51
|
Qin H, Gao F, Wang Y, Huang B, Peng L, Mo B, Wang C. Nur77 promotes cigarette smoke‑induced autophagic cell death by increasing the dissociation of Bcl2 from Beclin-1. Int J Mol Med 2019; 44:25-36. [PMID: 31115481 PMCID: PMC6559304 DOI: 10.3892/ijmm.2019.4184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 04/08/2019] [Indexed: 01/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by partially reversible airflow limitation and persistent alveolar destruction, and autophagy is involved in the pathogenesis of cigarette smoke (CS)‑induced COPD. Nuclear receptor 77 (Nur77) participates in a number of biological processes, including apoptosis, autophagy and in disease pathogenesis; however, the role of Nur77 in COPD remains unknown. Thus, in this study, we aimed to elucidate the role of Nur77 in COPD. We report that CS promotes Nur77 expression and nuclear export in vivo and in vitro, which increases cigarette smoke extract (CSE)‑induced autophagy. In addition, we found that lung tissues, human bronchial epithelial (HBE) cells and A549 cells exposed to CS or CSE expressed lower levels of LC3 and Beclin‑1 and contained fewer autophagosomes following Nur77 knockdown with siRNA‑Nur77. Moreover, a co‑immunoprecipitation assay demonstrated that CSE promoted autophagy, partly by accelerating the interaction between Nur77 and Bcl2, in turn leading to the increased dissociation of Bcl2 from Beclin‑1; by contrast, leptomycin B (LMB) suppressed the dissociation of Bcl2 from Beclin‑1. Taken together, the findings of this study demonstrate that Nur77 is involved in the CSE‑induced autophagic death of lung cells, and that this process is partially dependent on the increased interaction between Nur77 and Bcl2, and on the dissociation of Bcl2 from Beclin‑1. This study illustrates the role of Nur77 in bronchial and alveolar destruction following exposure to CS.
Collapse
Affiliation(s)
- Huiping Qin
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of The National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Feng Gao
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541002
| | - Yanni Wang
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of The National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Bin Huang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541002
| | - Ling Peng
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Key Site of The National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Biwen Mo
- Department of Respiratory Medicine, Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Changming Wang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541002,Correspondence to: Dr Changming Wang, Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, 12 Wenming Road, Guilin, Guangxi 541002, P.R. China, E-mail:
| |
Collapse
|
52
|
Yamaguchi MS, McCartney MM, Falcon AK, Linderholm AL, Ebeler SE, Kenyon NJ, Harper RH, Schivo M, Davis CE. Modeling cellular metabolomic effects of oxidative stress impacts from hydrogen peroxide and cigarette smoke on human lung epithelial cells. J Breath Res 2019; 13:036014. [PMID: 31063985 PMCID: PMC9798928 DOI: 10.1088/1752-7163/ab1fc4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The respiratory system is continuously exposed to variety of biological and chemical irritants that contain reactive oxygen species, and these are well known to cause oxidative stress responses in lung epithelial cells. There is a clinical need to identify biomarkers of oxidative stress which could potentially support early indicators of disease and health management. To identify volatile biomarkers of oxidative stress, we analyzed the headspace above human bronchial epithelial cell cultures (HBE1) before and after hydrogen peroxide (H2O2) and cigarette smoke extract (CSE) exposure. Using stir bar and headspace sorptive extraction-gas chromatography-mass spectrometry, we searched for volatile organic compounds (VOC) of these oxidative measures. In the H2O2 cell peroxidation experiments, four different H2O2 concentrations (0.1, 0.5, 10, 50 mM) were applied to the HBE1 cells, and VOCs were collected every 12 h over the time course of 48 h. In the CSE cell peroxidation experiments, four different smoke extract concentrations (0%, 10%, 30%, 60%) were applied to the cells, and VOCs were collected every 12 h over the time course of 48 h. We used partial-least squares (PLS) analysis to identify putative compounds from the mass spectrometry results that highly correlated with the known applied oxidative stress. We observed chemical emissions from the cells that related to both the intensity of the oxidative stress and followed distinct time courses. Additionally, some of these chemicals are aldehydes, which are thought to be non-invasive indicators of oxidative stress in exhaled human breath. Together, these results illustrate a powerful in situ cell culture model of oxidative stress that can be used to explore the putative biological genesis of exhaled breath biomarkers that are often observed in human clinical studies.
Collapse
Affiliation(s)
- Mei S. Yamaguchi
- Mechanical and Aerospace Engineering, University of California Davis, Davis, CA 95616, USA
| | - Mitchell M. McCartney
- Mechanical and Aerospace Engineering, University of California Davis, Davis, CA 95616, USA
| | - Alexandria K. Falcon
- Mechanical and Aerospace Engineering, University of California Davis, Davis, CA 95616, USA
| | - Angela L. Linderholm
- Center for Comparative Respiratory Biology and Medicine, UC Davis Medical School, Davis, CA 95616, USA
| | - Susan E. Ebeler
- Viticulture and Enology, University of California Davis, One Shields Avenue, Davis, California 95616, USA
| | - Nicholas J. Kenyon
- Center for Comparative Respiratory Biology and Medicine, UC Davis Medical School, Davis, CA 95616, USA,Department of Internal Medicine, 4150 V Street, Suite 3400, University of California, Davis, Sacramento, CA 95817, USA,VA Northern California Health Care System, 10535 Hospital Way, Mather, CA 95655, USA
| | - Richart H. Harper
- Center for Comparative Respiratory Biology and Medicine, UC Davis Medical School, Davis, CA 95616, USA,Department of Internal Medicine, 4150 V Street, Suite 3400, University of California, Davis, Sacramento, CA 95817, USA,VA Northern California Health Care System, 10535 Hospital Way, Mather, CA 95655, USA
| | - Michael Schivo
- Center for Comparative Respiratory Biology and Medicine, UC Davis Medical School, Davis, CA 95616, USA,Department of Internal Medicine, 4150 V Street, Suite 3400, University of California, Davis, Sacramento, CA 95817, USA,VA Northern California Health Care System, 10535 Hospital Way, Mather, CA 95655, USA
| | - Cristina E. Davis
- Mechanical and Aerospace Engineering, University of California Davis, Davis, CA 95616, USA,Corresponding author: Prof. Cristina E. Davis ()
| |
Collapse
|
53
|
Zhao J, Li M, Wang Z, Chen J, Zhao J, Xu Y, Wei X, Wang J, Xie J. Role of PM 2.5 in the development and progression of COPD and its mechanisms. Respir Res 2019; 20:120. [PMID: 31196090 PMCID: PMC6567502 DOI: 10.1186/s12931-019-1081-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/23/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A multitude of epidemiological studies have shown that ambient fine particulate matter 2.5 (diameter < 2.5um; PM2.5) was associated with increased morbidity and mortality of chronic obstructive pulmonary disease (COPD). However, the underlying associated mechanisms have not yet been elucidated. We conducted this study to investigate the role of PM2.5 in the development of COPD and associated mechanisms. METHODS We firstly conducted a cross-sectional study in Chinese han population to observe PM2.5 effects on COPD morbidity. Then, in vitro, we incubated human bronchial epithelial cells to different concentrations of PM2.5 for 24 h. The expression levels of IL-6 and IL-8 were detected by ELISA and the levels of MMPs, TGF-β1, fibronectin and collagen was determined by immunoblotting. In vivo, we subjected C57BL/6 mice to chronic prolonged exposure to PM2.5 for 48 weeks to study the influence of PM2.5 exposure on lung function, pulmonary structure and inflammation. RESULTS We found that the effect of PM2.5 on COPD morbidity was associated with its levels and that PM2.5 and cigarette smoke could have a synergistic impact on COPD development and progression. Both vitro and vivo studies demonstrated that PM2.5 exposure could induce pulmonary inflammation, decrease lung function, and cause emphysematous changes. Furthermore, PM2.5 could markedly aggravated cigarette smoke-induced changes. CONCLUSIONS In short, we found that prolonged chronic exposure to PM2.5 resulted in decreased lung function, emphysematous lesions and airway inflammation. Most importantly, long-term PM2.5 exposure exacerbateed cigarette smoke-induced changes in COPD.
Collapse
Affiliation(s)
- Junling Zhao
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Miao Li
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhihua Wang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinkun Chen
- Acadia Junior High School, Winnipeg, MB, Canada
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yongjian Xu
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jianmao Wang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
54
|
Lycopene mitigates pulmonary emphysema induced by cigarette smoke in a murine model. J Nutr Biochem 2019; 65:93-100. [DOI: 10.1016/j.jnutbio.2018.12.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 10/09/2018] [Accepted: 12/15/2018] [Indexed: 12/20/2022]
|
55
|
Bone morphogenetic protein 6 (BMP-6) modulates lung function, pulmonary iron levels and cigarette smoke-induced inflammation. Mucosal Immunol 2019; 12:340-351. [PMID: 30542109 DOI: 10.1038/s41385-018-0116-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 10/16/2018] [Accepted: 11/20/2018] [Indexed: 02/04/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with abnormal inflammatory responses and airway wall remodeling, leading to reduced lung function. An association between the bone morphogenetic protein (BMP-6) locus and forced vital capacity has been found in a genome-wide association study. However, the role of BMP-6 in the pathogenesis of COPD remains unknown. The pulmonary expression of BMP-6 was analyzed in patients with COPD and in cigarette smoke (CS)-exposed mice. We evaluated lung function and histology in BMP-6 KO mice at baseline. We exposed BMP-6 KO mice to CS for 4 weeks and measured pulmonary inflammation and iron levels. Pulmonary mRNA levels of BMP-6 were decreased in smokers with and without COPD and in CS-exposed mice. Importantly, BMP-6 expression was lowest in severe COPD. Accordingly, protein levels of BMP-6 were decreased in patients with COPD. Lung function measurements demonstrated a decreased compliance and total lung capacity in BMP-6 KO mice, whereas lung histology was normal. Furthermore, BMP-6 KO mice displayed elevated iron levels and an aggravated CS-induced inflammatory response. These results suggest that BMP-6 is important for normal lung function and that downregulation of BMP-6-as observed in patients with COPD-contributes to pulmonary inflammation after CS exposure.
Collapse
|
56
|
Cervilha DAB, Ito JT, Lourenço JD, Olivo CR, Saraiva-Romanholo BM, Volpini RA, Oliveira-Junior MC, Mauad T, Martins MA, Tibério IFLC, Vieira RP, Lopes FDTQS. The Th17/Treg Cytokine Imbalance in Chronic Obstructive Pulmonary Disease Exacerbation in an Animal Model of Cigarette Smoke Exposure and Lipopolysaccharide Challenge Association. Sci Rep 2019; 9:1921. [PMID: 30760822 PMCID: PMC6374436 DOI: 10.1038/s41598-019-38600-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/03/2019] [Indexed: 12/22/2022] Open
Abstract
We proposed an experimental model to verify the Th17/Treg cytokine imbalance in COPD exacerbation. Forty C57BL/6 mice were exposed to room air or cigarette smoke (CS) (12 ± 1 cigarettes, twice a day, 30 min/exposure and 5 days/week) and received saline (50 µl) or lipopolysaccharide (LPS) (1 mg/kg in 50 µl of saline) intratracheal instillations. We analyzed the mean linear intercept, epithelial thickness and inflammatory profiles of the bronchoalveolar lavage fluid and lungs. We evaluated macrophages, neutrophils, CD4+ and CD8+ T cells, Treg cells, and IL-10+ and IL-17+ cells, as well as STAT-3, STAT-5, phospho-STAT3 and phospho-STAT5 levels using immunohistochemistry and IL-17, IL-6, IL-10, INF-γ, CXCL1 and CXCL2 levels using ELISA. The study showed that CS exposure and LPS challenge increased the numbers of neutrophils, macrophages, and CD4+ and CD8+ T cells. Simultaneous exposure to CS/LPS intensified this response and lung parenchymal damage. The densities of Tregs and IL-17+ cells and levels of IL-17 and IL-6 were increased in both LPS groups, while IL-10 level was only increased in the Control/LPS group. The increased numbers of STAT-3, phospho-STAT3, STAT-5 and phospho-STAT5+ cells corroborated the increased numbers of IL-17+ and Treg cells. These findings point to simultaneous challenge with CS and LPS exacerbated the inflammatory response and induced diffuse structural changes in the alveolar parenchyma characterized by an increase in Th17 cytokine release. Although the Treg cell differentiation was observed, the lack of IL-10 expression and the decrease in the density of IL-10+ cells observed in the CS/LPS group suggest that a failure to release this cytokine plays a pivotal role in the exacerbated inflammatory response in this proposed model.
