight that the expression of IL-17A and IL-17C is induced by pathogens frequently found in lungs of COPD patients and that targeting IL-17-signaling is an interesting option for the treatment of acute exacerbation of COPD.
Collapse
Affiliation(s)
- Felix Ritzmann
- Department of Internal Medicine V - Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V - Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany.
| |
Collapse
|
10
|
Zhang XF, Xiang SY, Lu J, Li Y, Zhao SJ, Jiang CW, Liu XG, Liu ZB, Zhang J. Electroacupuncture inhibits IL-17/IL-17R and post-receptor MAPK signaling pathways in a rat model of chronic obstructive pulmonary disease. Acupunct Med 2021; 39:663-672. [PMID: 33715422 DOI: 10.1177/0964528421996720] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Interleukin (IL)-17, as a T-helper 17 cell (Th17) cytokine, plays a key role in chronic obstructive pulmonary disease (COPD) pathophysiology including chronic inflammation and airway obstruction, which lead to decreased pulmonary function. The aim of this study was to investigate the effect of acupuncture on IL-17, its receptor (IL-17R) and the mitogen-activated protein kinase (MAPK) signaling pathway, in a rat model of COPD. METHODS The COPD model was induced in Sprague Dawley rats by exposure to cigarette smoke for 12 weeks. The model rats were treated with electroacupuncture (EA) at BL13 and ST36. The lung function and histology of the rats were observed. IL-17, tumor necrosis factor (TNF)-α, and IL-10 were detected by enzyme-linked immunosorbent assay (ELISA) in bronchoalveolar lavage fluid (BALF) and in plasma. The leukocytes and macrophages in the BALF were counted. The expression levels of IL-17R were assayed in lung tissue by real-time polymerase chain reaction (PCR), western blotting, and immunohistochemistry. MAPK signaling pathway molecules including c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK)1/2 and p38, and their phosphorylated forms, were observed in the lung by western blotting. RESULTS Compared with the control group rats, lung function decreased and there was a severe inflammatory infiltration of the pulmonary parenchyma in the COPD rats. EA effectively improved lung function and alleviated the inflammatory infiltration in the lungs of COPD rats. EA also reversed the elevated total leukocyte and macrophage counts, the high levels of IL-17 and TNF-α, and the low IL-10 content in COPD rats. Meanwhile, EA downregulated the increased mRNA and protein expression of IL-17R, and significantly inhibited the elevated levels of phosphorylated JNK, ERK1/2, and p38 in the lungs of COPD rats. CONCLUSION Our results suggest that the protective effects of acupuncture therapy on the lungs of COPD rats are likely related to inhibition of IL-17/IL-17R and the post-receptor MAPK signaling pathways.
Collapse
Affiliation(s)
- Xin-Fang Zhang
- Department of Physiology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Shui-Ying Xiang
- Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Lu
- Department of Rehabilitation & Health Care, Anhui College of Traditional Chinese Medicine, Wuhu, China
| | - Yin Li
- Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei, China
| | - Shu-Jun Zhao
- Department of Physiology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chuan-Wei Jiang
- Department of Physiology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xiang-Guo Liu
- Department of Histology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Zi-Bing Liu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China.,Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei, China
| | - Jie Zhang
- Department of Immunology, Medical College of Nantong University, Nantong, China
| |
Collapse
|
11
|
Nascimento M, Huot-Marchand S, Gombault A, Panek C, Bourinet M, Fanny M, Savigny F, Schneider P, Le Bert M, Ryffel B, Riteau N, Quesniaux VFJ, Couillin I. B-Cell Activating Factor Secreted by Neutrophils Is a Critical Player in Lung Inflammation to Cigarette Smoke Exposure. Front Immunol 2020; 11:1622. [PMID: 32849550 PMCID: PMC7405926 DOI: 10.3389/fimmu.2020.01622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022] Open
Abstract
Cigarette smoke (CS) is the major cause of chronic lung injuries, such as chronic obstructive pulmonary disease (COPD). In patients with severe COPD, tertiary lymphoid follicles containing B lymphocytes and B cell-activating factor (BAFF) overexpression are associated with disease severity. In addition, BAFF promotes adaptive immunity in smokers and mice chronically exposed to CS. However, the role of BAFF in the early phase of innate immunity has never been investigated. We acutely exposed C57BL/6J mice to CS and show early BAFF expression in the bronchoalveolar space and lung tissue that correlates to airway neutrophil and macrophage influx. Immunostaining analysis revealed that neutrophils are the major source of BAFF. We confirmed in vitro that neutrophils secrete BAFF in response to cigarette smoke extract (CSE) stimulation. Antibody-mediated neutrophil depletion significantly dampens lung inflammation to CS exposure but only partially decreases BAFF expression in lung tissue and bronchoalveolar space suggesting additional sources of BAFF. Importantly, BAFF deficient mice displayed decreased airway neutrophil recruiting chemokines and neutrophil influx while the addition of exogenous BAFF significantly enhanced this CS-induced neutrophilic inflammation. This demonstrates that BAFF is a key proinflammatory cytokine and that innate immune cells in particular neutrophils, are an unconsidered source of BAFF in early stages of CS-induced innate immunity.
Collapse
Affiliation(s)
| | | | | | - Corinne Panek
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Manon Bourinet
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Manoussa Fanny
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | | | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Épalinges, Switzerland
| | - Marc Le Bert
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Bernhard Ryffel
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | - Nicolas Riteau
- University of Orleans and CNRS, INEM-UMR7355, Orléans, France
| | | | | |
Collapse
|
12
|
Ayhan E, Öztürk M, An İ, Abdelmaksoud A, Araç E. Potential role of anti-interleukin-17 in COVID-19 treatment. Dermatol Ther 2020; 33:e13715. [PMID: 32475041 DOI: 10.1111/dth.13715] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 12/25/2022]
Affiliation(s)
- Erhan Ayhan
- Department of Dermatology, University of Health Sciences Gazi Yaşargil Trainning and Research Hospital, Diyarbakır, Turkey
| | - Murat Öztürk
- Department of Dermatology, Van Trainning and Research Hospital, Van, Turkey
| | - İsa An
- Department of Dermatology, Şanlıurfa Trainning and Research Hospital, Şanlıurfa, Turkey
| | - Ayman Abdelmaksoud
- Mansoura Dermatology, Venerology and Leprology Hospital, Mansoura, Egypt
| | - Eşref Araç
- Department of Internal Medicine, University of Health Sciences Gazi Yaşargil Trainning and Research Hospital, Diyarbakır, Turkey
| |
Collapse
|
13
|
Yu Y, Zhao L, Xie Y, Xu Y, Jiao W, Wu J, Deng X, Fang G, Xue Q, Zheng Y, Gao Z. Th1/Th17 Cytokine Profiles are Associated with Disease Severity and Exacerbation Frequency in COPD Patients. Int J Chron Obstruct Pulmon Dis 2020; 15:1287-1299. [PMID: 32606639 PMCID: PMC7294048 DOI: 10.2147/copd.s252097] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/11/2020] [Indexed: 12/30/2022] Open
Abstract
Background T helper (Th) cell cytokine imbalances have been associated with the pathophysiology of chronic obstructive pulmonary disease (COPD), including the Th1/Th2 and Th17/T regulatory cells (Treg) paradigms. Clarifying cytokine profiles during COPD acute exacerbation (AE) and their relationships with clinical manifestations would help in understanding the pathogenesis of disease and improve clinical management. Materials and Methods Eighty seven patients admitted to the hospital with AEs of COPD were included in this study, and follow-up was conducted after discharge (every 30 days, for a total of 120 days). Sputum samples of patients at different time points (including at admission, discharge, and follow-up) were collected, and sputum cytokine profiling (12 cytokines in total) was performed using a Luminex assay. Results According to the cytokine profiles at admission, patients were divided into three clusters by a k-means clustering algorithm, namely, Th1high Th17high (n=26), Th1lowTh17low (n=56), and Th1high Th17low (n=5), which revealed distinct clinical characteristics. Patients with Th1high Th17low profile had a significantly longer length of non-invasive ventilation time and length of hospital stay than patients with Th1high Th17high profile (7 vs 0 days, 22 vs 11 days, respectively, p < 0.05), and had the highest AE frequency. Sputum levels of Th17 cytokines (IL-17A, IL-22, and IL-23) during AE were negatively correlated with AE frequency in the last 12 months (r = −0.258, −0.289 and −0.216, respectively, p < 0.05). Moreover, decreased sputum IL-17A levels were independently associated with increased AE frequency, with an OR (95% CI) of 0.975 (0.958–0.993) and p = 0.006. Conclusion Th1/Th17 imbalance during AE is associated with the severity of COPD. Decreased Th17 cytokine expression is correlated with increased AE frequency. The Th1/Th17 balance may be a specific target for the therapeutic manipulation of COPD.
Collapse
Affiliation(s)
- Yan Yu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, People's Republic of China
| | - Lili Zhao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, People's Republic of China
| | - Yu Xie
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, People's Republic of China
| | - Yu Xu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, People's Republic of China
| | - Weike Jiao
- Department of Pulmonary and Critical Care Medicine, Ningde Municipal Hospital Affiliated to Fujian Medical University, Ningde, Fujian 352100, People's Republic of China
| | - Jianhui Wu
- Department of Pulmonary and Critical Care Medicine, Ningde Municipal Hospital Affiliated to Fujian Medical University, Ningde, Fujian 352100, People's Republic of China
| | - Xinyu Deng
- Department of Pulmonary and Critical Care Medicine, Ningde Municipal Hospital Affiliated to Fujian Medical University, Ningde, Fujian 352100, People's Republic of China
| | - Guiju Fang
- Department of Pulmonary and Critical Care Medicine, Ningde Municipal Hospital Affiliated to Fujian Medical University, Ningde, Fujian 352100, People's Republic of China
| | - Qing Xue
- Department of Pulmonary and Critical Care Medicine, Ningde Municipal Hospital Affiliated to Fujian Medical University, Ningde, Fujian 352100, People's Republic of China
| | - Yali Zheng
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, People's Republic of China.,Department of Respiratory and Critical Care Medicine, Xiang'An Hospital of Xiamen University, Xiamen, Fujian 361100, People's Republic of China
| | - Zhancheng Gao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing 100044, People's Republic of China.,Department of Respiratory and Critical Care Medicine, Xiang'An Hospital of Xiamen University, Xiamen, Fujian 361100, People's Republic of China
| |
Collapse
|
14
|
Xiong J, Tian J, Zhou L, Le Y, Sun Y. Interleukin-17A Deficiency Attenuated Emphysema and Bone Loss in Mice Exposed to Cigarette Smoke. Int J Chron Obstruct Pulmon Dis 2020; 15:301-310. [PMID: 32103929 PMCID: PMC7020917 DOI: 10.2147/copd.s235384] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/22/2020] [Indexed: 12/20/2022] Open
Abstract
Background and Purpose Chronic obstructive pulmonary disease (COPD) is a common chronic inflammatory disease, which is associated with various comorbidities including osteoporosis. Interleukin(IL)-17 has been reported to play important roles in the pathogenesis of COPD and also associated with bone destruction in inflammatory diseases. However, the role of IL-17A in COPD-related osteoporosis is yet unknown. The purpose of our study was to investigate the potential contribution of IL-17A in COPD-related bone loss. Materials and Methods We examined the bone mass and bone microarchitecture in wild-type and IL-17A-/- mice exposed to long-term cigarette smoke (CS). Osteoclast activities and the expression of receptor activator of nuclear factor-κB ligand (RANKL) in bone tissues were assessed, and the blood levels of inflammatory cytokines were measured. Results Less bone loss as well as attenuated emphysema were shown in IL-17A-/- mice compared with wild-type mice. CS-exposed IL-17A-/- mice had decreased TRAP+ osteoclast numbers and lower RANKL expression compared with CS-exposed wild-type mice. Inflammatory cytokines including IL-6 and IL-1β in circulation were decreased in IL-17A-/- mice exposed to CS compared with wild-type mice. Conclusion This study indicates that IL-17A is involved in CS-induced bone loss and may be a common link between COPD and osteoporosis.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Jieyu Tian
- Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, Beijing 100045, People's Republic of China
| | - Lu Zhou
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Yanqing Le
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| |
Collapse
|
15
|
Lee JH, Hailey KL, Vitorino SA, Jennings PA, Bigby TD, Breen EC. Cigarette Smoke Triggers IL-33-associated Inflammation in a Model of Late-Stage Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2019; 61:567-574. [PMID: 30973786 PMCID: PMC6827064 DOI: 10.1165/rcmb.2018-0402oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/10/2019] [Indexed: 01/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a worldwide threat. Cigarette smoke (CS) exposure causes cardiopulmonary disease and COPD and increases the risk for pulmonary tumors. In addition to poor lung function, patients with COPD are susceptible to bouts of dangerous inflammation triggered by pollutants or infection. These severe inflammatory episodes can lead to additional exacerbations, hospitalization, further deterioration of lung function, and reduced survival. Suitable models of the inflammatory conditions associated with CS, which potentiate the downward spiral in patients with COPD, are lacking, and the underlying mechanisms that trigger exacerbations are not well understood. Although initial CS exposure activates a protective role for vascular endothelial growth factor (VEGF) functions in barrier integrity, chronic exposure depletes the pulmonary VEGF guard function in severe COPD. Thus, we hypothesized that mice with compromised VEGF production and challenged with CS would trigger human-like severe inflammatory progression of COPD. In this model, we discovered that CS exposure promotes an amplified IL-33 cytokine response and severe disease progression. Our VEGF-knockout model combined with CS recapitulates severe COPD with an influx of IL-33-expressing macrophages and neutrophils. Normally, IL-33 is quickly inactivated by a post-translational disulfide bond formation. Our results reveal that BAL fluid from the CS-exposed, VEGF-deficient cohort promotes a significantly prolonged lifetime of active proinflammatory IL-33. Taken together, our data demonstrate that with the loss of a VEGF-mediated protective barrier, the CS response switches from a localized danger to an uncontrolled long-term and long-range, amplified, IL-33-mediated inflammatory response that ultimately destroys lung function.
Collapse
Affiliation(s)
| | - Kendra L. Hailey
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California; and
| | | | - Patricia A. Jennings
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California; and
| | - Timothy D. Bigby
- Department of Medicine and
- Pulmonary and Critical Care, Veterans Affairs San Diego, La Jolla, California
| | | |
Collapse
|
16
|
Zuo L, Prather ER, Stetskiv M, Garrison DE, Meade JR, Peace TI, Zhou T. Inflammaging and Oxidative Stress in Human Diseases: From Molecular Mechanisms to Novel Treatments. Int J Mol Sci 2019; 20:E4472. [PMID: 31510091 PMCID: PMC6769561 DOI: 10.3390/ijms20184472] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
It has been proposed that a chronic state of inflammation correlated with aging known as inflammaging, is implicated in multiple disease states commonly observed in the elderly population. Inflammaging is associated with over-abundance of reactive oxygen species in the cell, which can lead to oxidation and damage of cellular components, increased inflammation, and activation of cell death pathways. This review focuses on inflammaging and its contribution to various age-related diseases such as cardiovascular disease, cancer, neurodegenerative diseases, chronic obstructive pulmonary disease, diabetes, and rheumatoid arthritis. Recently published mechanistic details of the roles of reactive oxygen species in inflammaging and various diseases will also be discussed. Advancements in potential treatments to ameliorate inflammaging, oxidative stress, and consequently, reduce the morbidity of multiple disease states will be explored.
Collapse
Affiliation(s)
- Li Zuo
- College of Arts and Sciences, University of Maine Presque Isle Campus, Presque Isle, ME 04769, USA.
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| | - Evan R Prather
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Mykola Stetskiv
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Davis E Garrison
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - James R Meade
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Timotheus I Peace
- College of Arts and Sciences, University of Maine Presque Isle Campus, Presque Isle, ME 04769, USA
| | - Tingyang Zhou
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
17
|
Rhinovirus-induces progression of lung disease in a mouse model of COPD via IL-33/ST2 signaling axis. Clin Sci (Lond) 2019; 133:983-996. [PMID: 30952808 DOI: 10.1042/cs20181088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/25/2019] [Accepted: 04/05/2019] [Indexed: 12/28/2022]
Abstract
Rhinovirus (RV), which is associated with acute exacerbations, also causes persistent lung inflammation in patients with chronic obstructive pulmonary disease (COPD), but the underlying mechanisms are not well-known. Recently, we demonstrated that RV causes persistent lung inflammation with accumulation of a subset of macrophages (CD11b+/CD11c+), and CD8+ T cells, and progression of emphysema. In the present study, we examined the mechanisms underlying the RV-induced persistent inflammation and progression of emphysema in mice with COPD phenotype. Our results demonstrate that at 14 days post-RV infection, in addition to sustained increase in CCL3, CXCL-10 and IFN-γ expression as previously observed, levels of interleukin-33 (IL-33), a ligand for ST2 receptor, and matrix metalloproteinase (MMP)12 are also elevated in mice with COPD phenotype, but not in normal mice. Further, MMP12 was primarily expressed in CD11b+/CD11c+ macrophages. Neutralization of ST2, reduced the expression of CXCL-10 and IFN-γ and attenuated accumulation of CD11b+/CD11c+ macrophages, neutrophils and CD8+ T cells in COPD mice. Neutralization of IFN-γ, or ST2 attenuated MMP12 expression and prevented progression of emphysema in these mice. Taken together, our results indicate that RV may stimulate expression of CXCL-10 and IFN-γ via activation of ST2/IL-33 signaling axis, which in turn promote accumulation of CD11b+/CD11c+ macrophages and CD8+ T cells. Furthermore, RV-induced IFN-γ stimulates MMP12 expression particularly in CD11b+/CD11c+ macrophages, which may degrade alveolar walls thus leading to progression of emphysema in these mice. In conclusion, our data suggest an important role for ST2/IL-33 signaling axis in RV-induced pathological changes in COPD mice.
Collapse
|
18
|
Shen F, Jiang L, Han F, Degos V, Chen S, Su H. Increased Inflammatory Response in Old Mice is Associated with More Severe Neuronal Injury at the Acute Stage of Ischemic Stroke. Aging Dis 2019; 10:12-22. [PMID: 30705764 PMCID: PMC6345332 DOI: 10.14336/ad.2018.0205] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/05/2018] [Indexed: 12/18/2022] Open
Abstract
Stroke occurs mostly in patients with advanced age. Elderly patients have a less favorable prognosis compared with young adult patients. To understand the underlying mechanisms, we tested our hypothesis that an increased inflammatory response to acute ischemic injury in old stroke mice leads to more severe brain damage and behavioral dysfunction. An ischemic stroke model was created in 2- and 12-month-old C57BL/6 mice through permanent occlusion of the left distal middle cerebral artery (dMCAO). Infarct/atrophy volumes were quantified by staining the brain sections with Cresyl Violet. Sensorimotor function was assessed using the corner test and adhesive removal test. Quantification of CD68+ cells in the peri-infarct region was performed at 1, 3 and 14 days after dMCAO. Interleukin-6 (IL-6), interleukin-1 β (IL-1β) and vascular endothelial growth factor (VEGF) levels in the ischemic brain tissue were measured using ELISA. Western blot was used to determine the expression levels of tight junction proteins, claudin-5 and zonula occludens (ZO)-1. Blood-brain barrier permeability was measured by Evans blue (EB) extravasation. Gelatinase B (MMP-9, type IV collagenase) was measured by gel zymography. Compared to 2-month-old mice, 12-month-old mice had more severe behavioral deficits at both the acute and chronic stages of stroke. Compared with the 2-month-old mice, 12-month-old mice had larger infarct/atrophy volumes at 1 and 14 days after dMCAO, higher levels of IL-6 and IL-1β, higher MMP9 activity, and lower levels of claudin-5 and ZO-1 at 1 and 3 days after dMCAO. 12-month-old mice also had more CD68+ cells in the peri-infarct region at 1, 3 and 14 days after dMCAO and more EB leakage at 3 days after dMCAO. A higher inflammatory response at the acute stage of ischemic stroke in old mice is associated with more severe neuronal injury and long-term behavioral dysfunction.
Collapse
Affiliation(s)
- Fanxia Shen
- 1Department of Neurology & Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Lidan Jiang
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Frank Han
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Vincent Degos
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| | - Shengdi Chen
- 1Department of Neurology & Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Su
- 2Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
19
|
Xiao C, Wu M, Liu J, Gu J, Jiao X, Lu D, He J, Lin C, Xue Y, Fu T, Wang H, Wang G, Yang X, Li Z. Acute tobacco smoke exposure exacerbates the inflammatory response to corneal wounds in mice via the sympathetic nervous system. Commun Biol 2019; 2:33. [PMID: 30701198 PMCID: PMC6345828 DOI: 10.1038/s42003-018-0270-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 12/17/2018] [Indexed: 12/18/2022] Open
Abstract
Exposure to tobacco smoke is a major public health concern that can also affect ophthalmic health. Based on previous work demonstrating the important role of the sympathetic nervous system (SNS) in corneal wound repair, we postulated that acute tobacco smoke exposure (ATSE) may act through the SNS in the impairment of corneal wound repair. Here we find that ATSE rapidly increases the markers of inflammatory response in normal corneal limbi. After an abrasion injury, ATSE exaggerates inflammation, impairs wound repair, and enhances the expression of nuclear factor-κB (NF-κB) and inflammatory molecules such as interleukin-6 (IL-6) and IL-17. We find that chemical SNS sympathectomy, local adrenergic receptor antagonism, NF-κB1 inactivation, and IL-6/IL-17A neutralization can all independently attenuate ATSE-induced excessive inflammatory responses and alleviate their impairment of the healing process. These findings highlight that the SNS may represent a major molecular sensor and mediator of ATSE-induced inflammation.
Collapse
Affiliation(s)
- Chengju Xiao
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Mingjuan Wu
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Jun Liu
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Jianqin Gu
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Xinwei Jiao
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Jingxin He
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
- Department of Ophthalmology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Cuipei Lin
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
- Department of Ophthalmology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yunxia Xue
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Ting Fu
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Hanqing Wang
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Guang Wang
- Department of Histology and Embryology, Jinan University Medical School, Guangzhou, China
| | - Xuesong Yang
- Department of Histology and Embryology, Jinan University Medical School, Guangzhou, China
| | - Zhijie Li
- International Ocular Surface Research Center, Institute of Ophthalmology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
- Section of Leukocyte Biology, Department of Pediatrics, Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
20
|
Fischer K, Doehn JM, Herr C, Lachner C, Heinrich A, Kershaw O, Voss M, Jacobson MH, Gruber AD, Clauss M, Witzenrath M, Bals R, Gutbier B, Slevogt H. Acute Moraxella catarrhalis Airway Infection of Chronically Smoke-Exposed Mice Increases Mechanisms of Emphysema Development: A Pilot Study. Eur J Microbiol Immunol (Bp) 2018; 8:128-134. [PMID: 30719329 PMCID: PMC6348706 DOI: 10.1556/1886.2018.00019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/07/2018] [Indexed: 12/20/2022] Open
Abstract
In chronic obstructive pulmonary disease (COPD), acute exacerbations and emphysema development are characteristics for disease pathology. COPD is complicated by infectious exacerbations with acute worsening of respiratory symptoms with Moraxella catarrhalis as one of the most frequent pathogens. Although cigarette smoke (CS) is the primary risk factor, additional molecular mechanisms for emphysema development induced by bacterial infections are incompletely understood. We investigated the impact of M. catarrhalis on emphysema development in CS exposed mice and asked whether an additional infection would induce a solubilization of pro-apoptotic and pro-inflammatory endothelial monocyte-activating-protein-2 (EMAPII) to exert its activities in the pulmonary microvas-culature and other parts of the lungs not exposed directly to CS. Mice were exposed to smoke (6 or 9 months) and/or infected with M. catarrhalis. Lungs, bronchoalveolar lavage fluid (BALF), and plasma were analyzed. CS exposure reduced ciliated area, caused rarefaction of the lungs, and induced apoptosis. EMAPII was increased independent of prior smoke exposure in BALF of infected mice. Importantly, acute M. catarrhalis infection increased release of matrixmetalloproteases-9 and -12, which are involved in emphysema development and comprise a mechanism of EMAPII release. Our data suggest that acute M. catarrhalis infection represents an independent risk factor for emphysema development in smoke-exposed mice.