Collapse
Affiliation(s)
- Daniela A B Cervilha
- Department of Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil.
| | - Juliana T Ito
- Department of Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Juliana D Lourenço
- Department of Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Clarice R Olivo
- Department of Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Department of post-graduation of Institute of Medical Assistance to the State Public Servant, University City of Sao Paulo, Sao Paulo, Brazil
| | - Beatriz M Saraiva-Romanholo
- Department of Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Department of post-graduation of Institute of Medical Assistance to the State Public Servant, University City of Sao Paulo, Sao Paulo, Brazil
| | - Rildo A Volpini
- Nephrology Department, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Thais Mauad
- Department of Pathology, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Milton A Martins
- Department of Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Iolanda F L C Tibério
- Department of Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Rodolfo P Vieira
- Post-graduation Program in Bioengineering and in Biomedical Engineering, Universidade Brasil, Sao Paulo, Brazil
- Post-graduation Program in Sciences of Human Movement and Rehabilitation, Federal University of Sao Paulo (UNIFESP), Santos, Brazil
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), Sao Jose dos Campos, Brazil
| | - Fernanda D T Q S Lopes
- Department of Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
57
|
Hussain M, Xu C, Yao M, Zhang Q, Wu J, Wu X, Lu M, Tang L, Wu F, Wu X. CRTH2 antagonist, CT‑133, effectively alleviates cigarette smoke-induced acute lung injury. Life Sci 2019; 216:156-167. [PMID: 30468833 DOI: 10.1016/j.lfs.2018.11.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/16/2018] [Accepted: 11/18/2018] [Indexed: 01/01/2023]
Abstract
AIMS Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), characterized by overwhelming lung inflammation, are associated with high mortality. Cigarette smoke (CS) is one of the major causes of ALI/ARDS. Since high expression of prostaglandin (PG) D2 has been observed in CS-induced lung injury. Currently, no effective pharmacological therapies are available to treat ALI, and supportive therapies remain the mainstay of treatment. Therefore, we investigated the protective effect of CT‑133, a newly discovered selective CRTH2 antagonist, on CS-induced ALI in vivo and in vitro. MAIN METHODS CT‑133 (10 and 30 mg/kg), dexamethasone (1 mg/kg) and normal saline were intratracheally administrated 1 hr prior to whole-body CS-exposure for seven consecutive days to study the key characteristics of ALI. Subsequently, CSE (4%)- and PGD2-stimulated RAW 264.7 macrophages were used to evaluate the protective effect of CT‑133. KEY FINDINGS CT‑133 remarkably attenuated infiltration of inflammatory cells, neutrophils, and macrophages in the BALF, albumin contents, expression of IL‑1β, IL‑6, TNF‑α and KC, lung myeloperoxidase (MPO) activity and lung histopathological alterations caused by CS exposure in mice. Moreover, CT‑133 not only reversed the uncontrolled secretion of IL‑1β, IL-6, TNF‑α and KC from CSE- and PGD2-stimulated RAW 264.7 macrophages but also augmented IL-10 production in both in vivo and in vitro studies. Additionally, CT‑133 alleviated in vitro neutrophil migration chemoattracted by PGD2. SIGNIFICANCE Our results provide the first evidence that targeting CRTH2 could be a new potential therapeutic option to treat CS-induced ALI.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Chengyun Xu
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Minli Yao
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Qin Zhang
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Junsong Wu
- Department of Critical Care Medicine and Orthopedics, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou City 310009, China
| | - Xiling Wu
- Department of Respiratory Medicine, the Affiliated Children Hospital, Zhejiang University School of Medicine, Hangzhou City 310052, China
| | - Meiping Lu
- Department of Respiratory Medicine, the Affiliated Children Hospital, Zhejiang University School of Medicine, Hangzhou City 310052, China
| | - Lanfang Tang
- Department of Respiratory Medicine, the Affiliated Children Hospital, Zhejiang University School of Medicine, Hangzhou City 310052, China
| | - Fugen Wu
- Department of Pediatrics, The First People's Hospital of Wenling City, Wenling City 317500, China.
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China.
| |
Collapse
|
58
|
Cheng Q, Fang L, Feng D, Tang S, Yue S, Huang Y, Han J, Lan J, Liu W, Gao L, Luo Z. Memantine ameliorates pulmonary inflammation in a mice model of COPD induced by cigarette smoke combined with LPS. Biomed Pharmacother 2019; 109:2005-2013. [PMID: 30551456 DOI: 10.1016/j.biopha.2018.11.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 02/07/2023] Open
Abstract
An enhanced chronic inflammatory response in the airways has been regarded as a critical characteristic of chronic obstructive pulmonary disease (COPD). Memantine, an N-methyl-d-aspartate (NMDA) receptors antagonist, has been reported to alleviate lung inflammation. In this study, we investigated the effect and mechanism of memantine on the COPD model induced by cigarette smoke (CS) combined with LPS. Mice and RAW264.7 cells were treated with LPS in the presence or absence of CS. We performed H&E staining to analysis the lung histopathological characteristics. Cytokines (IL-6, TNF-α, and IFN-γ) levels in bronchoalveolar lavage fluid (BALF), lung tissue homogenates and RAW264.7 cell culture medium were determined. Glutamate levels in plasma and culture medium of RAW264.7 were determined. The intracellular Ca2+ flux in RAW264.7 cells was measured by fluo-3 AM staining. The protein levels of NR-1, xCT, ERK1/2, and AKT signaling in the lung tissue and cells were investigated. The result showed that CS and LPS stimulation caused inflammation response, a significant increase in the release of cytokines, including TNF-α, IL-6, and IFN-γ, the elevated release of glutamate and protein levels of NR-1 and xCT, increased Ca2+ influx, and the activation of the ERK1/2 pathway in vitro and in vivo. The above effects of CS and LPS stimulation could be significantly attenuated by memantine treatment. In conclusion, memantine can effectively ameliorate pulmonary inflammation in CS + LPS-induced COPD in mice via reducing NR-1 and xCT expression, glutamate release, Ca2+ influx, and the phosphorylation of Erk1/2. We provided a possible mechanism by which memantine ameliorates COPD in mice.
Collapse
Affiliation(s)
- Qingmei Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lijuan Fang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Dandan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Siyuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Shaojie Yue
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanhong Huang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jianzhong Han
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jinrong Lan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Lihua Gao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
59
|
Guerrina N, Traboulsi H, Eidelman DH, Baglole CJ. The Aryl Hydrocarbon Receptor and the Maintenance of Lung Health. Int J Mol Sci 2018; 19:E3882. [PMID: 30563036 PMCID: PMC6320801 DOI: 10.3390/ijms19123882] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 01/09/2023] Open
Abstract
Much of what is known about the Aryl Hydrocarbon Receptor (AhR) centers on its ability to mediate the deleterious effects of the environmental toxicant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; dioxin). However, the AhR is both ubiquitously-expressed and evolutionarily-conserved, suggesting that it evolved for purposes beyond strictly mediating responses to man-made environmental toxicants. There is growing evidence that the AhR is required for the maintenance of health, as it is implicated in physiological processes such as xenobiotic metabolism, organ development and immunity. Dysregulation of AhR expression and activity is also associated with a variety of disease states, particularly those at barrier organs such as the skin, gut and lungs. The lungs are particularly vulnerable to inhaled toxicants such as cigarette smoke. However, the role of the AhR in diseases such as chronic obstructive pulmonary disease (COPD)-a respiratory illness caused predominately by cigarette smoking-and lung cancer remains largely unexplored. This review will discuss the growing body of literature that provides evidence that the AhR protects the lungs against the damaging effects of cigarette smoke.
Collapse
Affiliation(s)
- Necola Guerrina
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada.
| | - Hussein Traboulsi
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
| | - David H Eidelman
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada.
| | - Carolyn J Baglole
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada.
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada.
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
60
|
Yuan Y, Lin D, Feng L, Huang M, Yan H, Li Y, Chen Y, Lin B, Ma Y, Ye Z, Mei Y, Yu X, Zhou K, Zhang Q, Chen T, Zeng J. Upregulation of miR-196b-5p attenuates BCG uptake via targeting SOCS3 and activating STAT3 in macrophages from patients with long-term cigarette smoking-related active pulmonary tuberculosis. J Transl Med 2018; 16:284. [PMID: 30326918 PMCID: PMC6192289 DOI: 10.1186/s12967-018-1654-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 10/04/2018] [Indexed: 01/01/2023] Open
Abstract
Background Cigarette smoking (CS) triggers an intense and harmful inflammatory response in lungs mediated by alveolar and blood macrophages, monocytes, and neutrophils and is closely associated with prevalence of tuberculosis (TB). The risk of death in patients with long-term cigarette smoking-related pulmonary tuberculosis (LCS-PTB) is approximately 4.5 times higher than those with nonsmoking pulmonary tuberculosis (N-PTB). However, the mechanisms underlying the harmful inflammatory responses in the setting of LCS-PTB have not been well documented. Methods 28 cases LCS-PTB patients, 22 cases N-PTB patients and 20 cases healthy volunteers were enrolled in this study. Monocytes were isolated from peripheral blood mononuclear cells. Differentiated human MDM and U937 cell were prepared with M-CSF and PMA stimulation, respectively. The miR-196b-5p, STAT1, STAT3, STAT4, STAT5A, STAT5B, STAT6, SOCS1 and SOCS3 mRNA expression were detected by qRT-PCR. Western blot was performed according to SOCS1, SOCS3, and pSTAT3 expression. The mycobacterial uptake by MDMs from different groups of patients after Bacillus Calmette–Guérin (BCG) infection and agomir-196b-5p or antagomir-196b-5p transfection were used by flow cytometry analysis. Human IL-6, IL-10 and TNF-α levels on the plasma and cell culture supernatant samples were measured using ELISA. For dual-luciferase reporter assay, the SOCS3 3′-UTR segments, containing the binding elements of miR-196b-5p or its mutant versions were synthesized as sense and antisense linkers. Results In this study, we found that IL-6, TNF-α production, SOCS3 mRNA expression were downregulated, while miR-196b-5p and STAT3 mRNA expression were upregulated in monocytes from LCS-PTB patients as compared to N-PTB patients. Meanwhile, we demonstrated that miR-196b-5p could target SOCS3 and activate STAT3 signaling pathway, which may possibly contribute to attenuation of BCG uptake and decrease in IL-6 and TNF-α production in macrophages. Conclusions Our findings revealed that CS exposure regulates inflammatory responses in monocyte/macrophages from LCS-PTB patients via upregulating miR-196b-5p, and further understanding of the specific role of miR-196b-5p in inflammatory responses mightfacilitate elucidating the pathogenesis of LCS-PTB, thus leading to the development of new therapeutic strategies for PTB patients with long-term cigarette smoking. Electronic supplementary material The online version of this article (10.1186/s12967-018-1654-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yaoqin Yuan
- Dongguan Sixth People's Hospital, Dongguan, 523008, Guangdong, China
| | - Dongzi Lin
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong, China.,Dongguan Sixth People's Hospital, Dongguan, 523008, Guangdong, China
| | - Long Feng
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Mingyuan Huang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Huimin Yan
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong, China.,Provincial Tuberculosis Reference Laboratory of Guangdong, Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630, China
| | - Yumei Li
- Dongguan Sixth People's Hospital, Dongguan, 523008, Guangdong, China
| | - Yinwen Chen
- Dongguan Sixth People's Hospital, Dongguan, 523008, Guangdong, China
| | - Bihua Lin
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yan Ma
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Ziyu Ye
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuezhi Mei
- Dongguan Sixth People's Hospital, Dongguan, 523008, Guangdong, China
| | - Xiaolin Yu
- Dongguan Sixth People's Hospital, Dongguan, 523008, Guangdong, China
| | - Keyuan Zhou
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, 19104, USA
| | - Tao Chen
- Provincial Tuberculosis Reference Laboratory of Guangdong, Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630, China.
| | - Jincheng Zeng
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, Guangdong, China. .,Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, 19104, USA.
| |
Collapse
|
61
|
Zheng X, Zhang L, Chen J, Gu Y, Xu J, Ouyang Y. Dendritic cells and Th17/Treg ratio play critical roles in pathogenic process of chronic obstructive pulmonary disease. Biomed Pharmacother 2018; 108:1141-1151. [PMID: 30372815 DOI: 10.1016/j.biopha.2018.09.113] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/11/2018] [Accepted: 09/19/2018] [Indexed: 12/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common disorder of respiratory system. This study aimed to evaluate changes of mature dendritic cells (DCs) and regulatory T cells (Treg) in lung tissues and peripheral blood of COPD patients. For lung tissue analysis, patients were divided into no-smoking and no-COPD (CS-COPD-), smoking and no-COPD (CS + COPD-) and COPD group. For peripheral blood analysis, patients were divided into CS-COPD-, CS + COPD-, stable COPD (SCOPD) and acute exacerbation of COPD (AECOPD) group. Hematoxylin and eosin (HE) staining was used to evaluate inflammation of lung tissues. Immunohistochemistry assay was employed to examine CD80, CCR6, IL-17 A, FoxP3 in lung tissues. DCs and Treg cells were isolated from lung tissues and peripheral blood. Levels of CD80, FoxP3+ Treg, CCR6 and IL-17 A were detected by using flow cytometry. Results showed that FEV%, FVC% and FEV1/FVC were significantly reduced and Bosken scores were remarkably increased in COPD patients compared to non-COPD patients (p < 0.05). CD80 and FoxP3 levels were lower, and CCR6 and IL-17A levels were higher obviously in COPD compared to non-COPD patients (p < 0.05). COPD patients illustrated reduced mDCs levels and enhanced imDCs levels. COPD patients exhibited remarkably higher Th17 levels compared to no-smoking patients (p < 0.05). COPD patients illustrated obviously lower Treg levels and significantly higher Th17/Treg ratio compared to non-smoking patients (p < 0.05). Th17% (Th17/Treg) negatively and Treg% was positively correlated with FEV1%, FEVC%, FEV1/FEVC (p < 0.05). In conclusion, dendritic cells and Th17/Treg ratio play critical roles for pathogenic process of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Xiangru Zheng
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lanying Zhang
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jie Chen
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yanhui Gu
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jingqing Xu
- Tongji Medical College of HUST, Wuhan, China
| | - Yao Ouyang
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
62
|
Protective effect of water extract of guibi-tang against pulmonary inflammation induced by cigarette smoke and lipopolysaccharide. Lab Anim Res 2018; 34:92-100. [PMID: 30310405 PMCID: PMC6170225 DOI: 10.5625/lar.2018.34.3.92] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/12/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022] Open
Abstract
Water extract of guibi-tang (GB), a traditional Chinese, Japanese, and Korean herbal medicine, is used to treat memory impairment, insomnia, and peptic ulcers. The aim of this study was to investigate the protective effects of GB on pulmonary inflammation induced by cigarette smoke (CS) and lipopolysaccharide (LPS). C57BL/6 mice were used to develop a pulmonary inflammation model by exposing them to CS for 1 h per day for 7 days. LPS was intranasally administered to mice under mild anesthesia on day 5. GB was administered 1 h before CS exposure at doses of 50 or 100 mg/kg for 7 days. Our results showed that GB suppressed the CS and LPS induced elevation in inflammatory cell counts in the bronchoalveolar lavage fluid (BALF), with significant reductions in protein, tumor necrosis factor (TNF)-α, and interleukin (IL)-6 levels. Histological studies revealed that GB decreased the inflammatory cell infiltration into lung tissue caused by CS- and LPS-exposure. GB also significantly decreased the CS and LPS-induced expression of inducible nitric oxide synthase (iNOS) in the lung tissue. Taken together, GB effectively attenuated airway inflammation caused by CS and LPS. These results indicate that GB is a potential therapeutic herbal formula for pulmonary inflammatory disease.