Collapse
Affiliation(s)
- Katja Fischer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany.,Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Jan-Moritz Doehn
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
| | - Christian Herr
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, University of the Saarland, Homburg Saar, Germany
| | - Carolin Lachner
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Annina Heinrich
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Olivia Kershaw
- Department of Veterinary Pathology, Freie Universität, Berlin, Germany
| | - Meike Voss
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, University of the Saarland, Homburg Saar, Germany
| | - Max H Jacobson
- Pathology and Laboratory Medicine, IU School of Medicine, Indianapolis, Indiana, USA
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität, Berlin, Germany
| | - Matthias Clauss
- Indiana Center for Vascular Biology and Medicine and Department of Cellular and Integrative Physiology, Indiana University, Indianapolis, Indiana, USA
| | - Martin Witzenrath
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Division of Pulmonary Inflammation, Berlin, Germany
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, University of the Saarland, Homburg Saar, Germany
| | - Birgitt Gutbier
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Division of Pulmonary Inflammation, Berlin, Germany
| | - Hortense Slevogt
- Septomics Research Center, Jena University Hospital, Jena, Germany
| |
Collapse
|
21
|
Ramos CDO, Campos KKD, Costa GDP, Cangussú SD, Talvani A, Bezerra FS. Taurine treatment decreases inflammation and oxidative stress in lungs of adult mice exposed to cigarette smoke. Regul Toxicol Pharmacol 2018; 98:50-57. [PMID: 30026134 DOI: 10.1016/j.yrtph.2018.07.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 02/02/2023]
Abstract
Taurine is the major free amino acid found in mammalian cells and is known to be an antioxidant and membrane-stabilizing agent. This study aimed to evaluate the effects of taurine on oxidative stress and inflammatory response in the lungs of mice exposed to cigarette smoke. Fifty male C57BL/6 mice were divided into 5 groups: control group (CG), vehicle group (VG), taurine group (TG), cigarette smoke group (CSG), and cigarette smoke + taurine group (CSTG). For five consecutive days, CSG and CSTG were exposed to 4 cigarettes 3 times a day. Taurine administration was able to reduce total leukocytes in bronchoalveolar lavage fluid in CSTG compared to CSG. There was an increase in antioxidant superoxide dismutase and catalase activity in CSG compared to that in CG and TG, and a decrease in CSTG compared to CSG. There was an increase in the concentration of TNF and IL-17 in CSG and CSTG compared to CG and TG. There was an increase in the concentration of IL-22 in CSG compared to CG and TG, and a decrease in CSTG compared to CSG. The administration of taurine has been shown to reduce the inflammation and oxidative stress induced by short-term exposure to cigarette smoke.
Collapse
Affiliation(s)
- Camila de Oliveira Ramos
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - Keila Karine Duarte Campos
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - Guilherme de Paula Costa
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences(NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - Sílvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences(NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG, Brazil.
| |
Collapse
|
22
|
Mebratu YA, Tesfaigzi Y. IL-17 Plays a Role in Respiratory Syncytial Virus-induced Lung Inflammation and Emphysema in Elastase and LPS-injured Mice. Am J Respir Cell Mol Biol 2018; 58:717-726. [PMID: 29314865 PMCID: PMC6002655 DOI: 10.1165/rcmb.2017-0265oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/09/2018] [Indexed: 01/01/2023] Open
Abstract
Respiratory syncytial virus (RSV) is associated with enhanced progression of chronic obstructive pulmonary disease (COPD) and COPD exacerbations. However, little is known about the role of IL-17 in RSV-induced lung injury. We first investigated the role of RSV infection in enhancing mucous cell hyperplasia (MCH) and airspace enlargement in the lungs of mice injured with elastase and LPS (E/LPS). Mice injured with E/LPS had an enhanced and prolonged neutrophilic response to RSV that was associated with decreased levels of type I IFN and increased levels of IL-17, IL-23, CXCL-1, granulocyte colony stimulating factor (GCSF), CXCL-5, and matrix metalloproteinase (MMP)-9. In addition, extent of MCH and mean weighted alveolar space were increased significantly in the lungs of E/LPS-injured mice infected with RSV compared with E/LPS-only or RSV-only controls. Interestingly, immunodepletion of IL-17 before viral infection diminished the RSV-driven MCH and airspace enlargement in the E/LPS-injured animals, suggesting that IL-17 may be a therapeutic target for MCH and airspace enlargement when enhanced by RSV infection.
Collapse
Affiliation(s)
- Yohannes A Mebratu
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Yohannes Tesfaigzi
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| |
Collapse
|
23
|
Long Noncoding RNAs and mRNA Regulation in Peripheral Blood Mononuclear Cells of Patients with Chronic Obstructive Pulmonary Disease. Mediators Inflamm 2018; 2018:7501851. [PMID: 29725270 PMCID: PMC5872599 DOI: 10.1155/2018/7501851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/08/2017] [Accepted: 11/27/2017] [Indexed: 12/18/2022] Open
Abstract
Background Inflammation plays a pivotal role in the pathogenesis of chronic obstructive pulmonary disease (COPD). We evaluated the lncRNA and mRNA expression profile of peripheral blood mononuclear cells (PBMCs) from healthy nonsmokers, smokers without airflow limitation, and COPD patients. Methods lncRNA and mRNA profiling of PBMCs from 17 smokers and 14 COPD subjects was detected by high-throughput microarray. The expression of dysregulated lncRNAs was validated by qPCR. The lncRNA targets in dysregulated mRNAs were predicted and the GO enrichment was analyzed. The regulatory role of lncRNA ENST00000502883.1 on CXCL16 expression and consequently the effect on PBMC recruitment were investigated by siRNA knockdown and chemotaxis analysis. Results We identified 158 differentially expressed lncRNAs in PBMCs from COPD subjects compared with smokers. The dysregulated expression of 5 selected lncRNAs NR_026891.1 (FLJ10038), ENST00000502883.1 (RP11-499E18.1), HIT000648516, XR_429541.1, and ENST00000597550.1 (CTD-2245F17.3), was validated. The GO enrichment showed that leukocyte migration, immune response, and apoptosis are the main enriched processes that previously reported to be involved in the pathogenesis of COPD. The regulatory role of ENST00000502883.1 on CXCL16 expression and consequently the effect on PBMC recruitment was confirmed. Conclusion This study may provide clues for further studies targeting lncRNAs to control inflammation in COPD.
Collapse
|
24
|
Kaur G, Bagam P, Pinkston R, Singh DP, Batra S. Cigarette smoke-induced inflammation: NLRP10-mediated mechanisms. Toxicology 2018; 398-399:52-67. [PMID: 29501574 DOI: 10.1016/j.tox.2018.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 02/07/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive, life-threatening disease that causes irreversible lung damage. Cigarette smoking is the chief etiologic factor for the commencement of this condition. Despite constant efforts to develop therapeutic interventions and to ascertain the molecular mechanism leading to the pathophysiology of this disease, much remains unknown. However, pattern recognition receptors (PRRs), i.e., Toll-like-receptors (TLRs) and NOD-like receptors (NLRs) are believed to play important roles in COPD and could serve as effective therapeutic targets. Although the role of TLRs in COPD has been well studied, the importance of NLRs has not yet been explored in detail. The NLR family member NLRP10 (aka NOD8, PAN5, PYNOD) is the only member of this family of proteins that lacks the leucine rich repeat (LRR) domain responsible for detection of pathogen and danger-associated molecular patterns (PAMPs/DAMPs). Therefore, instead of functioning as a PRR, NLRP10 may have a broader regulatory role. To elucidate the role of NLRP10 in secondhand smoke (SHS)-induced inflammation, we exposed C57Bl/6 (WT) and Nlrp10-deficient mice (Nlrp10-/-) on the C57Bl/6 background to filtered air- or SHS- for 6 weeks (acute exposure) and assessed the resulting molecular events. Leukocyte recruitment in SHS-exposed Nlrp10-/- mice was found to be significantly lower compared to SHS-exposed WT mice. In addition, we observed an important role for NLRP10 in SHS-mediated caspase-1 activation, cytokine/chemokine production (IL-1β, IL-18, MCP-1 and IL-17A), and induction of NF-κB and MAPKs in the lungs of C57Bl/6 mice. The reduced influx of CD4+IL-17A+ and CD8+IL-17A+ cells into the lungs of SHS-exposed Nlrp10-/- mice and impaired differentiation of Nlrp10-/- Th0 cells into Th17 cells (ex vivo) provide insight into the mechanistic details underlying NLRP10-dependent IL-17 production. We further substantiated our in vivo findings by challenging human alveolar type II epithelial cells (A549) transfected with scrambled- or Nlrp10-siRNA with cigarette smoke extract (CSE). We observed an important role of NLRP10 in cytokine and chemokine production as well as expression of NF-κB and MAPKs in CSE-exposed A549 cells. Furthermore, replenishment of A549 cell culture with recombinant IL-17A (rIL-17A) during NLRP10 knockdown rescued CSE-induced inflammatory responses. To identify upstream mediators of NLRP10 regulation we investigated epigenetic markers within the Nlrp10 promoter following cigarette smoke exposure and observed significant changes in active as well as repressive gene markers on histone 3 and histone 4 using both in vivo and in vitro study models. Further, alterations in the respective histone acetyl- and methyltransferases (PCAF, SET1, ESET, SUV20H1) correlated well with the observed histone modifications. Overall, our findings suggest a novel role of epigenetically regulated NLRP10 in Th17/IL-17 signaling during CS exposure.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, United States
| | - Prathyusha Bagam
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, United States
| | - Rakeysha Pinkston
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, United States
| | - Dhirendra P Singh
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, United States
| | - Sanjay Batra
- Laboratory of Pulmonary Immuno-toxicology, Environmental Toxicology Department, Health Research Center, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, United States; Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States.
| |
Collapse
|
25
|
Mark NM, Kargl J, Busch SE, Yang GHY, Metz HE, Zhang H, Hubbard JJ, Pipavath SNJ, Madtes DK, Houghton AM. Chronic Obstructive Pulmonary Disease Alters Immune Cell Composition and Immune Checkpoint Inhibitor Efficacy in Non-Small Cell Lung Cancer. Am J Respir Crit Care Med 2018; 197:325-336. [PMID: 28934595 PMCID: PMC5803651 DOI: 10.1164/rccm.201704-0795oc] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/20/2017] [Indexed: 12/19/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC) are interrelated diseases with substantial mortality, and the pathogenesis of both involves aberrant immune functioning. OBJECTIVES To profile immune cell composition and function in patients with NSCLC and describe the effects of COPD on lung and tumor microenvironments. METHODS We profiled resected lung and tumor tissue using flow cytometry and T-cell receptor sequencing in patients with and without COPD from a prospective cohort of patients undergoing resection of NSCLC. A murine cigarette smoke exposure model was used to evaluate the effect on pulmonary immune populations. A separate retrospective cohort of patients who received immune checkpoint inhibitors (ICIs) was analyzed, and their survival was quantified. MEASUREMENTS AND MAIN RESULTS We observed an increased number of IFN-γ-producing CD8+ and CD4+ (T-helper cell type 1 [Th1]) lymphocytes in the lungs of patients with COPD. In both humans and mice, increased Th17 content was seen with smoke exposure, but was not associated with the development or severity of COPD. COPD-affected lung tissue displayed increased Th1 differentiation that was recapitulated in the matching tumor sample. PD-1 (programmed cell death protein 1) expression was increased in tumors of patients with COPD, and the presence of COPD was associated with progression-free survival in patients treated with ICIs. CONCLUSIONS In patients with COPD, Th1 cell populations were expanded in both lung and tumor microenvironments, and the presence of COPD was associated with longer progression-free intervals in patients treated with ICIs. This has implications for understanding the immune mediators of COPD and developing novel therapies for NSCLC.