Collapse
|
63
|
Heinemann F, Birk G, Schoenberger T, Stierstorfer B. Deep neural network based histological scoring of lung fibrosis and inflammation in the mouse model system. PLoS One 2018; 13:e0202708. [PMID: 30138413 PMCID: PMC6107205 DOI: 10.1371/journal.pone.0202708] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/07/2018] [Indexed: 01/08/2023] Open
Abstract
Preclinical studies of novel compounds rely on quantitative readouts from animal models. Frequently employed readouts from histopathological tissue scoring are time consuming, require highly specialized staff and are subject to inherent variability. Recent advances in deep convolutional neural networks (CNN) now allow automating such scoring tasks. Here, we demonstrate this for the case of the Ashcroft fibrosis score and a newly developed inflammation score to characterize fibrotic and inflammatory lung diseases. Sections of lung tissue from mice exhibiting a wide range of fibrotic and inflammatory states were stained with Masson trichrome. Whole slide scans using a 20x objective were acquired and cut into smaller tiles of 512x512 pixels. The tiles were subsequently classified by specialized CNNs, either an "Ashcroft fibrosis CNN" or an "inflammation CNN". For the Ashcroft fibrosis score the CNN was fine-tuned by using 14000 labelled tiles. For the inflammation score the CNN was trained with 3500 labelled tiles. After training, the Ashcroft fibrosis CNN achieved an accuracy of 79.5% and the inflammation CNN an accuracy of 80.0%. An error analysis revealed that misclassifications are almost exclusively with neighboring scores, which reflects the inherent ambiguity of parts of the data. The variability between two experts was found to be larger than the variability between the CNN classifications and the ground truth. The CNN generated Ashcroft score was in very good agreement with the score of a pathologist (r2 = 0.92). Our results demonstrate that costly and time consuming scoring tasks can be automated and standardized with deep learning. New scores such as the inflammation score can be easily developed with the approach presented here.
Collapse
Affiliation(s)
- Fabian Heinemann
- Target Discovery Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Gerald Birk
- Target Discovery Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Tanja Schoenberger
- Target Discovery Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Birgit Stierstorfer
- Target Discovery Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| |
Collapse
|
64
|
Ashare RL, Wetherill RR. The Intersection of Sex Differences, Tobacco Use, and Inflammation: Implications for Psychiatric Disorders. Curr Psychiatry Rep 2018; 20:75. [PMID: 30094593 PMCID: PMC7018440 DOI: 10.1007/s11920-018-0946-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Tobacco use, sex differences, and psychiatric disorders are associated with altered immune function. There are also sex differences in tobacco use and psychiatric disorders. This review summarizes findings from the small, but growing literature examining sex differences in the effects of tobacco use on inflammation and the implications for psychiatric disorders. RECENT FINDINGS We identified four studies that tested the interaction between sex and tobacco/nicotine on inflammation. Although males and females generally exhibited differential tobacco-induced immune responses, the pattern varied depending on the sample (rodents vs. humans) and the method to evaluate inflammation. Evidence suggests that sex modulates the effects of tobacco smoke on inflammation. Many inflammation markers associated with sex differences and tobacco use are related to psychiatric disorders. We propose a model in which sex, tobacco use, and inflammation interact to increase risk for psychiatric disorders. Future studies are needed to examine the mechanisms that explain this relationship.
Collapse
Affiliation(s)
- Rebecca L. Ashare
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Suite 4100, Philadelphia, PA 19104, USA
| | - Reagan R. Wetherill
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Suite 4100, Philadelphia, PA 19104, USA
| |
Collapse
|
65
|
Glynos C, Bibli SI, Katsaounou P, Pavlidou A, Magkou C, Karavana V, Topouzis S, Kalomenidis I, Zakynthinos S, Papapetropoulos A. Comparison of the effects of e-cigarette vapor with cigarette smoke on lung function and inflammation in mice. Am J Physiol Lung Cell Mol Physiol 2018; 315:L662-L672. [PMID: 30091379 DOI: 10.1152/ajplung.00389.2017] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Electronic cigarettes (e-cigs) are advertised as a less harmful nicotine delivery system or as a new smoking cessation tool. We aimed to assess the in vivo effects of e-cig vapor in the lung and to compare them to those of cigarette smoke (CS). We exposed C57BL/6 mice for either 3 days or 4 wk to ambient air, CS, or e-cig vapor containing 1) propylene glycol/vegetable glycerol (PG:VG-Sol; 1:1), 2) PG:VG with nicotine (G:VG-N), or 3) PG:VG with nicotine and flavor (PG:VG-N+F) and determined oxidative stress, inflammation, and pulmonary mechanics. E-cig vapors, especially PG:VG-N+F, increased bronchoalveolar lavage fluid (BALF) cellularity, Muc5ac production, as well as BALF and lung oxidative stress markers at least comparably and in many cases more than CS. BALF protein content at both time points studied was only elevated in the PG:VG-N+F group. After 3 days, PG:VG-Sol altered tissue elasticity, static compliance, and airway resistance, whereas after 4 wk CS was the only treatment adversely affecting these parameters. Airway hyperresponsiveness in response to methacholine was increased similarly in the CS and PG:VG-N+F groups. Our findings suggest that exposure to e-cig vapor can trigger inflammatory responses and adversely affect respiratory system mechanics. In many cases, the added flavor in e-cigs exacerbated the detrimental effects of e-cig vapor. We conclude that both e-cig vaping and conventional cigarette smoking negatively impact lung biology.
Collapse
Affiliation(s)
- Constantinos Glynos
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece
| | - Sofia-Iris Bibli
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens , Athens , Greece.,Institute for Vascular Signaling, Centre for Molecular Medicine, Goethe University , Frankfurt am Main , Germany
| | - Paraskevi Katsaounou
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece
| | - Athanasia Pavlidou
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece.,Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens , Athens , Greece
| | - Christina Magkou
- Department of Histopathology, Evangelismos Hospital , Athens , Greece
| | - Vassiliki Karavana
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece
| | - Stavros Topouzis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras , Patras , Greece
| | - Ioannis Kalomenidis
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece
| | - Spyros Zakynthinos
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece
| | - Andreas Papapetropoulos
- George P. Livanos and Marianthi Simou Laboratories, Evangelismos Hospital, 1st Department of Pulmonary and Critical Care, National and Kapodistrian University of Athens Medical School , Greece.,Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
66
|
Zuo J, Hu Z, Liu T, Chen C, Tao Z, Chen S, Li F. Calpeptin attenuates cigarette smoke-induced pulmonary inflammation via suppressing calpain/IκBα signaling in mice and BEAS-2B cells. Pathol Res Pract 2018; 214:1199-1209. [PMID: 30078403 DOI: 10.1016/j.prp.2018.06.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/19/2018] [Accepted: 06/25/2018] [Indexed: 11/29/2022]
Abstract
Exposure to cigarette smoke including secondhand smoking is the most important risk factor in the development of chronic obstructive pulmonary disease where incidence has substantially increased in recent decades. The mechanisms responsible for cigarette smoke-induced pulmonary inflammation remain unclear, and thus lack of effective treatment. The present study investigated the effect of calpeptin on attenuating cigarette smoke induced pulmonary inflammation and its potential mechanism and function. When BALB/c mice were exposed to cigarette smoke and received calpeptin intraperitoneally injection after 90 days, calpeptin histologically attenuated the accumulation of neutrophils (P < 0.001), eosinophils (P < 0.001), macrophages (P < 0.01), fibrinous exudation and proliferation within the interstitial and alveolar spaces. BEAS-2B cells were added with cigarette smoke extract in vitro and treated with calpeptin for 24 h in the treatment group. The markedly upregulation of μ-calpain (P < 0.01), m-calpain (P < 0.001) and IκBα (P < 0.01) in cigarette smoke-induced lungs were simultaneously decreased by calpeptin treatment (P < 0.05). The increased expression of μ-calpain, m-calpain and IκBα (P < 0.05) in cigarette smoke extract-stimulated BEAS-2B cells were also decreased by calpeptin treatment (P < 0.05). These data indicated that calpeptin attenuated cigarette smoke-induced pulmonary inflammation by suppressing the pathway of μ-calpain, m-calpain and IκBα in vivo and in vitro. Calpeptin might have a potential for prevention of the development of inflammatory pulmonary diseases and warrant further pharmaceutical investigation.
Collapse
Affiliation(s)
- Jingjing Zuo
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhangwei Hu
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Tao Liu
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Chen Chen
- Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zezhang Tao
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China.
| | - Shiming Chen
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China; Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Fen Li
- Research Institute of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| |
Collapse
|
67
|
Kerdidani D, Magkouta S, Chouvardas P, Karavana V, Glynos K, Roumelioti F, Zakynthinos S, Wauters E, Janssens W, Lambrechts D, Kollias G, Tsoumakidou M. Cigarette Smoke-Induced Emphysema Exhausts Early Cytotoxic CD8 + T Cell Responses against Nascent Lung Cancer Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:1558-1569. [PMID: 30037849 DOI: 10.4049/jimmunol.1700700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/25/2018] [Indexed: 01/08/2023]
Abstract
Chronic obstructive pulmonary disease is a chronic inflammatory disorder with an increased incidence of lung cancer. The emphysema component of chronic obstructive pulmonary disease confers the greatest proportion to lung cancer risk. Although tumors create inflammatory conditions to escape immunity, the immunological responses that control growth of nascent cancer cells in pre-established inflammatory microenvironments are unknown. In this study, we addressed this issue by implanting OVA-expressing cancer cells in the lungs of mice with cigarette smoke-induced emphysema. Emphysema augmented the growth of cancer cells, an effect that was dependent on T cytotoxic cells. OVA-specific OTI T cells showed early signs of exhaustion upon transfer in emphysema tumor hosts that was largely irreversible because sorting, expansion, and adoptive transfer failed to restore their antitumor activity. Increased numbers of PD-L1- and IDO-positive CD11c+ myeloid dendritic cells (DCs) infiltrated emphysema tumors, whereas sorted emphysema tumor DCs poorly stimulated OTI T cells. Upon adoptive transfer in immunocompetent hosts, T cells primed by emphysema tumor DCs were unable to halt tumor growth. DCs exposed to the emphysema tumor microenvironment downregulated MHC class II and costimulatory molecules, whereas they upregulated PD-L1/IDO via oxidative stress-dependent mechanisms. T cell activation increased upon PD-L1 blockade in emphysema DC-T cell cocultures and in emphysema tumor hosts in vivo. Analysis of the transcriptome of primary human lung tumors showed a strong association between computed tomography-based emphysema scoring and downregulation of immunogenic processes. Thus, suppression of adaptive immunity against lung cancer cells links a chronic inflammatory disorder, emphysema, to cancer, with clinical implications for emphysema patients to be considered optimal candidates for cancer immunotherapies.