Collapse
Affiliation(s)
- Nicholas M. Mark
- Division of Pulmonary and Critical Care, and
- Clinical Research Division and
| | - Julia Kargl
- Clinical Research Division and
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | - David K. Madtes
- Division of Pulmonary and Critical Care, and
- Clinical Research Division and
| | - A. McGarry Houghton
- Division of Pulmonary and Critical Care, and
- Clinical Research Division and
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; and
| |
Collapse
|
26
|
Xue H, Li MX. MicroRNA-150 protects against cigarette smoke-induced lung inflammation and airway epithelial cell apoptosis through repressing p53: MicroRNA-150 in CS-induced lung inflammation. Hum Exp Toxicol 2017; 37:920-928. [PMID: 29205062 DOI: 10.1177/0960327117741749] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cigarette smoke (CS) exposure is an important risk factor for chronic obstructive pulmonary disease (COPD). MicroRNA-150 (miR-150) is involved in several inflammatory diseases. However, little is known about the role of miR-150 in the pathogenesis of COPD. In this study, we established a CS-related mouse model of COPD and evaluated the impact of miR-150 on CS-induced lung inflammation. We further investigated the effects of miR-150 overexpression on pro-inflammatory cytokine production and apoptosis in airway epithelial cells exposed to CS extract (CSE). It was found that miR-150 was significantly ( p < 0.05) downregulated in the lungs of CS-exposed mice, compared to control mice under normal air. The CSE-exposed BEAS-2B airway epithelial cells displayed a four- to six-fold reduction in miR-150 levels, compared to control cells ( p < 0.05). Delivery of miR-150 mimic attenuated CS-induced lung inflammation and accumulation of neutrophils, lymphocytes, and macrophages in bronchoalveolar lavage fluid. Moreover, miR-150 overexpression prevented the induction of interleukin-6, tumor necrosis factor alpha, and interleukin-8 expression and nuclear factor kappa B (NF-κB) transcriptional activity in BEAS-2B cells by CSE. Additionally, miR-150 protected BEAS-2B cells from CSE-induced apoptosis, which was associated with reduced p53 expression. Co-expression of p53 restored apoptotic response to CSE in miR-150-overexpressing BEAS-2B cells. Collectively, miR-150 suppresses CS-induced lung inflammation and airway epithelial cell apoptosis, which is causally linked to repression of p53 expression and NF-κB activity. Restoration of miR-150 expression may represent a potential therapeutic strategy for CS-related COPD.
Collapse
Affiliation(s)
- H Xue
- 1 School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - M X Li
- 2 Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
27
|
Le Rouzic O, Pichavant M, Frealle E, Guillon A, Si-Tahar M, Gosset P. Th17 cytokines: novel potential therapeutic targets for COPD pathogenesis and exacerbations. Eur Respir J 2017; 50:1602434. [PMID: 29025886 DOI: 10.1183/13993003.02434-2016] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 07/14/2017] [Indexed: 12/31/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease of the airways caused mainly by cigarette smoke exposure. COPD progression is marked by exacerbations of the disease, often associated with infections. Recent data show the involvement in COPD pathophysiology of interleukin (IL)-17 and IL-22, two cytokines that are important in the control of lung inflammation and infection. During the initiation and progression of the disease, increased IL-17 secretion causes neutrophil recruitment, leading to chronic inflammation, airways obstruction and emphysema. In the established phase of COPD, a defective IL-22 response facilitates pathogen-associated infections and disease exacerbations. Altered production of these cytokines involves a complex network of immune cells and dysfunction of antigen-presenting cells. In this review, we describe current knowledge on the involvement of IL-17 and IL-22 in COPD pathophysiology at steady state and during exacerbations, and discuss implications for COPD management and future therapeutic approaches.
Collapse
Affiliation(s)
- Olivier Le Rouzic
- Université de Lille, U1019 - UMR 8204, Lung Infection and Innate Immunity, Center for Infection and Immunity of Lille (CIIL), Lille, France
- CNRS, UMR 8204, Lille, France
- INSERM, U1019, Lille, France
- Institut Pasteur de Lille, Lille, France
- Service de Pneumologie Immunologie et Allergologie, CHU Lille, Lille, France
| | - Muriel Pichavant
- Université de Lille, U1019 - UMR 8204, Lung Infection and Innate Immunity, Center for Infection and Immunity of Lille (CIIL), Lille, France
- CNRS, UMR 8204, Lille, France
- INSERM, U1019, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Emilie Frealle
- Université de Lille, U1019 - UMR 8204, Lung Infection and Innate Immunity, Center for Infection and Immunity of Lille (CIIL), Lille, France
- CNRS, UMR 8204, Lille, France
- INSERM, U1019, Lille, France
- Institut Pasteur de Lille, Lille, France
- Laboratoire de Parasitologie et Mycologie Médicale, CHU Lille, Lille, France
| | - Antoine Guillon
- Service de Réanimation Polyvalente, CHRU de Tours, Tours, France
- Inserm, U1100 - Centre d'Etude des Pathologies Respiratoires, Tours, France
- Université François Rabelais, Tours, France
| | - Mustapha Si-Tahar
- Inserm, U1100 - Centre d'Etude des Pathologies Respiratoires, Tours, France
- Université François Rabelais, Tours, France
| | - Philippe Gosset
- Université de Lille, U1019 - UMR 8204, Lung Infection and Innate Immunity, Center for Infection and Immunity of Lille (CIIL), Lille, France
- CNRS, UMR 8204, Lille, France
- INSERM, U1019, Lille, France
- Institut Pasteur de Lille, Lille, France
| |
Collapse
|
28
|
Wang W, Qu X, Dang X, Shang D, Yang L, Li Y, Xu D, Martin JG, Hamid Q, Liu J, Chang Y. Human β-defensin-3 induces IL-8 release and apoptosis in airway smooth muscle cells. Clin Exp Allergy 2017; 47:1138-1149. [PMID: 28437599 DOI: 10.1111/cea.12943] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/12/2017] [Accepted: 04/14/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Human airway smooth muscle cells (ASMCs) may have a pro-inflammatory role through the release of inflammatory mediators. Increasing evidence indicates that human β-defensins (HBDs) are related to pathogenesis of asthma. OBJECTIVES To examine the plasma level of HBD-1, HBD-2 and HBD-3 in asthmatic patients and the expression of their mouse orthologues in the lung tissue of a mouse model of chronic severe asthma. Further to investigate the effect of HBD-3 on the release of the pro-inflammatory cytokine IL-8 and to explore the mechanisms. METHODS The plasma levels of HBD-1, HBD-2 and HBD-3 from 34 healthy controls and 25 asthmatic patients were determined by ELISA. The expression of mouse β-defensins MBD-1, MBD-3 and MBD-14 in the lung tissue of asthmatic mice was detected by Western blot. The ASMCs were cultured with HBD-3 for 24 hour, and then the supernatant level of IL-8 was evaluated by ELISA and the cell viability was examined by WST-1 assay. The signalling pathway was investigated with blocking antibodies or pharmacological inhibitors. RESULTS The plasma levels of HBD-1 and HBD-3 were elevated in asthmatic patients, and the expression of MBD-14, the mouse orthologue for HBD-3, was increased in asthmatic mice. HBD-3-induced IL-8 production in a CCR6 receptor-specific manner and was dependent on multiple signalling pathways. Moreover, HBD-3-induced cell apoptosis concurrently, which was dependent on the ERK1/2 MAPK pathway. Mitochondrial ROS regulated both HBD-3-induced IL-8 production and cell apoptosis. CONCLUSIONS AND CLINICAL RELEVANCE These observations provide clear evidence of an important new mechanism for the promotion of airway inflammation and tissue remodelling with potential relevance for the treatment of asthma.
Collapse
Affiliation(s)
- W Wang
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - X Qu
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - X Dang
- Department of Respiration, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - D Shang
- Department of Respiration, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - L Yang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Y Li
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - D Xu
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - J G Martin
- Meakins-Christie Laboratories and Respiratory Division, The Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Q Hamid
- Meakins-Christie Laboratories and Respiratory Division, The Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada.,College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - J Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Y Chang
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
29
|
Loss of Peripheral Tolerance in Emphysema. Phenotypes, Exacerbations, and Disease Progression. Ann Am Thorac Soc 2016; 12 Suppl 2:S164-8. [PMID: 26595734 DOI: 10.1513/annalsats.201503-115aw] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Heterogeneity in the development and progression of cigarette smoke-induced lung diseases strongly argues for a need to improve the clinical and phenotypic characterization of patients with chronic obstructive lung disease and emphysema. Smokers with emphysema are at a much higher risk for accelerated loss of lung function, increased cardiovascular morbidity, and development of lung cancer. Recent evidence in human translational studies and animal models suggests that emphysema is associated with activation of specialized antigen-presenting cells and that cigarette smoke can disrupt the induction of immune tolerance in the lungs. Quantitative assessment of cytokines expressed by autoreactive T lymphocytes in response to human lung elastin fragments has shown a strong positive correlation between T helper Type 1 (Th1) and Th17 cells' immune responses and emphysema. In search of factors that could reduce the threshold for induction of autoimmune inflammation, we have discovered that cleavage of complement protein 3 (C3) generates bioactive molecules (e.g., C3a) and activates lung antigen-presenting cells. The autocrine and paracrine function of C3a and its receptor are required in T cell-mediated inflammatory responses to cigarette smoke in both human and preclinical models of emphysema. Targeting upstream molecules that reduce the potential for generation of autoreactive T cells could lead to the development of novel therapeutics to prevent progression of emphysema in smokers.