Collapse
Affiliation(s)
- Dimitra Kerdidani
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming,' 16672 Vari, Athens, Greece.,Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Sophia Magkouta
- Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Panagiotis Chouvardas
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming,' 16672 Vari, Athens, Greece.,Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Vassiliki Karavana
- Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Konstantinos Glynos
- Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Fani Roumelioti
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming,' 16672 Vari, Athens, Greece
| | - Spyros Zakynthinos
- Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Els Wauters
- Respiratory Oncology Unit, University Hospitals KU Leuven, 3000 Leuven, Belgium.,Leuven Lung Cancer Group, University Hospitals KU Leuven, 3000 Leuven, Belgium.,Laboratory of Pneumology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Wim Janssens
- Respiratory Oncology Unit, University Hospitals KU Leuven, 3000 Leuven, Belgium.,Leuven Lung Cancer Group, University Hospitals KU Leuven, 3000 Leuven, Belgium.,Laboratory of Pneumology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Diether Lambrechts
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium; and.,Laboratory for Translational Genetics, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - George Kollias
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming,' 16672 Vari, Athens, Greece.,Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Maria Tsoumakidou
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming,' 16672 Vari, Athens, Greece;
| |
Collapse
|
68
|
High affinity sugar ligands of C-type lectin receptor langerin. Biochim Biophys Acta Gen Subj 2018; 1862:1592-1601. [PMID: 29631057 DOI: 10.1016/j.bbagen.2018.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Langerin, a C-type lectin receptor (CLR) expressed in a subset of dendritic cells (DCs), binds to glycan ligands for pathogen capture and clearance. Previous studies revealed that langerin has an unusual binding affinity toward 6-sulfated galactose (Gal), a structure primarily found in keratan sulfate (KS). However, details and biological outcomes of this interaction have not been characterized. Based on a recent discovery that the disaccharide L4, a KS component that contains 6-sulfo-Gal, exhibits anti-inflammatory activity in mouse lung, we hypothesized that L4-related compounds are useful tools for characterizing the langerin-ligand interactions and their therapeutic application. METHODS We performed binding analysis between purified long and short forms of langerin and a series of KS disaccharide components. We also chemically synthesized oligomeric derivatives of L4 to develop a new high-affinity ligand of langerin. RESULTS We show that the binding critically requires the 6-sulfation of Gal and that the long form of langerin displays higher affinity than the short form. The synthesized trimeric (also designated as triangle or Tri) and polymeric (pendant) L4 derivatives displayed over 1000-fold higher affinity toward langerin than monomeric L4. The pendant L4, but not the L4 monomer, was found to effectively transduce langerin signaling in a model cell system. CONCLUSIONS L4 is a specific ligand for langerin. Oligomerization of L4 unit increased the affinity toward langerin. GENERAL SIGNIFICANCE These results suggest that oligomeric L4 derivatives will be useful for clarifying the langerin functions and for the development of new glycan-based anti-inflammatory drugs.
Collapse
|
69
|
Onishi M, Kobayashi T, D'Alessandro-Gabazza CN, Fujimoto H, Chelakkot-Govindalayathil AL, Takahashi Y, Yasuma T, Nishihama K, Toda M, Takei Y, Taguchi O, Gabazza EC. Mice overexpressing latent matrix metalloproteinase-2 develop lung emphysema after short-term exposure to cigarette smoke extract. Biochem Biophys Res Commun 2018; 497:332-338. [PMID: 29428733 DOI: 10.1016/j.bbrc.2018.02.081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 02/08/2018] [Indexed: 11/29/2022]
Abstract
Chronic obstructive pulmonary disease is the major growing cause of mortality and morbidity worldwide, and it is going to become the third most common cause of death by 2020. Chronic obstructive pulmonary disease is pathologically characterized by lung emphysema and small airway inflammation. Animal models are very important to get insights into the disease pathogenesis but current models of chronic obstructive pulmonary disease take a long time to develop. The need of a new model is compelling. In the present study we focus on the role of matrix metalloproteinases in the pathogenesis of chronic obstructive pulmonary disease and hypothesized that lung overexpression of latent matrix metalloproteinases-2 would allow the development of emphysema after short-term exposure to cigarette smoke extract inhalation. Human latent matrix metalloproteinases-2 transgenic mouse expressing high level of the protein in the lungs and wild type mouse were exposed to aerosolized cigarette smoke extract for two weeks. Transgenic mice showed significant lung emphysematous changes, increased infiltration of inflammatory cells and enhanced lung concentrations of inflammatory cytokines in the lungs compared to their wild type counterparts after inhalation of cigarette smoke extract. This novel mouse model will be a very useful tool for evaluating the mechanistic pathways and for development of novel therapies in cigarette smoke-associated lung emphysema.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Osamu Taguchi
- Mie University Center for Physical and Mental Health, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu, Mie, 514-8507, Japan
| | | |
Collapse
|
70
|
Gu W, Yuan Y, Yang H, Wu H, Wang L, Tang Z, Li Q. Role of miR-195 in cigarette smoke-induced chronic obstructive pulmonary disease. Int Immunopharmacol 2017; 55:49-54. [PMID: 29223853 DOI: 10.1016/j.intimp.2017.11.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 01/03/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is regarded as a persistent respiratory symptom, mainly caused by cigarette smoking. Recent data have suggested that some miRNAs are involved in the pathogenesis of COPD. Here, we found that miR-195 was significantly upregulated in the lung tissues of patients with COPD compared to in never smokers. miR-195 expression was also upregulated in cigarette smoke (CS)-exposed mice. Lentivirus-mediated knockdown of miR-195 alleviated CS-induced lung pathological changes and reduced inflammatory cell infiltration as well as production of interleukin-6 and tumor necrosis factor-α in bronchoalveolar lavage fluid. Mechanically, a positive correlation was found between miR-195 and phosphorylation of Akt in lung tissues of COPD patients. PHLPP2 was confirmed as a direct downstream target of miR-195 and negative regulator of miR-195 expression. Inhibition of PHLPP2 enhanced Akt phosphorylation and increased interleukin-6 and tumor necrosis factor-α production in BEAS-2B cells, resembling the effects of miR-195 overexpression. Collectively, our data indicate that miR-195 has a pathogenetic role in CS-induced COPD and regulates Akt signaling by suppressing PHLPP2 expression. miR-195 may be an effective therapeutic target in COPD.
Collapse
Affiliation(s)
- Wenchao Gu
- Department of Respiratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200080, China; Department of Respiratory Medicine, Shanghai Pudong New Area People' s Hospital, Shanghai 201200, China
| | - Yaping Yuan
- Department of Respiratory Medicine, Shanghai Pudong New Area People' s Hospital, Shanghai 201200, China
| | - Hua Yang
- Department of Respiratory Medicine, Shanghai Pudong New Area People' s Hospital, Shanghai 201200, China
| | - Hao Wu
- Department of Respiratory Medicine, Shanghai Pudong New Area People' s Hospital, Shanghai 201200, China
| | - Linxuan Wang
- Department of Respiratory Medicine, Shanghai Pudong New Area People' s Hospital, Shanghai 201200, China
| | - Zhijun Tang
- Department of Respiratory Medicine, Shanghai Pudong New Area People' s Hospital, Shanghai 201200, China
| | - Qiang Li
- Department of Respiratory Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200080, China.
| |
Collapse
|
71
|
Elevated GDF-15 contributes to pulmonary inflammation upon cigarette smoke exposure. Mucosal Immunol 2017; 10:1400-1411. [PMID: 28145442 DOI: 10.1038/mi.2017.3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 01/03/2017] [Indexed: 02/04/2023]
Abstract
The molecular mechanisms underlying the pathogenesis of chronic obstructive pulmonary disease (COPD) are still unclear, however signaling pathways associated with lung development, such as the transforming growth factor (TGF)-β superfamily, could be implicated in COPD. Growth differentiation factor (GDF)-15, a member of the TGF-β superfamily, is involved in inflammation, mucus secretion, and cachexia. We analyzed the pulmonary expression of GDF-15 in smokers and patients with COPD, in cigarette smoke (CS)-exposed cultures of primary human bronchial epithelial cells (pHBECs), and in CS-exposed mice. Next, we exposed GDF-15 KO and control mice to air or CS and evaluated pulmonary inflammation. GDF-15 levels were higher in sputum supernatant and lung tissue of patients with COPD and smokers without COPD compared with never smokers. Immunohistochemistry revealed GDF-15 staining in the airway epithelium. Increased expression and secretion of GDF-15 was confirmed in vitro in CS-exposed pHBECs compared with air-exposed pHBECs. Similarly, GDF-15 levels were increased in lungs of CS-exposed mice. Importantly, GDF-15 deficiency attenuated the CS-induced pulmonary inflammation. These results suggest that increased GDF-15-as observed in lungs of smokers and patients with COPD-contributes to CS-induced pulmonary inflammation.
Collapse
|
72
|
Krüger K, Seimetz M, Ringseis R, Wilhelm J, Pichl A, Couturier A, Eder K, Weissmann N, Mooren FC. Exercise training reverses inflammation and muscle wasting after tobacco smoke exposure. Am J Physiol Regul Integr Comp Physiol 2017; 314:R366-R376. [PMID: 29092860 DOI: 10.1152/ajpregu.00316.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Long-term cigarette smoking induces inflammatory processes in the pulmonary system that are suggested to "spill over" into systemic inflammation. Regular exercise has been shown to have anti-inflammatory properties. The aim of the study was to investigate the effects of therapeutic exercise on inflammation and muscle wasting in smoke-exposed mice. C57BL/6J mice ( n = 30) were separated into three groups to receive either 1) no specific treatment (control group), 2) 8-mo exposure to cigarette smoke [smoke-exposed (SE) group], or 3) 8 mo of cigarette smoke combined with exercise training during the last 2 mo (SEex group). The inflammatory status was analyzed by quantifying levels of various plasma proteins using multiplex ELISA and detection of lymphocyte surface markers by flow cytometry. Muscle tissue was analyzed by histological techniques and measurements of RNA/protein expression. SE led to decreased maximal O2 uptake (V̇o2max) and maximal running speed ( Vmax), which was reversed by exercise ( P < 0.05). Expression of ICAM-1, VCAM-1, and CD62L on T cells increased and was reversed by exercise ( P < 0.05). Similarly, SE induced an increase of various inflammatory cytokines, which were downregulated by exercise. In muscle, exercise improved the structure, oxidative capacity, and metabolism by reducing ubiquitin proteasome system activation, stimulating insulin-like growth factor 1 expression, and the SE-induced inhibition of mammalian target of rapamycin signaling pathway ( P < 0.05). Exercise training reverses smoke-induced decline in exercise capacity, systemic inflammation, and muscle wasting by addressing immune-regulating, anabolic, and metabolic pathways.
Collapse
Affiliation(s)
- Karsten Krüger
- Institute of Sports Science, Department Exercise and Health, Leibniz University Hannover , Germany.,Department of Sports Medicine, University of Giessen , Giessen , Germany
| | - Michael Seimetz
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Cente, Member of the German Center for Lung Research
| | - Robert Ringseis
- Institute of Animal Nutrition and Nutritional Physiology, Justus-Liebig-University, Heinrich-Buff-Ring, Giessen , Germany
| | - Jochen Wilhelm
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Cente, Member of the German Center for Lung Research
| | - Alexandra Pichl
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Cente, Member of the German Center for Lung Research
| | - Aline Couturier
- Institute of Animal Nutrition and Nutritional Physiology, Justus-Liebig-University, Heinrich-Buff-Ring, Giessen , Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutritional Physiology, Justus-Liebig-University, Heinrich-Buff-Ring, Giessen , Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Cente, Member of the German Center for Lung Research
| | - Frank C Mooren
- Department of Sports Medicine, University of Giessen , Giessen , Germany.,Klinik Königsfeld, Ennepetal, Germany
| |
Collapse
|
73
|
Jia R, Zhang H, Yang Z, Zhao H, Liu F, Wang H, Miao M, Wang Q, Liu Y. Protective effects of Schisandrin B on cigarette smoke-induced airway injury in mice through Nrf2 pathway. Int Immunopharmacol 2017; 53:11-16. [PMID: 29031142 DOI: 10.1016/j.intimp.2017.09.030] [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: 07/15/2017] [Revised: 09/25/2017] [Accepted: 09/28/2017] [Indexed: 12/18/2022]
Abstract
Schisandrin B (SchB), a dibenzocyclooctadiene derivative isolated from Schisandra chinensis, has been reported to have anti-inflammatory effects. However, the protective effects of SchB on cigarette smoke (CS)-induced lung inflammation remain unclear. This study was to investigate the effects of SchB on CS-induced lung inflammation in mice. The mice were exposed to CS to develop lung inflammation. SchB was given 1h before CS exposure daily for five consecutive days. The levels of inflammatory mediators TNF-α, IL-1β, and IL-6 in bronchoalveolar lavage fluid (BALF) were measured in this study. SOD, GSH, MPO and MDA contents were also detected. Furthermore, the expression of Nrf-2 and NF-κB were detected by western blot analysis. Histopathological analyses showed that SchB had protective effects against CS-induced lung inflammation. The levels of inflammatory mediators TNF-α, IL-1β, and IL-6 in BALF were also inhibited by SchB. CS-induced MPO activity and MDA content were inhibited by SchB. The levels of SOD and GSH were up-regulated by SchB. SchB significantly inhibited CS-induced NF-κB activation and up-regulated the expression of Nrf2 and HO-1. In conclusion, these data suggest that SchB protects against CS-induced lung inflammation by activating Nrf2 and inhibiting NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ruichun Jia
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Haogang Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zhiping Yang
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Hong Zhao
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Fei Liu
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Hui Wang
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Meijuan Miao
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Qiushi Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yanhong Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
74
|
microRNA profiling in lung tissue and bronchoalveolar lavage of cigarette smoke-exposed mice and in COPD patients: a translational approach. Sci Rep 2017; 7:12871. [PMID: 28993685 PMCID: PMC5634489 DOI: 10.1038/s41598-017-13265-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/20/2017] [Indexed: 01/01/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by a progressive airflow limitation and is associated with a chronic inflammatory response in both airways and lungs. microRNAs (miRNAs) are often highly conserved between species and have an intricate role within homeostatic conditions and immune responses. Also, miRNAs are dysregulated in smoking-associated diseases. We investigated the miRNA profile of 523 miRNAs by stem-loop RT-qPCR in lung tissue and cell-free bronchoalveolar lavage (BAL) supernatant of mice exposed to air or cigarette smoke (CS) for 4 or 24 weeks. After 24 weeks of CS exposure, 31 miRNAs were differentially expressed in lung tissue and 78 in BAL supernatant. Next, we correlated the miRNA profiling data to inflammation in BAL and lung, obtained by flow cytometry or ELISA. In addition, we surveyed for overlap with newly assessed miRNA profiles in bronchial biopsies and with previously assessed miRNA profiles in lung tissue and induced sputum supernatant of smokers with COPD. Several miRNAs showed concordant differential expression between both species including miR-31*, miR-155, miR-218 and let-7c. Thus, investigating miRNA profiling data in different compartments and both species provided accumulating insights in miRNAs that may be relevant in CS-induced inflammation and the pathogenesis of COPD.