Collapse
|
30
|
Polverino F, Seys LJM, Bracke KR, Owen CA. B cells in chronic obstructive pulmonary disease: moving to center stage. Am J Physiol Lung Cell Mol Physiol 2016; 311:L687-L695. [PMID: 27542809 DOI: 10.1152/ajplung.00304.2016] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 08/15/2016] [Indexed: 12/22/2022] Open
Abstract
Chronic inflammatory responses in the lungs contribute to the development and progression of chronic obstructive pulmonary disease (COPD). Although research studies focused initially on the contributions of the innate immune system to the pathogenesis of COPD, more recent studies have implicated adaptive immune responses in COPD. In particular, studies have demonstrated increases in B cell counts and increases in the number and size of B cell-rich lymphoid follicles in COPD lungs that correlate directly with COPD severity. There are also increases in lung levels of mediators that promote B cell maturation, activation, and survival in COPD patients. B cell products such as autoantibodies directed against lung cells, components of cells, and extracellular matrix proteins are also present in COPD lungs. These autoantibodies may contribute to lung inflammation and injury in COPD patients, in part, by forming immune complexes that activate complement components. Studies of B cell-deficient mice and human COPD patients have linked B cells most strongly to the emphysema phenotype. However, B cells have protective activities during acute exacerbations of COPD by promoting adaptive immune responses that contribute to host defense against pathogens. This review outlines the evidence that links B cells and B cell-rich lymphoid follicles to the pathogenesis of COPD and the mechanisms involved. It also reviews the potential and limitations of B cells as therapeutic targets to slow the progression of human COPD.
Collapse
Affiliation(s)
- Francesca Polverino
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico; University of Parma, Parma, Italy; and
| | - Leen J M Seys
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico;
| |
Collapse
|
31
|
Matera MG, Page C, Rogliani P, Calzetta L, Cazzola M. Therapeutic Monoclonal Antibodies for the Treatment of Chronic Obstructive Pulmonary Disease. Drugs 2016; 76:1257-1270. [DOI: 10.1007/s40265-016-0625-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
32
|
Zhang M, Fei X, Zhang GQ, Zhang PY, Li F, Bao WP, Zhang YY, Zhou X. Role of neutralizing anti-murine interleukin-17A monoclonal antibody on chronic ozone-induced airway inflammation in mice. Biomed Pharmacother 2016; 83:247-256. [PMID: 27380433 DOI: 10.1016/j.biopha.2016.06.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 12/19/2022] Open
Abstract
Exposure to ozone has led to airway inflammation and airway hyperresponsiveness, which potential mechanisms relate to ozone-induced oxidative stress. IL-17 is a growing target for autoimmune and inflammatory diseases. The aim of the study was to examine the inhibitory effects of anti-murine interleukin-17A monoclonal antibody (IL-17mAb) on adverse effects of ozone which are noted above. After C57/BL6 mice were exposed to ozone (2.5ppm; 3h) for 12 times over 6 weeks, IL-17mAb, PBS was intraperitoneally injected into mice 1h after ozone or air exposure for 6 weeks and mice were studied 24h after final exposure, monitoring bronchial responsiveness, airway inflammatory cells, lung histology, levels of neutrophil-related chemokine and proinflammatory cytokines in bronchoalveolar lavage (BAL) fluid and serum, the expression of IL-17A mRNA and protein, glucocorticoid receptors (GR), and the phosphorylation of p38MAPK in lung tissues. The administration of IL-17mAb reduced the ozone-induced increases in total cells, especially neutrophils; decreased levels of cytokines, including IL-8 in BAL fluid, IL-8 and IL-17A in serum; mitigated the severity of airway hyperresponsiveness; attenuated lung inflammation scores and histologic analysis confirmed the suppression of lung inflammation, compared with the administration of a control PBS. Exposure to ozone results in increases in IL-17A production rate, mRNA and protein levels of IL-17A and the protein level of GR. These effects were halted and reversed by IL-17mAb treatment. Furthermore, IL-17mAb also reduced the phosphorylation of p38MAPK. Therefore, we conclude that IL-17mAb may be a useful therapy in ozone-related diseases, including COPD.
Collapse
Affiliation(s)
- Min Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Fei
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Qing Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Peng-Yu Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Li
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wu-Ping Bao
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Ying Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Zhou
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
33
|
Dessalle K, Narayanan V, Kyoh S, Mogas A, Halayko AJ, Nair P, Baglole CJ, Eidelman DH, Ludwig MS, Hamid Q. Human bronchial and parenchymal fibroblasts display differences in basal inflammatory phenotype and response to IL-17A. Clin Exp Allergy 2016; 46:945-56. [PMID: 27079765 DOI: 10.1111/cea.12744] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 04/08/2016] [Accepted: 04/08/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND Chronic inflammation, typified by increased expression of IL-17A, together with airway and parenchymal remodelling are features of chronic lung diseases. Emerging evidence suggests that phenotypic heterogeneity of repair and inflammatory capacities of fibroblasts may contribute to the differential structural changes observed in different regions of the lung. OBJECTIVE To investigate phenotypic differences in parenchymal and bronchial fibroblasts, either in terms of inflammation and remodelling or the ability of these fibroblasts to respond to IL-17A. METHODS Four groups of primary fibroblasts were used: normal human bronchial fibroblast (NHBF), normal human parenchymal fibroblast (NHPF), COPD human bronchial fibroblast (CHBF) and COPD human parenchymal fibroblast (CHPF). Cytokine and extracellular matrix (ECM) expression were measured at baseline and after stimulation with IL-17A. Actinomycin D was used to measure cytokine mRNA stability. RESULTS At baseline, we observed higher protein production of IL-6 in NHPF than NHBF, but higher levels of IL-8 and GRO-α in NHBF. IL-17A induced a higher expression of GRO-α (CXCL1) and IL-6 in NHPF than in NHBF, and a higher level of IL-8 expression in NHBF. IL-17A treatment decreased the mRNA stability of IL-6 in NHBF when compared with NHPF. CHPF expressed higher protein levels of fibronectin, collagen-I and collagen-III than CHBF, NHBF and NHPF. IL-17A increased fibronectin and collagen-III protein only in NHPF and collagen-III protein production in CHBF and CHPF. CONCLUSIONS AND CLINICAL RELEVANCE These findings provide insight into the inflammatory and remodelling processes that may be related to the phenotypic heterogeneity of fibroblasts from airway and parenchymal regions and in their response to IL-17A.
Collapse
Affiliation(s)
- K Dessalle
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - V Narayanan
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - S Kyoh
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - A Mogas
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - A J Halayko
- Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - P Nair
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare and Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - C J Baglole
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - D H Eidelman
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - M S Ludwig
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - Q Hamid
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada.,College of Medicine, University of Sharjah, UAE
| |
Collapse
|
34
|
Pabon MA, Ma KC, Choi AMK. Autophagy and Obesity-Related Lung Disease. Am J Respir Cell Mol Biol 2016; 54:636-46. [PMID: 26900794 PMCID: PMC5455357 DOI: 10.1165/rcmb.2016-0045ps] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 02/22/2016] [Indexed: 12/11/2022] Open
Abstract
Obesity-related disease is a significant source of premature death and economic burden globally. It is also a common comorbidity in patients suffering from lung disease, affecting both severity and treatment success. However, this complex association between obesity and the lung is poorly understood. Autophagy is a self-recycling homeostatic process that has been linked to beneficial or deleterious effects, depending on the specific lung disease. Obesity affects autophagy in a tissue-specific manner, activating autophagy in adipocytes and impairing autophagy in hepatocytes, immune cells, and pancreatic β-cells, among others. Obesity is also characterized by chronic low-grade inflammation that can be modulated by the pro- and antiinflammatory effects of the autophagic machinery. Scant evidence exists regarding the impact of autophagy in obesity-related lung diseases, but there are communal pathways that could be related to disease pathogenesis. Important signaling molecules in obesity, including IL-17, leptin, adiponectin, NLRP3 inflammasome, and TLR-4, have been implicated in the pathogenesis of lung disease. These mediators are known to be modulated by autophagy activity. In this perspective, we highlight the recent advances in the understanding of autophagy in obesity-related conditions, as well as the potential mechanisms that can link autophagy and obesity in the pathogenesis of lung disease.
Collapse
Affiliation(s)
- Maria A Pabon
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Kevin C Ma
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
35
|
Yu A, Duan H, Zhang T, Pan Y, Kou Z, Zhang X, Lu Y, Wang S, Yang Z. IL-17A promotes microglial activation and neuroinflammation in mouse models of intracerebral haemorrhage. Mol Immunol 2016; 73:151-7. [PMID: 27107665 DOI: 10.1016/j.molimm.2016.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 04/04/2016] [Accepted: 04/05/2016] [Indexed: 10/21/2022]
Abstract
Microglial activation is an important contributor to neuroinflammation in intracerebral haemorrhage (ICH). IL-17A has been demonstrated to be involved in neuroinflammatory diseases such as multiple sclerosis. However, the exact mechanism of IL-17A mediated microglial activation in ICH has not been well identified. The purpose of this experiment is to investigate the role of IL-17A in ICH induced microglial activation and neuroinflammation. ICH mice were made by injection of autologous blood model. IL-17A expression and inflammatory factors in perihematomal region, and neurological function of mice were examined after ICH. In addition, IL-17A-neutralizing antibody was utilized to potentially prevent microglial activation and neuroinflammation in ICH mice. The expression of IL-17A, inflammatory factors and microglial activation in perihematomal region were significantly increased, and neurological function of mice was impaired after ICH. In addition, IL-17A Ab prevented ICH-induced cytokine expression, including TNF-α, IL-1β and IL-6, and downstream signaling molecules, including MyD88, TRIF, IκBα, and NF-κBp65 expression, and attenuated microglial activation. IL-17A Ab significantly reduced brain water content and improved neurological function of ICH mice. In conclusion, our results demonstrated that IL-17A was involved in ICH-induced microglial activation and neuroinflammation. IL-17A Ab might also provide a promising therapeutic strategy in ICH.
Collapse
Affiliation(s)
- Anyong Yu
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Haizhen Duan
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Tianxi Zhang
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Yong Pan
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Zhi Kou
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Xiaojun Zhang
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Yuanlan Lu
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Song Wang
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Zhao Yang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China.
| |
Collapse
|
36
|
Inflammatory and Immune Response Genes Polymorphisms are Associated with Susceptibility to Chronic Obstructive Pulmonary Disease in Tatars Population from Russia. Biochem Genet 2016; 54:388-412. [DOI: 10.1007/s10528-016-9726-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/11/2016] [Indexed: 02/08/2023]
|
37
|
Jiang Z, Zhu L. Update on molecular mechanisms of corticosteroid resistance in chronic obstructive pulmonary disease. Pulm Pharmacol Ther 2016; 37:1-8. [PMID: 26805715 DOI: 10.1016/j.pupt.2016.01.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/14/2016] [Accepted: 01/20/2016] [Indexed: 12/24/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is an inflammatory and irreversible pulmonary disorder that is characterized by inflammation and airway destruction. In recent years, COPD has become a global epidemic due to increased air pollution and exposure to cigarette smoke. Current therapeutics using bronchiodialator and anti-inflammatory corticosteroids are most widely used for all patients with persistent COPD, but these approaches are disappointing due to limited improvement in symptom control and survival rate. More importantly, a certain number of COPD patients are resistant to the corticosteroid treatment and their symptoms worsen. Therefore, more effective anti-inflammatory drugs and combinational treatment are required. Understanding of the underlying molecular and immunological mechanisms is critical to developing new therapeutics. Lung inflammation and the released pro-inflammatory cytokines affect glucocorticoid receptor (GR), histone deacetylase 2 (HDAC2) and surfactant protein D (SP-D) activities in many cell types. Macrophages, neutrophils, airway epithelial cells and lymphocytes are involved in the induction of corticosteroid resistance. This review updated the recent advances in molecular and immunological mechanisms of steroid resistance among patients and animal models with COPD. Meanwhile we discussed novel therapeutic approaches in controlling lung inflammation and improving corticosteroid sensitivity among the steroid resistant patients with COPD.
Collapse
Affiliation(s)
- Zhilong Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China.
| | - Lei Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China.
| |
Collapse
|
38
|
Montalbano AM, Riccobono L, Siena L, Chiappara G, Di Sano C, Anzalone G, Gagliardo R, Ricciardolo FLM, Sorbello V, Pipitone L, Vitulo P, Profita M. Cigarette smoke affects IL-17A, IL-17F and IL-17 receptor expression in the lung tissue: Ex vivo and in vitro studies. Cytokine 2015; 76:391-402. [PMID: 26198032 DOI: 10.1016/j.cyto.2015.07.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/16/2015] [Accepted: 07/13/2015] [Indexed: 12/28/2022]
Abstract
Cigarette smoke is a risk factor for Chronic Obstructive Pulmonary Disease (COPD). Th-17 cytokines are involved in the pathogenesis of COPD. We aimed to evaluate the role of cigarette smoke on the expression of IL-17A, IL-17F and IL-17R in airways of COPD patients. Epithelial and subepithelial immunoreactivity for IL-17A, IL-17F and IL-17R was assessed in surgical specimens from COPD patients (n=15) and from healthy subjects (HC) (n=10) by immunohistochemistry. In vitro, human epithelial cell line 16HBE and A549 as well as PBMC from normal donors were stimulated with cigarette smoke extract (CSE) (0%, 2.5%, 5%, 10%) to evaluate the IL-17A, IL-17F and IL-17R expression by flow cytometry. Furthermore, rhIL-17A and CSE stimulation was evaluated on proliferation and apoptosis in 16HBE and in A549. In central and distal airways immunoreactivity for IL-17A, IL-17F and IL-17R significantly increased in the epithelium and IL-17A in the subepithelium from COPD than in HC. In distal airway, immunoreactivity for IL-17F increased in the subepithelium of COPD than in HC. IL-17A immunoreactivity positively correlate with IL-17R and total pack years in the epithelium from central and distal airways of COPD patients. In vitro, CSE stimulation significantly increased IL-17F and IL-17R in 16HBE (2.5%) and A549 (5%) while IL-17A and IL-17F in PBMC (10%). IL-17A and CSE stimulation, rather than CSE or rhIL-17A alone, significantly increased proliferation in 16HBE and apoptosis in A549. Cigarette smoke increases Th17 immunity in lung tissue of COPD patients, promoting the mechanism of proliferation and apoptosis in airway epithelial cells.
Collapse
Affiliation(s)
- Angela Marina Montalbano
- Unit: "Ex vivo/In vitro Models to Study the Immunopathology and Pharmacology of Airway Diseases", Institute of Biomedicine and Molecular Immunology (IBIM), Italian National Research Council (CNR), Palermo, Italy
| | - Loredana Riccobono
- Unit: "Ex vivo/In vitro Models to Study the Immunopathology and Pharmacology of Airway Diseases", Institute of Biomedicine and Molecular Immunology (IBIM), Italian National Research Council (CNR), Palermo, Italy
| | - Liboria Siena
- Unit: "Ex vivo/In vitro Models to Study the Immunopathology and Pharmacology of Airway Diseases", Institute of Biomedicine and Molecular Immunology (IBIM), Italian National Research Council (CNR), Palermo, Italy
| | - Giuseppina Chiappara
- Unit: "Ex vivo/In vitro Models to Study the Immunopathology and Pharmacology of Airway Diseases", Institute of Biomedicine and Molecular Immunology (IBIM), Italian National Research Council (CNR), Palermo, Italy
| | - Caterina Di Sano
- Unit: "Ex vivo/In vitro Models to Study the Immunopathology and Pharmacology of Airway Diseases", Institute of Biomedicine and Molecular Immunology (IBIM), Italian National Research Council (CNR), Palermo, Italy
| | - Giulia Anzalone
- Unit: "Ex vivo/In vitro Models to Study the Immunopathology and Pharmacology of Airway Diseases", Institute of Biomedicine and Molecular Immunology (IBIM), Italian National Research Council (CNR), Palermo, Italy
| | - Rosalia Gagliardo
- Unit: "Ex vivo/In vitro Models to Study the Immunopathology and Pharmacology of Airway Diseases", Institute of Biomedicine and Molecular Immunology (IBIM), Italian National Research Council (CNR), Palermo, Italy
| | | | - Valentina Sorbello
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Loredana Pipitone
- Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (ISMETT), Palermo, Italy
| | - Patrizio Vitulo
- Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (ISMETT), Palermo, Italy
| | - Mirella Profita
- Unit: "Ex vivo/In vitro Models to Study the Immunopathology and Pharmacology of Airway Diseases", Institute of Biomedicine and Molecular Immunology (IBIM), Italian National Research Council (CNR), Palermo, Italy.
| |
Collapse
|
39
|
Bozinovski S, Seow HJ, Chan SPJ, Anthony D, McQualter J, Hansen M, Jenkins BJ, Anderson GP, Vlahos R. Innate cellular sources of interleukin-17A regulate macrophage accumulation in cigarette- smoke-induced lung inflammation in mice. Clin Sci (Lond) 2015; 129:785-96. [PMID: 26201093 PMCID: PMC4613531 DOI: 10.1042/cs20140703] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 05/27/2015] [Accepted: 07/01/2015] [Indexed: 01/17/2023]
Abstract
Cigarette smoke (CS) is the major cause of chronic obstructive pulmonary disease (COPD). Interleukin-17A (IL-17A) is a pivotal cytokine that regulates lung immunity and inflammation. The aim of the present study was to investigate how IL-17A regulates CS-induced lung inflammation in vivo. IL-17A knockout (KO) mice and neutralization of IL-17A in wild-type (WT) mice reduced macrophage and neutrophil recruitment and chemokine (C-C motif) ligand 2 (CCL2), CCL3 and matrix metalloproteinase (MMP)-12 mRNA expression in response to acute CS exposure. IL-17A expression was increased in non-obese diabetic (NOD) severe combined immunodeficiency SCID) mice with non-functional B- and T-cells over a 4-week CS exposure period, where macrophages accumulated to the same extent as in WT mice. Gene expression analysis by QPCR (quantitative real-time PCR) of isolated immune cell subsets detected increased levels of IL-17A transcript in macrophages, neutrophils and NK/NKT cells in the lungs of CS-exposed mice. In order to further explore the relative contribution of innate immune cellular sources, intracellular IL-17A staining was performed. In the present study, we demonstrate that CS exposure primes natural killer (NK), natural killer T (NKT) and γδ T-cells to produce more IL-17A protein and CS alone increased the frequency of IL17+ γδ T-cells in the lung, whereas IL-17A protein was not detected in macrophages and neutrophils. Our data suggest that activation of innate cellular sources of IL-17A is an essential mediator of macrophage accumulation in CS-exposed lungs. Targeting non-conventional T-cell sources of IL-17A may offer an alternative strategy to reduce pathogenic macrophages in COPD.
Collapse
MESH Headings
- Animals
- Bronchoalveolar Lavage Fluid/cytology
- Bronchoalveolar Lavage Fluid/immunology
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/immunology
- Chemokine CCL2/metabolism
- Chemokine CCL3/genetics
- Chemokine CCL3/immunology
- Chemokine CCL3/metabolism
- Flow Cytometry
- Gene Expression/immunology
- Immunity, Innate/genetics
- Immunity, Innate/immunology
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Lung/immunology
- Lung/metabolism
- Lung/pathology
- Macrophages/immunology
- Macrophages/metabolism
- Matrix Metalloproteinase 12/genetics
- Matrix Metalloproteinase 12/immunology
- Matrix Metalloproteinase 12/metabolism
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Neutrophil Infiltration/immunology
- Pneumonia/genetics
- Pneumonia/immunology
- Pneumonia/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Smoke
- Nicotiana/chemistry
Collapse
Affiliation(s)
- Steven Bozinovski
- School of Health Sciences and Health Innovations Research Institute, RMIT University, Bundoora, VIC 3083, Australia Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, VIC 3010, Australia
| | - Huei Jiunn Seow
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, VIC 3010, Australia
| | - Sheau Pyng Jamie Chan
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, VIC 3010, Australia
| | - Desiree Anthony
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, VIC 3010, Australia
| | - Jonathan McQualter
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, VIC 3010, Australia
| | - Michelle Hansen
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, VIC 3010, Australia
| | - Brendan J Jenkins
- Hudson Institute of Medical Research, Monash University, Clayton, VIC 3168, Australia
| | - Gary P Anderson
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, VIC 3010, Australia
| | - Ross Vlahos
- School of Health Sciences and Health Innovations Research Institute, RMIT University, Bundoora, VIC 3083, Australia Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, VIC 3010, Australia
| |
Collapse
|
40
|
Maneechotesuwan K, Wongkajornsilp A, Adcock IM, Barnes PJ. Simvastatin Suppresses Airway IL-17 and Upregulates IL-10 in Patients With Stable COPD. Chest 2015; 148:1164-76. [PMID: 26043025 PMCID: PMC4631035 DOI: 10.1378/chest.14-3138] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/15/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Statins have immunomodulatory properties that may provide beneficial effects in the treatment of COPD. We investigated whether a statin improves the IL-17/IL-10 imbalance in patients with COPD, as has previously been demonstrated in patients with asthma. METHODS Thirty patients with stable COPD were recruited to a double-blind, randomized, controlled, crossover trial comparing the effect of simvastatin, 20 mg po daily, with that of a matched placebo on sputum inflammatory markers and airway inflammation. Each treatment was administered for 4 weeks separated by a 4-week washout period. The primary outcome was the presence of T-helper 17 cytokines and indoleamine 2,3-dioxygenase (IDO) in induced sputum. Secondary outcomes included sputum inflammatory cells, FEV1, and symptoms using the COPD Assessment Test (CAT). RESULTS At 4 weeks, there was a significant reduction in sputum IL-17A, IL-22, IL-6, and CXCL8 concentrations (mean difference, -16.4 pg/mL, P = .01; -48.6 pg/mL, P < .001; -45.3 pg/mL, P = .002; and -190.9 pg/mL, P = .007, respectively), whereas IL-10 concentrations, IDO messenger RNA expression (fold change), and IDO activity (kynurenine to tryptophan ratio) were markedly increased during simvastatin treatment compared with placebo treatment periods (mean difference, 24.7 pg/mL, P < .001; 1.02, P < .001; and 0.47, P < .001, respectively). The absolute sputum macrophage count, proportion of macrophages, and CAT score were reduced after simvastatin compared with placebo (mean difference, -0.16 × 106, P = .004; -14.1%, P < .001; and -3.2, P = .02, respectively). Values for other clinical outcomes were similar between the simvastatin and placebo treatments. CONCLUSIONS Simvastatin reversed the IL-17A/IL-10 imbalance in the airways and reduced sputum macrophage but not neutrophil counts in patients with COPD. TRIAL REGISTRY ClinicalTrials.gov; No.: NCT01944176; www.clinicaltrials.gov.