Collapse
|
75
|
Oliveira da Silva C, Monte-Alto-Costa A, Renovato-Martins M, Viana Nascimento FJ, Dos Santos Valença S, Lagente V, Pôrto LC, Victoni T. Time Course of the Phenotype of Blood and Bone Marrow Monocytes and Macrophages in the Lung after Cigarette Smoke Exposure In Vivo. Int J Mol Sci 2017; 18:ijms18091940. [PMID: 28891938 PMCID: PMC5618589 DOI: 10.3390/ijms18091940] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/30/2017] [Accepted: 09/04/2017] [Indexed: 12/20/2022] Open
Abstract
Alveolar macrophages play a central role in the pathogenesis of chronic obstructive pulmonary disease (COPD). Monocytes are recruited from blood during inflammation and then mature into alveolar macrophages. The aim of this study was to investigate the effect of cigarette smoke (CS) at different times in lung macrophages and monocytes from blood and bone marrow in mice. Male mice (C57BL/6, n = 45) were divided into groups: control, CS 5 days, CS 14 days and CS 30 days. Five days’ CS exposure induced a pronounced influx of neutrophils and macrophages in the lung associated with increased levels of keratinocyte chemoattractant (KC), tumor necrosis factor-α (TNF-α), nitric oxide (NO) and matrix metalloproteinase (MMP)-12. After 14 days of CS exposure, neutrophil recruitment and cytokine production were greatly reduced. Moreover, chronic CS exposure led to increased recruitment of macrophages (with high expression of CD206), transforming growth factor-β (TGF-β) production as well as no detection of TNF-α, interleukin (IL)-6 and KC. CS can also change the monocyte phenotype in the blood and bone marrow, with an increase in Ly6Clow cells. These results show for the first time that CS can change not only macrophage polarization but also monocyte. These results suggest that continued recruitment of Ly6Clow monocytes may help the distinct renewing macrophage M2 population required for COPD progression.
Collapse
Affiliation(s)
- Camila Oliveira da Silva
- Laboratório e Histocompatibilidade e Criopreservação, HLA/Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20950-000, Brazil.
| | - Andréa Monte-Alto-Costa
- Laboratório de Reparo Tecidual, DHE/IBRAG/Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20950-003, Brazil.
| | - Mariana Renovato-Martins
- Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20551-030, Brazil.
| | - Filipe Jorge Viana Nascimento
- Laboratório e Histocompatibilidade e Criopreservação, HLA/Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20950-000, Brazil.
| | - Samuel Dos Santos Valença
- Laboratório de Biologia Redox, ICB/Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| | - Vincent Lagente
- Nutrition, Métabolismes et Cancer, NUMECAN Unité, Institut national de la santé et de la recherche médicale, INSERM 1241/Institut national de la recherche agronomique, INRA 1341/Université de Rennes 1, 35000 Rennes, France.
| | - Luís Cristóvão Pôrto
- Laboratório e Histocompatibilidade e Criopreservação, HLA/Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20950-000, Brazil.
| | - Tatiana Victoni
- Laboratório e Histocompatibilidade e Criopreservação, HLA/Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20950-000, Brazil.
- Nutrition, Métabolismes et Cancer, NUMECAN Unité, Institut national de la santé et de la recherche médicale, INSERM 1241/Institut national de la recherche agronomique, INRA 1341/Université de Rennes 1, 35000 Rennes, France.
| |
Collapse
|
76
|
Toledo-Arruda AC, Vieira RP, Guarnier FA, Suehiro CL, Caleman-Neto A, Olivo CR, Arantes PMM, Almeida FM, Lopes FDTQS, Ramos EMC, Cecchini R, Lin CJ, Martins MA. Time-course effects of aerobic physical training in the prevention of cigarette smoke-induced COPD. J Appl Physiol (1985) 2017; 123:674-683. [PMID: 28729393 DOI: 10.1152/japplphysiol.00819.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 07/13/2017] [Accepted: 07/13/2017] [Indexed: 11/22/2022] Open
Abstract
A previous study by our group showed that regular exercise training (ET) attenuated pulmonary injury in an experimental model of chronic exposure to cigarette smoke (CS) in mice, but the time-course effects of the mechanisms involved in this protection remain poorly understood. We evaluated the temporal effects of regular ET in an experimental model of chronic CS exposure. Male C57BL/6 mice were divided into four groups: Control (sedentary + air), Exercise (aerobic training + air), Smoke (sedentary + smoke), and Smoke + Exercise (aerobic training + smoke). Mice were exposed to CS and ET for 4, 8, or 12 wk. Exercise protected mice exposed to CS from emphysema and reductions in tissue damping and tissue elastance after 12 wk (P < 0.01). The total number of inflammatory cells in the bronchoalveolar lavage increased in the Smoke group, mainly due to the recruitment of macrophages after 4 wk, neutrophils and lymphocytes after 8 wk, and lymphocytes and macrophages after 12 wk (P < 0.01). Exercise attenuated this increase in mice exposed to CS. The protection conferred by exercise was mainly observed after exercise adaptation. Exercise increased IL-6 and IL-10 in the quadriceps and lungs (P < 0.05) after 12 wk. Total antioxidant capacity and SOD was increased and TNF-α and oxidants decreased in lungs of mice exposed to CS after 12 wk (P < 0.05). The protective effects of exercise against lung injury induced by cigarette smoke exposure suggests that anti-inflammatory mediators and antioxidant enzymes play important roles in chronic obstructive pulmonary disease development mainly after the exercise adaptation.NEW & NOTEWORTHY These experiments investigated for the first time the temporal effects of regular moderate exercise training in cigarette smoke-induced chronic obstructive pulmonary disease. We demonstrate that aerobic conditioning had a protective effect in emphysema development induced by cigarette smoke exposure. This effect was most likely secondary to an effect of exercise on oxidant-antioxidant balance and anti-inflammatory mediators.
Collapse
Affiliation(s)
| | - Rodolfo P Vieira
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology, School of Medical Sciences Humanitas, Universidade Brasil and Laboratory of Pulmonary and Exercise Immunology, Nove de Julho University, Sao Paulo, Brazil
| | - Flávia A Guarnier
- Department of Pathology, Londrina State University, Londrina, Brazil; and
| | - Camila L Suehiro
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Agostinho Caleman-Neto
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Clarice R Olivo
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Petra M M Arantes
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Francine M Almeida
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Fernanda D T Q S Lopes
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Ercy M C Ramos
- Department of Physiotherapy, State University of Sao Paulo, Presidente Prudente, Brazil
| | - Rubens Cecchini
- Department of Pathology, Londrina State University, Londrina, Brazil; and
| | - Chin Jia Lin
- Department of Pathology, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Milton Arruda Martins
- Department of Clinical Medicine, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
77
|
Effect of IRAK-M on Airway Inflammation Induced by Cigarette Smoking. Mediators Inflamm 2017; 2017:6506953. [PMID: 28951634 PMCID: PMC5603328 DOI: 10.1155/2017/6506953] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 05/16/2017] [Accepted: 05/29/2017] [Indexed: 12/27/2022] Open
Abstract
Background IRAK-M, negatively regulating Toll-like receptor, is shown the dual properties in the varied disease contexts. We studied the effect of IRAK-M deficiency on cigarette smoking- (CS-) induced airway inflammation under acute or subacute conditions in a mouse model. Methods A number of cellular and molecular techniques were used to detect the differences between IRAK-M knockout (KO) and wild type (WT) mice exposed to 3-day or 7-week CS. Results Airway inflammation was comparable between IRAK-M KO and WT mice under 3-day CS exposure. Upon short-term CS exposure and lipopolysaccharide (LPS) inhalation, IRAK-M KO mice demonstrated worse airway inflammation, significantly higher percentage of Th17 cells and concentrations of proinflammatory cytokines in the lungs, and significantly elevated expression of costimulatory molecules CD40 and CD86 by lung dendritic cells (DCs) or macrophages. Conversely, 7-week CS exposed IRAK-M KO mice demonstrated significantly attenuated airway inflammation, significantly lower concentrations of proinflammatory cytokines in the lungs, significantly increased percentage of Tregs, and lower expression of CD11b and CD86 by lung DCs or macrophages. Conclusions IRAK-M plays distinctive effect on CS-induced airway inflammation, and influences Treg/Th17 balance and expression of costimulatory molecules by DCs and macrophages, depending on duration and intensity of stimulus.
Collapse
|
78
|
Du Y, Ding Y, Chen X, Mei Z, Ding H, Wu Y, Jie Z. MicroRNA-181c inhibits cigarette smoke-induced chronic obstructive pulmonary disease by regulating CCN1 expression. Respir Res 2017; 18:155. [PMID: 28806967 PMCID: PMC5557525 DOI: 10.1186/s12931-017-0639-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/07/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is an obstinate pulmonary disease, causing irreversible alveoli collapse and increasing the risk for cardiovascular disease. Accumulating evidence has shown that the dysregulation of miRNAs is crucially involved in the pathogenesis and development of COPD. However, the effects and role of microRNA-181c (miR-181c) have not been investigated in a murine model of COPD. METHODS miR-181c expression was detected in human lung tissue samples of 34 patients, an in vivo murine model of CS exposure, and primary human bronchial epithelial cells (HBECs) by qRT-PCR. Degeneration of lung tissue, necrosis, infiltration and neutrophil cells were assessed with H&E and flow cytometry. Interleukin (IL)-6 and IL-8 levels were determined by an enzyme-linked immunosorbent assay and qRT-PCR. Luciferase reporter assay and correlation analyses were used to confirm and measure the levels between miR-181c and its target CCN1. RESULTS We showed that miR-181c was significantly down-regulated in lung tissues from patients with COPD compared to individuals who had never smoked (p < 0.01). We also observed a down-regulation of miR-181c in HBECs and a mouse model after cigarette smoke (CS) exposure. Functional assays demonstrated that miR-181c over-expression decreased the inflammatory response, neutrophil infiltration, reactive oxygen species (ROS) generation, and inflammatory cytokines induced by CS, while its down-regulation produced the opposite effects. Subsequent investigation found that CCN1 was a direct target of miR-181c. CCN1 expression was increased in lung tissues of COPD patients, and was negatively correlated with miR-181c expression in human COPD samples (p < 0.01). CONCLUSIONS Taken together, our data suggest the critical roles of miR-181c and its target CCN1 in COPD development, and provide potential therapeutic targets for COPD treatment.
Collapse
Affiliation(s)
- Yong Du
- Department of Respiratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, No.801 Heqing Road, MinhangDistrict, Shanghai, 200240, China
| | - Yi Ding
- Department of Respiratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, No.801 Heqing Road, MinhangDistrict, Shanghai, 200240, China
| | - Xuru Chen
- Department of Respiratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, No.801 Heqing Road, MinhangDistrict, Shanghai, 200240, China
| | - Zhoufang Mei
- Department of Respiratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, No.801 Heqing Road, MinhangDistrict, Shanghai, 200240, China
| | - Heyuan Ding
- Department of Endocrinology, The Fifth People's Hospital of Shanghai, Fudan University, No.801 Heqing Road, MinhangDistrict, Shanghai, 200240, China
| | - Yi Wu
- Department of Respiratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, No.801 Heqing Road, MinhangDistrict, Shanghai, 200240, China
| | - Zhijun Jie
- Department of Respiratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, No.801 Heqing Road, MinhangDistrict, Shanghai, 200240, China.
| |
Collapse
|
79
|
Bowen TS, Aakerøy L, Eisenkolb S, Kunth P, Bakkerud F, Wohlwend M, Ormbostad AM, Fischer T, Wisloff U, Schuler G, Steinshamn S, Adams V, Bronstad E. Exercise Training Reverses Extrapulmonary Impairments in Smoke-exposed Mice. Med Sci Sports Exerc 2017; 49:879-887. [PMID: 28009790 DOI: 10.1249/mss.0000000000001195] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Cigarette smoking is the main risk factor for chronic obstructive pulmonary disease and emphysema. However, evidence on the extrapulmonary effects of smoke exposure that precede lung impairments remains unclear at present, as are data on nonpharmacological treatments such as exercise training. METHODS Three groups of mice, including control (n = 10), smoking (n = 10), and smoking with 6 wk of high-intensity interval treadmill running (n = 11), were exposed to 20 wk of fresh air or whole-body cigarette smoke. Exercise capacity (peak oxygen uptake) and lung destruction (histology) were subsequently measured, whereas the heart, peripheral endothelium (aorta), and respiratory (diaphragm) and limb (extensor digitorum longus and soleus) skeletal muscles were assessed for in vivo and in vitro function, in situ mitochondrial respiration, and molecular alterations. RESULTS Smoking reduced body weight by 26% (P < 0.05) without overt airway destruction (P > 0.05). Smoking impaired exercise capacity by 15% while inducing right ventricular dysfunction by ~20%, endothelial dysfunction by ~20%, and diaphragm muscle weakness by ~15% (all P < 0.05), but these were either attenuated or reversed by exercise training (P < 0.05). Compared with controls, smoking mice had normal limb muscle and mitochondrial function (cardiac and skeletal muscle fibers); however, diaphragm measures of oxidative stress and protein degradation were increased by 111% and 65%, respectively (P < 0.05), but these were attenuated by exercise training (P < 0.05). CONCLUSIONS Prolonged cigarette smoking reduced exercise capacity concomitant with functional impairments to the heart, peripheral endothelium, and respiratory muscle that preceded the development of overt emphysema. However, high-intensity exercise training was able to reverse these smoke-induced extrapulmonary impairments.
Collapse
Affiliation(s)
- T Scott Bowen
- 1Department of Internal Medicine and Cardiology, Leipzig University-Heart Center, Leipzig, GERMANY; 2Faculty of Medicine, Department of Circulation and Medical Imaging, K.G. Jebsen Center of Exercise in Medicine, Norwegian University of Science and Technology, Trondheim, NORWAY; and 3Department of Thoracic Medicine, Clinic of Thoracic and Occupational Medicine, St. Olav's University Hospital, Trondheim, NORWAY
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Mulligan JK, O’Connell BP, Pasquini W, Mulligan RM, Smith S, Soler ZM, Atkinson C, Schlosser RJ. Impact of tobacco smoke on upper airway dendritic cell accumulation and regulation by sinonasal epithelial cells. Int Forum Allergy Rhinol 2017; 7:777-785. [PMID: 28574651 PMCID: PMC5544557 DOI: 10.1002/alr.21955] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/10/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND In these studies we examined the impact of environmental tobacco smoke (ETS) and active smoking on sinonasal dendritic cell (DC) subsets in controls or patients with chronic rhinosinusitis with nasal polyps (CRSwNP). In subsequent in-vitro investigations, we examined the influence of cigarette smoke extract (CSE) on human sinonasal epithelial cells' (HSNECs) ability to regulate DC functions. METHODS Sinonasal tissue, blood, and hair were collected from patients undergoing sinus surgery. Smoking status and ETS exposure were determined by hair nicotine. DC subsets were examined by flow cytometric analysis. Monocyte-derived dendritic cells (moDCs) were treated with conditioned medium from non-smoked-exposed HSNECs (NS-HSNECs) or cigarette-smoke-extract-exposed HSNECs (CSE-HSNECs) to assess the impact of CSE exposure on HSNEC regulation of moDC functions. RESULTS Control subjects who were active smokers displayed increased sinonasal moDC and myeloid dendritic 1 (mDC1) cells and reduced mDC2 cells, whereas, in CRSwNP patients, only moDC and mDC2 cells were altered. ETS was found to increase only moDCs in the CRSwNP patients. In vitro, CSE stimulated HSNEC secretion of the moDC regulatory products chemokine (C-C motif) ligand 20, prostaglandin E2 , and granulocyte-macrophage colony-stimulating factor. CSE exposure also promoted HSNECs to stimulate monocyte and moDC migration. moDCs treated with CSE-HSNEC media stimulated an increase in antigen uptake and expression of CD80 and CD86. Last, CSE-HSNEC-treated moDCs secreted increased levels of interleukin-10, interferon-γ, and thymic stromal lymphopoietin. CONCLUSION Active smoking, and to a lesser degree ETS, alters the sinonasal composition of DCs. A potential mechanism to account for this is that cigarette smoke stimulates HSNECs to induce moDC migration, maturation, and activation.
Collapse
Affiliation(s)
- Jennifer K. Mulligan
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Brendan P. O’Connell
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Whitney Pasquini
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Ryan M. Mulligan
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Sarah Smith
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Zachary M. Soler
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Carl Atkinson
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Rodney J. Schlosser
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina
| |
Collapse
|
81
|
Conickx G, Mestdagh P, Avila Cobos F, Verhamme FM, Maes T, Vanaudenaerde BM, Seys LJM, Lahousse L, Kim RY, Hsu AC, Wark PA, Hansbro PM, Joos GF, Vandesompele J, Bracke KR, Brusselle GG. MicroRNA Profiling Reveals a Role for MicroRNA-218-5p in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2017; 195:43-56. [PMID: 27409149 DOI: 10.1164/rccm.201506-1182oc] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
RATIONALE Aberrant expression of microRNAs (miRNAs) can have a detrimental role in disease pathogenesis. OBJECTIVES To identify dysregulated miRNAs in lung tissue of patients with chronic obstructive pulmonary disease (COPD). METHODS We performed miRNA and mRNA profiling using high throughput stem-loop reverse-transcriptase quantitative polymerase chain reaction and mRNA microarray, respectively, on lung tissue of 30 patients (screening cohort) encompassing 8 never-smokers, 10 smokers without airflow limitation, and 12 smokers with COPD. Differential expression of miRNA-218-5p (miR-218-5p) was validated by reverse-transcriptase quantitative polymerase chain reaction in an independent cohort of 71 patients, an in vivo murine model of COPD, and primary human bronchial epithelial cells. Localization of miR-218-5p was assessed by in situ hybridization. In vitro and in vivo perturbation of miR-218-5p combined with RNA sequencing and gene set enrichment analysis was used to elucidate its functional role in COPD pathogenesis. MEASUREMENTS AND MAIN RESULTS Several miRNAs were differentially expressed among the different patient groups. Interestingly, miR-218-5p was significantly down-regulated in smokers without airflow limitation and in patients with COPD compared with never-smokers. Decreased pulmonary expression of miR-218-5p was validated in an independent validation cohort, in cigarette smoke-exposed mice, and in human bronchial epithelial cells. Importantly, expression of miR-218-5p strongly correlated with airway obstruction. Furthermore, cellular localization of miR-218-5p in human and murine lung revealed highest expression of miR-218-5p in the bronchial airway epithelium. Perturbation experiments with a miR-218-5p mimic or inhibitor demonstrated a protective role of miR-218-5p in cigarette smoke-induced inflammation and COPD. CONCLUSIONS We highlight a role for miR-218-5p in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Griet Conickx
- 1 Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Pieter Mestdagh
- 2 Center for Medical Genetics, Ghent University, Ghent, Belgium
| | | | - Fien M Verhamme
- 1 Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Tania Maes
- 1 Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Bart M Vanaudenaerde
- 3 Department of Clinical and Experimental Medicine, Laboratory for Respiratory Diseases, Lung Transplantation Unit, KU Leuven-University of Leuven, Leuven, Belgium
| | - Leen J M Seys
- 1 Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Lies Lahousse
- 1 Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Richard Y Kim
- 4 Priority Research Centres for Asthma and Respiratory Diseases and Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia; and
| | - Alan C Hsu
- 4 Priority Research Centres for Asthma and Respiratory Diseases and Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia; and
| | - Peter A Wark
- 4 Priority Research Centres for Asthma and Respiratory Diseases and Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia; and.,5 Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- 4 Priority Research Centres for Asthma and Respiratory Diseases and Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia; and
| | - Guy F Joos
- 1 Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Jo Vandesompele
- 2 Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Ken R Bracke
- 1 Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Guy G Brusselle
- 1 Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
82
|
Lewis BW, Sultana R, Sharma R, Noël A, Langohr I, Patial S, Penn AL, Saini Y. Early Postnatal Secondhand Smoke Exposure Disrupts Bacterial Clearance and Abolishes Immune Responses in Muco-Obstructive Lung Disease. THE JOURNAL OF IMMUNOLOGY 2017; 199:1170-1183. [PMID: 28667160 DOI: 10.4049/jimmunol.1700144] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/05/2017] [Indexed: 01/15/2023]
Abstract
Secondhand smoke (SHS) exposure has been linked to the worsening of ongoing lung diseases. However, whether SHS exposure affects the manifestation and natural history of imminent pediatric muco-obstructive airway diseases such as cystic fibrosis remains unclear. To address these questions, we exposed Scnn1b transgenic (Scnn1b-Tg+) mice to SHS from postnatal day (PND) 3-21 and lung phenotypes were examined at PND22. Although a majority of filtered air (FA)-exposed Scnn1b-Tg+ (FA-Tg+) mice successfully cleared spontaneous bacterial infections by PND22, the SHS-exposed Scnn1b-Tg+ (SHS-Tg+) mice failed to resolve these infections. This defect was associated with suppressed antibacterial defenses, i.e., phagocyte recruitment, IgA secretion, and Muc5b expression. Whereas the FA-Tg+ mice exhibited marked mucus obstruction and Th2 responses, SHS-Tg+ mice displayed a dramatic suppression of these responses. Mechanistically, downregulated expression of IL-33, a stimulator of type II innate lymphoid cells, in lung epithelial cells was associated with suppression of neutrophil recruitment, IgA secretions, Th2 responses, and delayed bacterial clearance in SHS-Tg+ mice. Cessation of SHS exposure for 21 d restored previously suppressed responses, including phagocyte recruitment, IgA secretion, and mucous cell metaplasia. However, in contrast with FA-Tg+ mice, the SHS-Tg+ mice had pronounced epithelial necrosis, alveolar space consolidation, and lymphoid hyperplasia; indicating lagged unfavorable effects of early postnatal SHS exposure in later life. Collectively, our data show that early postnatal SHS exposure reversibly suppresses IL-33 levels in airspaces which, in turn, results in reduced neutrophil recruitment and diminished Th2 response. Our data indicate that household smoking may predispose neonates with muco-obstructive lung disease to bacterial exacerbations.
Collapse
Affiliation(s)
- Brandon W Lewis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Razia Sultana
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Rahul Sharma
- National Hansen's Disease Program, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803; and
| | - Alexandra Noël
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Ingeborg Langohr
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803.,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Arthur L Penn
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803;
| |
Collapse
|
83
|
Huang D, Ma Z, He Y, Xiao Y, Luo H, Liang Q, Zhong X, Bai J, He Z. Long-term cigarette smoke exposure inhibits histone deacetylase 2 expression and enhances the nuclear factor-κB activation in skeletal muscle of mice. Oncotarget 2017; 8:56726-56736. [PMID: 28915625 PMCID: PMC5593596 DOI: 10.18632/oncotarget.18089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/27/2017] [Indexed: 01/21/2023] Open
Abstract
Long-term cigarette smoke induces lung inflammatory injury and chronic obstructive pulmonary disease (COPD), associated with skeletal muscle inflammation. This study aimed at investigating how cigarette smoke promotes skeletal muscle inflammation and its molecular pathogenesis. Mice were exposed to air or cigarette smoke for 12 or 24 weeks, and C2C12 cells were stimulated with cigarette smoke extract (CSE). The mass and function, myotube formation, inflammatory cytokine production, histone deacetylase 2 (HDAC2) and nuclear factor-κB (NF-κB) p65 expression were detected in the gastrocnemius muscles of mice and C2C12 cells. In comparison with the control mice, cigarette smoke significantly damaged the lung and reduced the gastrocnemius muscle mass and body weights in mice. Cigarette smoke significantly down-regulated myosin heavy chain (MHC)-IIβ and HDAC2 expression, but enhanced NF-κBp65, keratinocyte chemoattractant (KC) and tumor necrosis factor (TNF)-α expression in the gastrocnemius muscles. CSE stimulation significantly inhibited the myotube formation, MyoD and HDAC2 expression, but enhanced NF-κBp65 expression, KC and TNF-α production in C2C12 cells, which were enhanced by HDAC2 knockdown and abrogated by a NF-κB inhibitor. CSE significantly inhibited the interaction of HDAC2 with NF-κBp65, and increased the levels of acetyl-NF-κBp65 in C2C12 cells. These data indicated that cigarette smoke inhibited HDAC2 expression and its interaction with NF-κBp65 to stimulate inflammation, contributing to the pathogenesis of COPD-related skeletal muscle atrophy in mice.
Collapse
Affiliation(s)
- Dongmei Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zhiying Ma
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yili He
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ying Xiao
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Honglin Luo
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Qiuli Liang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiaoning Zhong
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jing Bai
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zhiyi He
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| |
Collapse
|
84
|
Zou Y, Chen X, Liu J, Zhou DB, Kuang X, Xiao J, Yu Q, Lu X, Li W, Xie B, Chen Q. Serum IL-1β and IL-17 levels in patients with COPD: associations with clinical parameters. Int J Chron Obstruct Pulmon Dis 2017; 12:1247-1254. [PMID: 28490868 PMCID: PMC5413485 DOI: 10.2147/copd.s131877] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
COPD is a chronic airway inflammatory disease characterized mainly by neutrophil airway infiltrations. Interleukin (IL)-1β and IL-17 are the key mediators of neutrophilic airway inflammation in COPD. This study was undertaken to evaluate the serum IL-1β and IL-17 levels and associations between these two key mediators with clinical parameters in COPD patients. Serum samples were collected from 60 COPD subjects during the acute exacerbation of COPD, 60 subjects with stable COPD and 40 healthy control subjects. Commercial enzyme-linked immunosorbent assay kits were used to measure the serum IL-1β and IL-17 concentrations. The association between serum IL-1β and IL-17 with FEV1% predicted, C-reactive protein, neutrophil percentage and smoking status (pack-years) was assessed in the COPD patients. We found that serum IL-1β and IL-17 levels in acute exacerbation of COPD subjects were significantly higher than that in stable COPD or control subjects and were positively correlated to serum C-reactive protein levels, neutrophil % and smoking status (pack-years) but negatively correlated with FEV1% predicted in COPD patients. More importantly, serum IL-1β levels were markedly positively associated with serum IL-17 levels in patients with COPD (P=0.741, P<0.001). In conclusion, elevated serum IL-1β and IL-17 levels may be used as a biomarker for indicating persistent neutrophilic airway inflammation and potential ongoing exacerbation of COPD.