Collapse
|
41
|
You R, Lu W, Shan M, Berlin JM, Samuel EL, Marcano DC, Sun Z, Sikkema WK, Yuan X, Song L, Hendrix AY, Tour JM, Corry DB, Kheradmand F. Nanoparticulate carbon black in cigarette smoke induces DNA cleavage and Th17-mediated emphysema. eLife 2015; 4:e09623. [PMID: 26437452 PMCID: PMC4612775 DOI: 10.7554/elife.09623] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/15/2015] [Indexed: 12/24/2022] Open
Abstract
Chronic inhalation of cigarette smoke is the major cause of sterile inflammation and pulmonary emphysema. The effect of carbon black (CB), a universal constituent of smoke derived from the incomplete combustion of organic material, in smokers and non-smokers is less known. In this study, we show that insoluble nanoparticulate carbon black (nCB) accumulates in human myeloid dendritic cells (mDCs) from emphysematous lung and in CD11c+ lung antigen presenting cells (APC) of mice exposed to smoke. Likewise, nCB intranasal administration induced emphysema in mouse lungs. Delivered by smoking or intranasally, nCB persisted indefinitely in mouse lung, activated lung APCs, and promoted T helper 17 cell differentiation through double-stranded DNA break (DSB) and ASC-mediated inflammasome assembly in phagocytes. Increasing the polarity or size of CB mitigated many adverse effects. Thus, nCB causes sterile inflammation, DSB, and emphysema and explains adverse health outcomes seen in smokers while implicating the dangers of nCB exposure in non-smokers. DOI:http://dx.doi.org/10.7554/eLife.09623.001 Smoking for many years damages the lungs and leads to a disease called emphysema that makes it difficult to breathe and is often deadly. There are thousands of chemicals in cigarette smoke and many of them have been linked to the development of lung cancer, although it has been difficult to pinpoint those that are responsible for smoking-related emphysema. Moreover, cigarette smoke also contains large numbers of small particles and relatively little is known about the role played by these particles in smoking-related disease. One of the hallmarks of long-term smoking is a blackening of the lung tissue that persists even if someone stops smoking. Previously, little was known about the composition of the substance that causes this blackening, or its significance in the development of emphysema. Now, by studying lung tissue taken from smokers with emphysema, You et al. have shown that this black substance is made of nano-sized particles of a material called carbon black (which is also known as elemental carbon). These nanoparticles are produced by the incomplete combustion of the cigarettes. You et al. also confirmed that nanoparticles of carbon black can cause emphysema in mice. Closer examination of the lung damage caused by the nanoparticles revealed that they trigger breakages in DNA, which leads to inflammation of the lung. And because the nanoparticles cannot be cleared, they are released into the lung when cells die, which perpetuates lung inflammation and damage. You et al. then went on to show that nanoparticles of carbon black can be modified in a way that allows them to be cleared from the lungs. Such modifications could potentially protect people who are exposed to carbon black nanoparticles in the environment or in workplaces where carbon black is used, such as factories that produce automobile tires and other rubber products. DOI:http://dx.doi.org/10.7554/eLife.09623.002
Collapse
Affiliation(s)
- Ran You
- Department of Medicine, Baylor College of Medicine, Houston, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Biology of Inflammation Center, Baylor College of Medicine, Houston, United States
| | - Wen Lu
- Department of Medicine, Baylor College of Medicine, Houston, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Biology of Inflammation Center, Baylor College of Medicine, Houston, United States
| | - Ming Shan
- Department of Medicine, Baylor College of Medicine, Houston, United States
| | - Jacob M Berlin
- Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, United States.,Irell & Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, United States
| | - Errol Lg Samuel
- Department of Chemistry, Rice University, Houston, United States
| | | | - Zhengzong Sun
- Department of Chemistry, Rice University, Houston, United States
| | | | - Xiaoyi Yuan
- Department of Medicine, Baylor College of Medicine, Houston, United States
| | - Lizhen Song
- Department of Medicine, Baylor College of Medicine, Houston, United States
| | - Amanda Y Hendrix
- Department of Medicine, Baylor College of Medicine, Houston, United States
| | - James M Tour
- Department of Chemistry, Rice University, Houston, United States
| | - David B Corry
- Department of Medicine, Baylor College of Medicine, Houston, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Biology of Inflammation Center, Baylor College of Medicine, Houston, United States.,Michael E. DeBakey VA Center, US Department of Veterans Affairs, Houston, United States
| | - Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Biology of Inflammation Center, Baylor College of Medicine, Houston, United States.,Michael E. DeBakey VA Center, US Department of Veterans Affairs, Houston, United States
| |
Collapse
|
42
|
Prakash YS, Tschumperlin DJ, Stenmark KR. Coming to terms with tissue engineering and regenerative medicine in the lung. Am J Physiol Lung Cell Mol Physiol 2015; 309:L625-38. [PMID: 26254424 DOI: 10.1152/ajplung.00204.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/04/2015] [Indexed: 01/10/2023] Open
Abstract
Lung diseases such as emphysema, interstitial fibrosis, and pulmonary vascular diseases cause significant morbidity and mortality, but despite substantial mechanistic understanding, clinical management options for them are limited, with lung transplantation being implemented at end stages. However, limited donor lung availability, graft rejection, and long-term problems after transplantation are major hurdles to lung transplantation being a panacea. Bioengineering the lung is an exciting and emerging solution that has the ultimate aim of generating lung tissues and organs for transplantation. In this article we capture and review the current state of the art in lung bioengineering, from the multimodal approaches, to creating anatomically appropriate lung scaffolds that can be recellularized to eventually yield functioning, transplant-ready lungs. Strategies for decellularizing mammalian lungs to create scaffolds with native extracellular matrix components vs. de novo generation of scaffolds using biocompatible materials are discussed. Strengths vs. limitations of recellularization using different cell types of various pluripotency such as embryonic, mesenchymal, and induced pluripotent stem cells are highlighted. Current hurdles to guide future research toward achieving the clinical goal of transplantation of a bioengineered lung are discussed.
Collapse
Affiliation(s)
- Y S Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota;
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota; Division of Pulmonary Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| |
Collapse
|
43
|
Dai H, Xu L, Tang Y, Liu Z, Sun T. Treatment with a neutralising anti-rat interleukin-17 antibody after multiple-trauma reduces lung inflammation. Injury 2015; 46:1465-70. [PMID: 26100210 DOI: 10.1016/j.injury.2015.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/13/2015] [Accepted: 05/01/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND It has been well recognised that a deficit of numbers and function of CD4(+)CD25(+)Foxp3(+) cells (Treg) is attributed to the development of autoimmune diseases and inflammatory diseases; additionally, IL-17-producing cells (Th17) have a pro-inflammatory role. The balance between Th17 and Treg may be essential for maintaining immune homeostasis and has long been thought as one of the important factors in the development/prevention of autoimmune diseases and inflammatory diseases. In our previous research, we explored that cytokines (IL-17) and the balance of Treg/Th17 had a significant relevance with tissue (lung) inflammation and injury in acute-phase after multiple-trauma. OBJECTIVE To more verify whether an imbalance of Treg/Th17 is characteristic of rats suffering from multiple trauma. METHODS AND SUBJECTIVE Using IL-17 monoclonal antibody (IL-17mAb)-treated multiple-trauma rat, we tested the pathogenic role of IL-17 in the development of multiple-trauma. Rat models were treated respectively with IL-17mAb or rat IgG 2A isotype control or phosphate-buffered solution after model was established. Normal rats only received anaesthesia and cannulation were taken as sham. Rats in each group were killed respectively at the end of 1h, 4h, 8h after injection. Collected serum and lung samples for assessment dynamically of MPO, IL-17, IL-6, and TGF-β-mRNA, and cytokine (IL-17, IL-6, TGF-β) and lung tissue for pulmonary histological analysis. RESULTS Neutralisation of IL-17 with anti-IL-17 can decrease serum IL-17 level and the IL-17-mRNA transcript level in lung, and ameliorate tissue inflammatory, defer disease course. CONCLUSION Our data suggest that IL-17 is crucially involved in the pathogenesis of multiple-trauma in rat, IL-17 inhibition might ameliorate the lung inflammation in acute-phase after multiple-trauma.
Collapse
Affiliation(s)
- Heling Dai
- Institute of Orthopaedics, Chinese PLA, Beijing Army General Hospital, Dongcheng District, Nanmencang No. 5, Beijing 100700, China.
| | - Li Xu
- Institute of Orthopaedics, Chinese PLA, Beijing Army General Hospital, Dongcheng District, Nanmencang No. 5, Beijing 100700, China; Laboratory, Department of Second Hospital, Jilin University, Changchun, China
| | - Yu Tang
- Institute of Orthopaedics, Chinese PLA, Beijing Army General Hospital, Dongcheng District, Nanmencang No. 5, Beijing 100700, China
| | - Zhi Liu
- Institute of Orthopaedics, Chinese PLA, Beijing Army General Hospital, Dongcheng District, Nanmencang No. 5, Beijing 100700, China
| | - Tiansheng Sun
- Institute of Orthopaedics, Chinese PLA, Beijing Army General Hospital, Dongcheng District, Nanmencang No. 5, Beijing 100700, China. suntiansheng-@163.com
| |
Collapse
|
44
|
Yuan X, Shan M, You R, Frazier MV, Hong MJ, Wetsel RA, Drouin S, Seryshev A, MD LZS, Cornwell L, Rossen RD, Corry DB, Kheradmand F. Activation of C3a receptor is required in cigarette smoke-mediated emphysema. Mucosal Immunol 2015; 8:874-85. [PMID: 25465103 PMCID: PMC4454642 DOI: 10.1038/mi.2014.118] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 10/23/2014] [Indexed: 02/04/2023]
Abstract
Exposure to cigarette smoke can initiate sterile inflammatory responses in the lung and activate myeloid dendritic cells (mDCs) that induce differentiation of T helper type 1 (Th1) and Th17 cells in the emphysematous lungs. Consumption of complement proteins increases in acute inflammation, but the contribution of complement protein 3 (C3) to chronic cigarette smoke-induced immune responses in the lung is not clear. Here, we show that following chronic exposure to cigarette smoke, C3-deficient (C3(-/-)) mice develop less emphysema and have fewer CD11b(+)CD11c(+) mDCs infiltrating the lungs as compared with wild-type mice. Proteolytic cleavage of C3 by neutrophil elastase releases C3a, which in turn increases the expression of its receptor (C3aR) on lung mDCs. Mice deficient in the C3aR (C3ar(-/-)) partially phenocopy the attenuated responses to chronic smoke observed in C3(-/-) mice. Consistent with a role for C3 in emphysema, C3 and its active fragments are deposited on the lung tissue of smokers with emphysema, and smoke-exposed mice. Together, these findings suggest a critical role for C3a through autocrine/paracrine induction of C3aR in the pathogenesis of cigarette smoke-induced sterile inflammation and provide new therapeutic targets for the treatment of emphysema.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Ming Shan
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Ran You
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Michael V. Frazier
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Monica Jeongsoo Hong
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Rick A. Wetsel
- Brown Foundation Institute of Molecular Medicine-Research Center for Immunology and Autoimmune Diseases, The University of Texas Medical School at Houston, Houston TX 77030
| | - Scott Drouin
- Brown Foundation Institute of Molecular Medicine-Research Center for Immunology and Autoimmune Diseases, The University of Texas Medical School at Houston, Houston TX 77030
| | - Alexander Seryshev
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Li-zhen Song MD
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | | | - Roger D Rossen
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Pathology and Immunology, Baylor College of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Michael E DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston TX 77030
| | - David B. Corry
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Pathology and Immunology, Baylor College of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Biology of Inflammation Center, Baylor College of Medicine, Houston TX 77030
- Michael E DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston TX 77030
| | - Farrah Kheradmand
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Pathology and Immunology, Baylor College of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Biology of Inflammation Center, Baylor College of Medicine, Houston TX 77030
- Michael E DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston TX 77030
| |
Collapse
|
45
|
Bozinovski S, Vlahos R. Multifaceted Role for IL-17A in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2015; 191:1213-4. [DOI: 10.1164/rccm.201504-0714ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
46
|
Expression variations of connective tissue growth factor in pulmonary arteries from smokers with and without chronic obstructive pulmonary disease. Sci Rep 2015; 5:8564. [PMID: 25708588 PMCID: PMC4338434 DOI: 10.1038/srep08564] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 01/19/2015] [Indexed: 12/19/2022] Open
Abstract
Cigarette smoking contributes to the development of pulmonary hypertension (PH) complicated with chronic obstructive pulmonary disease (COPD), and the pulmonary vascular remodeling, the structural basis of PH, could be attributed to abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs).In this study, morphometrical analysis showed that the pulmonary vessel wall thickness in smoker group and COPD group was significantly greater than in nonsmokers. In addition, we determined the expression patterns of connective tissue growth factor (CTGF) and cyclin D1 in PASMCs harvested from smokers with normal lung function or mild to moderate COPD, finding that the expression levels of CTGF and cyclin D1 were significantly increased in smoker group and COPD group. In vitro experiment showed that the expression of CTGF, cyclin D1 and E2F were significantly increased in human PASMCs (HPASMCs) treated with 2% cigarette smoke extract (CSE), and two CTGF siRNAs with different mRNA hits successfully attenuated the upregulated cyclin D1 and E2F, and significantly restored the CSE-induced proliferation of HPASMCs by causing cell cycle arrest in G0. These findings suggest that CTGF may contribute to the pathogenesis of abnormal proliferation of HPASMCs by promoting the expression of its downstream effectors in smokers with or without COPD.
Collapse
|
47
|
Chang Y, Al-Alwan L, Alshakfa S, Audusseau S, Mogas AK, Chouiali F, Nair P, Baglole CJ, Hamid Q, Eidelman DH. Upregulation of IL-17A/F from human lung tissue explants with cigarette smoke exposure: implications for COPD. Respir Res 2014; 15:145. [PMID: 25427574 PMCID: PMC4268887 DOI: 10.1186/s12931-014-0145-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/04/2014] [Indexed: 01/10/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is an inflammatory disorder marked by relative resistance to steroids. The IL-17 superfamily, which mediates cross-talk between the adaptive and innate immune systems, has been associated with diminished responses to steroids. Increasing evidence supports elevated IL-17 expression in the lung of COPD subjects. However, whether cells of the immune system (systemic) and/or local lung cells are contributing to the elevated IL-17 remains unclear. To address this issue, we utilized a human parenchymal lung tissue explant culture system with cigarette smoke exposure to investigate the expression of IL-17 and the mechanisms involved. Methods Parenchymal lung tissue removed from 10 non-COPD and 8 COPD patients was sectioned and cultured with different concentrations of cigarette smoke extract (CSE) for 3 or 6 hours. Tissue viability was evaluated by LDH (lactate dehydrogenase) in culture supernatants. Western blot and real-time PCR were performed to evaluate IL-17A/F expression. To investigate the mechanisms, pharmacological inhibitors for MAPK p38, ERK1/2, NF-κB and PI3K pathways were added into the culture media. Results No tissue damage was observed after the cigarette smoke exposure for 3 h or 6 h compared with the control media. At the protein level, the expression of both IL-17A (2.4 ± 0.6 fold) and IL-17 F (3.7 ± 0.7 fold) in the tissue from non-COPD subjects was significantly increased by 5% of CSE at 3 h. For COPD subjects, IL-17A/F expression were significantly increased only at 6 h with 10% of CSE (IL-17A: 4.2 ± 0.8 fold; IL-17 F: 3.3 ± 0.8 fold). The increased expression of IL-17A/F is also regulated at the mRNA level. The inhibitors for NF-κB and PI3K pathways significantly inhibited CSE-induced IL-17A/F expression from lung tissue of non-COPD subjects. Conclusions We found the evidence that the expression of both IL-17A and IL-17 F is increased by the cigarette smoke exposure in explants from both non-COPD and COPD subjects, supporting that local lung cells contribute IL-17 production. The elevated IL-17A/F expression is dependent on NF-κB and PI3K pathways. These observations add to the growing evidence which suggests that Th17 cytokines play a significant role in COPD.
Collapse
Affiliation(s)
- Ying Chang
- Meakins-Christie Laboratories and Respiratory Division, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada. .,Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Laila Al-Alwan
- Meakins-Christie Laboratories and Respiratory Division, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Sama Alshakfa
- Meakins-Christie Laboratories and Respiratory Division, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Severine Audusseau
- Meakins-Christie Laboratories and Respiratory Division, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Andrea Karen Mogas
- Meakins-Christie Laboratories and Respiratory Division, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Fazila Chouiali
- Meakins-Christie Laboratories and Respiratory Division, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Parameswaran Nair
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare and Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | - Carolyn J Baglole
- Meakins-Christie Laboratories and Respiratory Division, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Qutayba Hamid
- Meakins-Christie Laboratories and Respiratory Division, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada.
| | - David H Eidelman
- Meakins-Christie Laboratories and Respiratory Division, McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
48
|
Hansen MJ, Chan SPJ, Langenbach SY, Dousha LF, Jones JE, Yatmaz S, Seow HJ, Vlahos R, Anderson GP, Bozinovski S. IL-17A and serum amyloid A are elevated in a cigarette smoke cessation model associated with the persistence of pigmented macrophages, neutrophils and activated NK cells. PLoS One 2014; 9:e113180. [PMID: 25405776 PMCID: PMC4236152 DOI: 10.1371/journal.pone.0113180] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/20/2014] [Indexed: 12/29/2022] Open
Abstract
While global success in cessation advocacy has seen smoking rates fall in many developed countries, persistent lung inflammation in ex-smokers is an increasingly important clinical problem whose mechanistic basis remains poorly understood. In this study, candidate effector mechanisms were assessed in mice exposed to cigarette smoke (CS) for 4 months following cessation from long term CS exposure. BALF neutrophils, CD4+ and CD8+ T cells and lung innate NK cells remained significantly elevated following smoking cessation. Analysis of neutrophil mobilization markers showed a transition from acute mediators (MIP-2α, KC and G-CSF) to sustained drivers of neutrophil and macrophage recruitment and activation (IL-17A and Serum Amyoid A (SAA)). Follicle-like lymphoid aggregates formed with CS exposure and persisted with cessation, where they were in close anatomical proximity to pigmented macrophages, whose number actually increased 3-fold following CS cessation. This was associated with the elastolytic protease, MMP-12 (macrophage metallo-elastase) which remained significantly elevated post-cessation. Both GM-CSF and CSF-1 were significantly increased in the CS cessation group relative to the control group. In conclusion, we show that smoking cessation mediates a transition to accumulation of pigmented macrophages, which may contribute to the expanded macrophage population observed in COPD. These macrophages together with IL-17A, SAA and innate NK cells are identified here as candidate persistence determinants and, we suggest, may represent specific targets for therapies directed towards the amelioration of chronic airway inflammation.
Collapse
Affiliation(s)
- Michelle J. Hansen
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
- * E-mail:
| | - Sheau Pyng J. Chan
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Shenna Y. Langenbach
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Lovisa F. Dousha
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Jessica E. Jones
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Selcuk Yatmaz
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Huei Jiunn Seow
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Ross Vlahos
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Gary P. Anderson
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| | - Steven Bozinovski
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Victoria, Australia
| |
Collapse
|
49
|
Yu XL, Zhang J, Zhao F, Pan XM. Relationships of COX2 and MMP12 genetic polymorphisms with chronic obstructive pulmonary disease risk: a meta-analysis. Mol Biol Rep 2014; 41:8149-62. [PMID: 25209967 DOI: 10.1007/s11033-014-3715-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 08/28/2014] [Indexed: 01/11/2023]
Abstract
We performed the present meta-analysis in an attempt to confirm the correlation of genetic polymorphisms in the COX2 and MMP12 genes with the susceptibility to chronic obstructive pulmonary disease (COPD). We searched English database such as PubMed, CISCOM, CINAHL, Web of Science, Google Scholar and several Chinese database for meta-analysis. There were no specific language restrictions. Two investigators systematically extracted relevant data within those included studies. Crude ORs with its corresponding 95 % CI were calculated. STATA 12.0 software was adopted for statistical analysis. The impact of COX2 and MMP12 genetic polymorphisms on the pathogenesis of COPD was investigated in the current study with a total of 10 case-control studies, which includes 1,751 COPD patients and 2,472 healthy subjects. Four common polymorphisms, including rs689466 G > A and rs20417 G > C in the COX2 gene, rs652438 A > G and rs2276109 A > G were evaluated in the MMP12 gene. Pooled OR of the present studies and results showed that the frequency of COX2 rs20417 polymorphism was prevalent in COPD patients than those of healthy subjects (C allele vs. G allele OR = 1.33, 95 % CI 1.06-1.67, P = 0.014; GC + CC vs. GG OR = 1.86, 95 % CI 1.07-3.24, P = 0.029; respectively). However, we found no significant correlation between COX2 rs689466 polymorphism and the risk of COPD (all P > 0.05). Furthermore, our meta-analysis illustrated that individuals with MMP12 rs652438 polymorphism had significantly increased risk of developing COPD (G allele vs. A allele OR = 1.62, 95 % CI 1.08-2.42, P = 0.020; AG + GG vs. AA OR = 2.14, 95 % CI 1.12-4.09, P = 0.021; respectively). Nevertheless, no positive relation was detected between MMP12 rs2276109 variant and the risk of COPD. Our meta-analysis indicates that COX2 and MMP12 genetic polymorphisms may be strongly implicated in the development of COPD, especially for the COX2 rs20417 and MMP12 rs652438 polymorphisms. Thus, COX2 and MMP12 genetic polymorphisms could potentially be utilized as helpful biomarkers for early diagnosis of COPD.
Collapse
Affiliation(s)
- Xiao-Ling Yu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, South Seven Road No. 102, Heping District, Shenyang, 110005, People's Republic of China,
| | | | | | | |
Collapse
|
50
|
Zuo L, He F, Sergakis GG, Koozehchian MS, Stimpfl JN, Rong Y, Diaz PT, Best TM. Interrelated role of cigarette smoking, oxidative stress, and immune response in COPD and corresponding treatments. Am J Physiol Lung Cell Mol Physiol 2014; 307:L205-18. [DOI: 10.1152/ajplung.00330.2013] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cigarette smoking (CS) can impact the immune system and induce pulmonary disorders such as chronic obstructive pulmonary disease (COPD), which is currently the fourth leading cause of chronic morbidity and mortality worldwide. Accordingly, the most significant risk factor associated with COPD is exposure to cigarette smoke. The purpose of the present study is to provide an updated overview of the literature regarding the effect of CS on the immune system and lungs, the mechanism of CS-induced COPD and oxidative stress, as well as the available and potential treatment options for CS-induced COPD. An extensive literature search was conducted on the PubMed/Medline databases to review current COPD treatment research, available in the English language, dating from 1976 to 2014. Studies have investigated the mechanism by which CS elicits detrimental effects on the immune system and pulmonary function through the use of human and animal subjects. A strong relationship among continued tobacco use, oxidative stress, and exacerbation of COPD symptoms is frequently observed in COPD subjects. In addition, therapeutic approaches emphasizing smoking cessation have been developed, incorporating counseling and nicotine replacement therapy. However, the inability to reverse COPD progression establishes the need for improved preventative and therapeutic strategies, such as a combination of intensive smoking cessation treatment and pharmaceutical therapy, focusing on immune homeostasis and redox balance. CS initiates a complex interplay between oxidative stress and the immune response in COPD. Therefore, multiple approaches such as smoking cessation, counseling, and pharmaceutical therapies targeting inflammation and oxidative stress are recommended for COPD treatment.
Collapse
Affiliation(s)
- Li Zuo
- Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Feng He
- Department of Health and Kinesiology, Purdue University, Lafayette, Indiana
| | - Georgianna G. Sergakis
- Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Majid S. Koozehchian
- Exercise and Sport Nutrition Laboratory, Department of Health & Kinesiology, Texas A&M University, College Station, Texas
| | - Julia N. Stimpfl
- Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Yi Rong
- Department of Radiation Oncology, James Cancer Hospital, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Philip T. Diaz
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Thomas M. Best
- Division of Sports Medicine, Department of Family Medicine, Sports Health & Performance Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|