Collapse
Affiliation(s)
- Yong Zou
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Xi Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Jie Liu
- Department of Emergency, The First People's Hospital of Changsha, Changsha, People's Republic of China
| | - Dong Bo Zhou
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Xiao Kuang
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Jian Xiao
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Qiao Yu
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Xiaoxiao Lu
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Wei Li
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Bin Xie
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| | - Qiong Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University
| |
Collapse
|
85
|
Duan MC, Zhang JQ, Liang Y, Liu GN, Xiao J, Tang HJ, Liang Y. Infiltration of IL-17-Producing T Cells and Treg Cells in a Mouse Model of Smoke-Induced Emphysema. Inflammation 2017; 39:1334-44. [PMID: 27150336 DOI: 10.1007/s10753-016-0365-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive and irreversible chronic inflammatory disease associated with the accumulation of activated T cells. To date, there is little information concerning the intrinsic association among Th17, Tc17, and regulatory T (Treg) cells in COPD. The objective of this study was to investigate the variation of lungs CD4(+)Foxp3(+) Treg cells and IL-17-producing CD4 and CD8 (Th17 and Tc17) lymphocytes in mice with cigarette-induced emphysema. Groups of mice were exposed to cigarette smoke or room air. At weeks 12 and 24, mice were sacrificed to observe histological changes by HE stain. The frequencies of Th17 (CD4(+)IL-17(+)T), Tc17 (CD8(+)IL-17(+)T), and Treg (CD4(+)Foxp3(+)T) cells in lungs from these mice were analyzed by flow cytometry. The mRNA levels of orphan nuclear receptor ROR γt and Foxp3 were performed by real-time quantitative polymerase chain reaction. The protein levels of interleukin-17 (IL-17), IL-6, IL-10, and transforming growth factor-beta (TGF-β1) were measured by enzyme-linked immunosorbent assay. Cigarette smoke caused substantial enlargement of the air spaces accompanied by the destruction of the normal alveolar architecture and led to emphysema. The frequencies of Th17 and Tc17 cells, as well as the expressions of IL-6, IL-17, TGF-β1, and ROR γt were greater in the lungs of cigarette smoke (CS)-exposed mice, particularly in the 24-week CS-exposed mice. The frequencies of Treg cells and the expressions of IL-10 and Foxp3 were lower in CS-exposed mice compared to control group. More important, the frequencies of Tregs were negatively correlated with Th17 cells and with Tc17 cells. Interestingly, a significant portion of the cells that infiltrate the lungs was skewed towards a Tc17 phenotype. Our findings suggest the contribution of Th17, Tc17, and Treg cells in the pathogenesis of COPD. Rebalance of these cells will be helpful for developing and refining the new immunological therapies for COPD.
Collapse
Affiliation(s)
- Min-Chao Duan
- Department of Respiratory Medicine, The Eighth People's Hospital of Nanning, Nanning, Guangxi, 530001, China
- Department of Respiratory Medicine, The First Afiltrated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jian-Quan Zhang
- Department of Respiratory Medicine, The First Afiltrated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Yue Liang
- Department of Respiratory Medicine, The Eighth People's Hospital of Nanning, Nanning, Guangxi, 530001, China
| | - Guang-Nan Liu
- Department of Respiratory Medicine, The First Afiltrated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jin Xiao
- Department of Respiratory Medicine, The Eighth People's Hospital of Nanning, Nanning, Guangxi, 530001, China
| | - Hai-Juan Tang
- Department of Respiratory Medicine, The First Afiltrated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yi Liang
- Department of Respiratory Medicine, The First Afiltrated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| |
Collapse
|
86
|
Al-Sawalha NA, Migdadi AM, Alzoubi KH, Khabour OF, Qinna NA. Effect of waterpipe tobacco smoking on airway inflammation in murine model of asthma. Inhal Toxicol 2017; 29:46-52. [PMID: 28330430 DOI: 10.1080/08958378.2017.1280105] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE There has been an increase in the popularity of waterpipe tobacco smoking (WTS) worldwide, especially in the younger population, including asthma patients. In this study, we investigated the effects of waterpipe smoking on airway inflammation, cytokine levels and oxidative stress markers in an antigen-driven murine model of asthma. MATERIALS AND METHODS Balb/c mice were divided into four groups; (1) control (received fresh air, ovalbumin sensitization and saline challenge), (2) WTS (received WTS, ovalbumin sensitization and saline challenge), (3) Ova S/C (received fresh air, ovalbumin sensitization and ovalbumin challenge) and (4) simultaneous WTS and Ova S/C (received WTS, ovalbumin sensitization and ovalbumin challenge). Airway inflammatory cells were evaluated in the broncho-alveolar lavage fluid. Cytokines [interleukin (IL)-13, 10 and 18] and oxidative stress markers [superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx)] were evaluated in the lung homogenates. RESULTS Chronic exposure to WTS significantly increased the number of airway inflammatory cells in mice, specifically: eosinophils, neutrophils, macrophages and lymphocytes. The level of IL-13 in the lungs was increased and the level of IL-10 was reduced (p < 0.05) by WTS. Chronic WTS potentiated the increase in inflammatory cells induced by Ova S/C (p < 0.05). The level of IL-13 in the lungs was increased by simultaneous WTS and Ova S/C (p < 0.05) while, levels of IL-10, IL-18, SOD, catalase and GPx in the lungs were not affected. CONCLUSIONS Chronic WTS exposure induced airway inflammation in control mice and enhanced airway inflammation in murine model of asthma.
Collapse
Affiliation(s)
- Nour A Al-Sawalha
- a Faculty of Pharmacy , Jordan University of Science and Technology , Irbid , Jordan
| | - Ala'a M Migdadi
- a Faculty of Pharmacy , Jordan University of Science and Technology , Irbid , Jordan
| | - Karem H Alzoubi
- a Faculty of Pharmacy , Jordan University of Science and Technology , Irbid , Jordan
| | - Omar F Khabour
- b Faculty of Applied Medical Sciences , Jordan University of Science and Technology , Irbid , Jordan.,c Faculty of Applied Medical Sciences , Taibah University , Medina , Saudi Arabia
| | - Nidal A Qinna
- d Faculty of Pharmacy and Biomedical Sciences , University of Petra , Amman , Jordan
| |
Collapse
|
87
|
Huang F, Cheng H, Zhang YT, Ju YH, Li YN. Early Postnatal Exposure to Cigarette Smoke Leads to Later Airway Inflammation in Asthmatic Mice. PLoS One 2017; 12:e0171021. [PMID: 28135326 PMCID: PMC5279757 DOI: 10.1371/journal.pone.0171021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 01/13/2017] [Indexed: 11/24/2022] Open
Abstract
Background and objective Asthma is one of the most common airway inflammatory diseases. In most cases, asthma development is related to ubiquitous harmful environmental exposure factors in early-life. Previous studies have indicated that smoking can promote asthma development and increase the difficulty of asthma control. The aim of this study was to determine the effects of early-life CS exposure on ovalbumin (OVA)-sensitized asthmatic mice. Methods Pathological and immunological functions were analyzed in an adult asthma mice model in which mice were sensitized with OVA combined with early-life CS exposure. Results Mice exposed to CS for only 5 weeks demonstrated significantly reduced pulmonary compliance, increased airway inflammation, and augmented cellular and humoral immune responses. In addition, CS inhalation was sufficient to facilitate OVA sensitization and challenge asthmatic development. Meanwhile, CS exposure amplified regulatory T cell-mediated immunity inhibition, but still did not offset the increased effector T cell-mediated inflammatory response. Conclusion Early-life CS exposure is significantly associated with later pulmonary injury and aggravation of T-cell immunologic derangement in asthmatic mice.
Collapse
Affiliation(s)
- Fei Huang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Hang Cheng
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Yu-Tong Zhang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Yang-Hua Ju
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Ya-Nan Li
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, PR China.,Department of Molecular Biology, Basic Medical College of Jilin University, Changchun, PR China
| |
Collapse
|
88
|
Białas AJ, Sitarek P, Miłkowska-Dymanowska J, Piotrowski WJ, Górski P. The Role of Mitochondria and Oxidative/Antioxidative Imbalance in Pathobiology of Chronic Obstructive Pulmonary Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7808576. [PMID: 28105251 PMCID: PMC5220474 DOI: 10.1155/2016/7808576] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 10/23/2016] [Indexed: 12/12/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common preventable and treatable disease, characterized by persistent airflow limitation that is usually progressive and associated with an enhanced chronic inflammatory response in the airways and the lung to noxious particles or gases. The major risk factor of COPD, which has been proven in many studies, is the exposure to cigarette smoke. However, it is 15-20% of all smokers who develop COPD. This is why we should recognize the pathobiology of COPD as involving a complex interaction between several factors, including genetic vulnerability. Oxidant-antioxidant imbalance is recognized as one of the significant factors in COPD pathogenesis. Numerous exogenous and endogenous sources of ROS are present in pathobiology of COPD. One of endogenous sources of ROS is mitochondria. Although leakage of electrons from electron transport chain and forming of ROS are the effect of physiological functioning of mitochondria, there are various intra- and extracellular factors which may increase this amount and significantly contribute to oxidative-antioxidative imbalance. With the coexistence with impaired antioxidant defence, all these issues lead to oxidative and carbonyl stress. Both of these states play a significant role in pathobiology of COPD and may account for development of major comorbidities of this disease.
Collapse
Affiliation(s)
- Adam Jerzy Białas
- Department of Pneumology and Allergy, 1st Chair of Internal Medicine, Medical University of Lodz, Łódź, Poland
- Healthy Aging Research Centre (HARC), Medical University of Lodz, Łódź, Poland
| | - Przemysław Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Łódź, Łódź, Poland
| | - Joanna Miłkowska-Dymanowska
- Department of Pneumology and Allergy, 1st Chair of Internal Medicine, Medical University of Lodz, Łódź, Poland
- Healthy Aging Research Centre (HARC), Medical University of Lodz, Łódź, Poland
| | - Wojciech Jerzy Piotrowski
- Department of Pneumology and Allergy, 1st Chair of Internal Medicine, Medical University of Lodz, Łódź, Poland
- Healthy Aging Research Centre (HARC), Medical University of Lodz, Łódź, Poland
| | - Paweł Górski
- Department of Pneumology and Allergy, 1st Chair of Internal Medicine, Medical University of Lodz, Łódź, Poland
- Healthy Aging Research Centre (HARC), Medical University of Lodz, Łódź, Poland
| |
Collapse
|
89
|
Cheng Y, Wang D, Wang B, Li H, Xiong J, Xu S, Chen Q, Tao K, Yang X, Zhu Y, He S. HMGB1 translocation and release mediate cigarette smoke-induced pulmonary inflammation in mice through a TLR4/MyD88-dependent signaling pathway. Mol Biol Cell 2016; 28:201-209. [PMID: 27807045 PMCID: PMC5221624 DOI: 10.1091/mbc.e16-02-0126] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 10/20/2016] [Accepted: 10/26/2016] [Indexed: 02/05/2023] Open
Abstract
Cigarette smoke (CS) increases up-regulation of TLR4-mediated signaling and induces TLR4-dependent inflammation in lungs. CS exposure–induced HMGB1 translocation and release of HMGB1 controls CS-induced inflammatory response. MGB1 induces TLR4-mediated proinflammatory cytokine production and activates NF-κB and JNK/p38 pathways. We performed studies to determine the role of high-mobility group box 1 (HMGB1) in cigarette smoke (CS)–induced pulmonary inflammation. After mice were exposed to five cigarettes four times a day for 3 d, toll-like receptor 4 (TLR4) expression and TLR4-mediated signaling were significantly up-regulated, and HMGB1 had translocated from the nucleus to the cytoplasm in lung epithelial cells and then been released into the extracellular lung space. On CS exposure, inflammatory cell recruitment and proinflammatory cytokine production were significantly increased in lung tissue and bronchoalveolar lavage, and these effects depended on the TLR4 signaling pathway. HMGB1 inhibition decreased the CS-induced inflammatory response, whereas treatment with exogenous HMGB1 aggravated the damage and increased the phosphorylation of JNK, p38, and IκBα in the lungs of wild-type mice but not in TLR4-knockout mice. Blockade of TLR4 action or TLR4 knockout significantly inhibited HMGB1-induced proinflammatory cytokine production in mouse tracheal epithelial (MTE) cells and lung tissues. In addition, a MyD88 deficiency inhibited JNK, p38, and IκBα phosphorylation, and this effect was associated with the suppressed production of TNF-α and IL-1β in MTE cells and lung tissues in response to CS stimulation. Thus HMGB1 activates the NF-κB and JNK/p38 pathways through TLR4/MyD88-dependent signaling and induces an inflammatory response in lungs exposed to CS.
Collapse
Affiliation(s)
- Yao Cheng
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chong-qing 400016, China.,Department of Hepatobiliary Surgery, Second Affiliated Hospital, Chongqing Medical University, Chong-qing 400016, China
| | - Dan Wang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chong-qing 400016, China.,Department of Respiration Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bin Wang
- Institute of Life Science, Chongqing Medical University, Chong-qing 400016, China
| | - Huanan Li
- College of Biomedical Engineering, Chongqing Medical University, Chong-qing 400016, China
| | - Junjie Xiong
- Department of General Surgery, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuyun Xu
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Quan Chen
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chong-qing 400016, China
| | - Kun Tao
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chong-qing 400016, China
| | - Xiaoyan Yang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chong-qing 400016, China
| | - Yu Zhu
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chong-qing 400016, China
| | - Sirong He
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chong-qing 400016, China
| |
Collapse
|
90
|
Jones B, Donovan C, Liu G, Gomez HM, Chimankar V, Harrison CL, Wiegman CH, Adcock IM, Knight DA, Hirota JA, Hansbro PM. Animal models of COPD: What do they tell us? Respirology 2016; 22:21-32. [DOI: 10.1111/resp.12908] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 08/01/2016] [Accepted: 08/02/2016] [Indexed: 01/28/2023]
Affiliation(s)
- Bernadette Jones
- Priority Research Centre for Healthy Lungs; Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia; London UK
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs; Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia; London UK
| | - Gang Liu
- Priority Research Centre for Healthy Lungs; Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia; London UK
| | - Henry M. Gomez
- Priority Research Centre for Healthy Lungs; Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia; London UK
| | - Vrushali Chimankar
- Priority Research Centre for Healthy Lungs; Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia; London UK
| | - Celeste L. Harrison
- Priority Research Centre for Healthy Lungs; Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia; London UK
| | - Cornelis H. Wiegman
- The Airways Disease Section, National Heart and Lung Institute; Imperial College London; London UK
| | - Ian M. Adcock
- The Airways Disease Section, National Heart and Lung Institute; Imperial College London; London UK
| | - Darryl A. Knight
- Priority Research Centre for Healthy Lungs; Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia; London UK
| | - Jeremy A. Hirota
- James Hogg Research Centre; University of British Columbia; Vancouver British Columbia Canada
| | - Philip M. Hansbro
- Priority Research Centre for Healthy Lungs; Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia; London UK
| |
Collapse
|
91
|
Pan Z, Yu H, Liao JL. Probing Cellular and Molecular Mechanisms of Cigarette Smoke-Induced Immune Response in the Progression of Chronic Obstructive Pulmonary Disease Using Multiscale Network Modeling. PLoS One 2016; 11:e0163192. [PMID: 27669518 PMCID: PMC5036797 DOI: 10.1371/journal.pone.0163192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 09/06/2016] [Indexed: 01/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disorder characterized by progressive destruction of lung tissues and airway obstruction. COPD is currently the third leading cause of death worldwide and there is no curative treatment available so far. Cigarette smoke (CS) is the major risk factor for COPD. Yet, only a relatively small percentage of smokers develop the disease, showing that disease susceptibility varies significantly among smokers. As smoking cessation can prevent the disease in some smokers, quitting smoking cannot halt the progression of COPD in others. Despite extensive research efforts, cellular and molecular mechanisms of COPD remain elusive. In particular, the disease susceptibility and smoking cessation effects are poorly understood. To address these issues in this work, we develop a multiscale network model that consists of nodes, which represent molecular mediators, immune cells and lung tissues, and edges describing the interactions between the nodes. Our model study identifies several positive feedback loops and network elements playing a determinant role in the CS-induced immune response and COPD progression. The results are in agreement with clinic and laboratory measurements, offering novel insight into the cellular and molecular mechanisms of COPD. The study in this work also provides a rationale for targeted therapy and personalized medicine for the disease in future.
Collapse
Affiliation(s)
- Zhichao Pan
- Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui Province, 230026, People’s Republic of China
| | - Haishan Yu
- Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui Province, 230026, People’s Republic of China
| | - Jie-Lou Liao
- Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui Province, 230026, People’s Republic of China
| |
Collapse
|
92
|
Jia J, Conlon TM, Ballester Lopez C, Seimetz M, Bednorz M, Zhou-Suckow Z, Weissmann N, Eickelberg O, Mall MA, Yildirim AÖ. Cigarette smoke causes acute airway disease and exacerbates chronic obstructive lung disease in neonatal mice. Am J Physiol Lung Cell Mol Physiol 2016; 311:L602-10. [PMID: 27448665 DOI: 10.1152/ajplung.00124.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/12/2016] [Indexed: 11/22/2022] Open
Abstract
Epidemiological evidence demonstrates a strong link between postnatal cigarette smoke (CS) exposure and increased respiratory morbidity in young children. However, how CS induces early onset airway disease in young children, and how it interacts with endogenous risk factors, remains poorly understood. We, therefore, exposed 10-day-old neonatal wild-type and β-epithelial sodium ion channel (β-ENaC)-transgenic mice with cystic fibrosis-like lung disease to CS for 4 days. Neonatal wild-type mice exposed to CS demonstrated increased numbers of macrophages and neutrophils in the bronchoalveolar lavage fluid (BALF), which was accompanied by increased levels of Mmp12 and Cxcl1 BALF from β-ENaC-transgenic mice contained greater numbers of macrophages, which did not increase following acute CS exposure; however, there was significant increase in airway neutrophilia compared with filtered air transgenic and CS-exposed wild-type controls. Interestingly, wild-type and β-ENaC-transgenic mice demonstrated epithelial airway and vascular remodeling following CS exposure. Morphometric analysis of lung sections revealed that CS exposure caused increased mucus accumulation in the airway lumen of neonatal β-ENaC-transgenic mice compared with wild-type controls, which was accompanied by an increase in the number of goblet cells and Muc5ac upregulation. We conclude that short-term CS exposure 1) induces acute airway disease with airway epithelial and vascular remodeling in neonatal wild-type mice; and 2) exacerbates airway inflammation, mucus hypersecretion, and mucus plugging in neonatal β-ENaC-transgenic mice with chronic lung disease. Our results in neonatal mice suggest that young children may be highly susceptible to develop airway disease in response to tobacco smoke exposure, and that adverse effects may be aggravated in children with underlying chronic lung diseases.
Collapse
Affiliation(s)
- Jie Jia
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Thomas M Conlon
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Carolina Ballester Lopez
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Michael Seimetz
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Justus-Liebig-University, Giessen, Germany; Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Mariola Bednorz
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Justus-Liebig-University, Giessen, Germany; Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Zhe Zhou-Suckow
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany; and
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Justus-Liebig-University, Giessen, Germany; Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Oliver Eickelberg
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany; University Hospital of the Ludwig Maximilians University (LMU), Munich, Germany
| | - Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany; and
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany;
| |
Collapse
|
93
|
Kuang LJ, Deng TT, Wang Q, Qiu SL, Liang Y, He ZY, Zhang JQ, Bai J, Li MH, Deng JM, Liu GN, Liu JF, Zhong XN. Dendritic cells induce Tc1 cell differentiation via the CD40/CD40L pathway in mice after exposure to cigarette smoke. Am J Physiol Lung Cell Mol Physiol 2016; 311:L581-9. [PMID: 27448664 DOI: 10.1152/ajplung.00002.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/20/2016] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells and CD8(+) T cells participate in the pathology of chronic obstructive pulmonary disease, including emphysema, but little is known of the involvement of the CD40/CD40L pathway. We investigated the role of the CD40/CD40L pathway in Tc1 cell differentiation induced by dendritic cells in a mouse model of emphysema, and in vitro. C57BL/6J wild-type and CD40(-/-) mice were exposed to cigarette smoke (CS) or not (control), for 24 wk. In vitro experiments involved wild-type and CD40(-/-) dendritic cells treated with CS extract (CSE) or not. Compared with the control groups, the CS mice (both wild type and CD40(-/-)) had a greater percentage of lung dendritic cells and higher levels of major histocompatability complex (MHC) class I molecules and costimulatory molecules CD40 and CD80. Relative to the CS CD40(-/-) mice, the CS wild type showed greater signs of lung damage and Tc1 cell differentiation. In vitro, the CSE-treated wild-type cells evidenced more cytokine release (IL-12/p70) and Tc1 cell differentiation than did the CSE-treated CD40(-/-) cells. Exposure to cigarette smoke increases the percentage of lung dendritic cells and promotes Tc1 cell differentiation via the CD40/CD40L pathway. Blocking the CD40/CD40L pathway may suppress development of emphysema in mice exposed to cigarette smoke.
Collapse
Affiliation(s)
- Liang-Jian Kuang
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Ting-Ting Deng
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Qin Wang
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Shi-Lin Qiu
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Yi Liang
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Zhi-Yi He
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Jian-Quan Zhang
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Jing Bai
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Mei-Hua Li
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Jing-Min Deng
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Guang-Nan Liu
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| | - Ji-Feng Liu
- Department of Respiratory Medicine, Tenth Affiliated Hospital of Guangxi Medical University, Qinzhou, Guangxi, China
| | - Xiao-Ning Zhong
- Department of Respiratory Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; and
| |
Collapse
|
94
|
Sun D, Ouyang Y, Gu Y, Liu X. Cigarette smoke-induced chronic obstructive pulmonary disease is attenuated by CCL20-blocker: a rat model. Croat Med J 2016; 57:363-70. [PMID: 27586551 PMCID: PMC5048234 DOI: 10.3325/cmj.2016.57.363] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 07/04/2016] [Indexed: 01/06/2023] Open
Abstract
AIM To evaluate whether the effect of dendritic cells (DCs) on chronic obstructive pulmonary disease (COPD) can be relieved by blocking CCL20. METHODS 30 Wistar rats were randomly divided into three groups: control, COPD, and COPD treated with CCL20 monoclonal antibody. In the latter two groups, COPD was induced by four-week cigarette smoke exposure and trachea injection of lipopolysaccharide solution on two occasions. CCL20 monoclonal antibody was injected intraperitoneally on the first day. All animals were sacrificed on the 29th day. Pathomorphology of the lung and bronchiole was analyzed using hematoxylin and eosin staining. The CCR6 content in the bronchoalveolar lavage fluid was detected using ELISA. DC distribution in the lung was examined by immunohistochemistry for OX62. RESULTS COPD rat models showed pathological alterations similar to those in COPD patients. DCs, CCR6, and the severity of emphysema were significantly increased in the COPD group than in controls (all P values <0.001), and they were significantly reduced after anti-CCL20 treatment compared with the COPD group (all P values <0.05). CONCLUSION The interaction between CCR6 and its ligand CCL20 promotes the effect of DCs in the COPD pathogenesis, which can be reduced by blocking CCL20.
Collapse
Affiliation(s)
| | - Yao Ouyang
- Yao Ouyang, Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical College, Dalian Road 149, Zunyi City, Guizhou Province, China,
| | | | | |
Collapse
|
95
|
Kammerl IE, Dann A, Mossina A, Brech D, Lukas C, Vosyka O, Nathan P, Conlon TM, Wagner DE, Overkleeft HS, Prasse A, Rosas IO, Straub T, Krauss-Etschmann S, Königshoff M, Preissler G, Winter H, Lindner M, Hatz R, Behr J, Heinzelmann K, Yildirim AÖ, Noessner E, Eickelberg O, Meiners S. Impairment of Immunoproteasome Function by Cigarette Smoke and in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2016; 193:1230-41. [PMID: 26756824 DOI: 10.1164/rccm.201506-1122oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
RATIONALE Patients with chronic obstructive pulmonary disease (COPD) and in particular smokers are more susceptible to respiratory infections contributing to acute exacerbations of disease. The immunoproteasome is a specialized type of proteasome destined to improve major histocompatibility complex (MHC) class I-mediated antigen presentation for the resolution of intracellular infections. OBJECTIVES To characterize immunoproteasome function in COPD and its regulation by cigarette smoke. METHODS Immunoproteasome expression and activity were determined in bronchoalveolar lavage (BAL) and lungs of human donors and patients with COPD or idiopathic pulmonary fibrosis (IPF), as well as in cigarette smoke-exposed mice. Smoke-mediated alterations of immunoproteasome activity and MHC I surface expression were analyzed in human blood-derived macrophages. Immunoproteasome-specific MHC I antigen presentation was evaluated in spleen and lung immune cells that had been smoke-exposed in vitro or in vivo. MEASUREMENTS AND MAIN RESULTS Immunoproteasome and MHC I mRNA expression was reduced in BAL cells of patients with COPD and in isolated alveolar macrophages of patients with COPD or IPF. Exposure of immune cells to cigarette smoke extract in vitro reduced immunoproteasome activity and impaired immunoproteasome-specific MHC I antigen presentation. In vivo, acute cigarette smoke exposure dynamically regulated immunoproteasome function and MHC I antigen presentation in mouse BAL cells. End-stage COPD lungs showed markedly impaired immunoproteasome activities. CONCLUSIONS We here show that the activity of the immunoproteasome is impaired by cigarette smoke resulting in reduced MHC I antigen presentation. Regulation of immunoproteasome function by cigarette smoke may thus alter adaptive immune responses and add to prolonged infections and exacerbations in COPD and IPF.
Collapse
Affiliation(s)
- Ilona E Kammerl
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Angela Dann
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Alessandra Mossina
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Dorothee Brech
- 2 Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Christina Lukas
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Oliver Vosyka
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Petra Nathan
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Thomas M Conlon
- 3 Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the DZL, Neuherberg, Germany
| | - Darcy E Wagner
- 2 Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Hermen S Overkleeft
- 4 Department of Bio-organic Synthesis, Leiden University, Leiden, the Netherlands
| | - Antje Prasse
- 5 Department of Pneumology, Hannover Medical School, Hannover, Germany
| | - Ivan O Rosas
- 6 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tobias Straub
- 7 Biomedical Center, Bioinformatics Unit, Ludwig-Maximilians University, Munich, Germany
| | - Susanne Krauss-Etschmann
- 8 Division of Experimental Asthma Research, Research Center Borstel, Airway Research Center North, Member of the DZL, Borstel, Germany
- 9 Institute of Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Germany
| | - Melanie Königshoff
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Gerhard Preissler
- 10 Thoraxchirurgisches Zentrum, Klinik für Allgemeine-, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität, Member of the DZL, Munich, Germany
| | - Hauke Winter
- 10 Thoraxchirurgisches Zentrum, Klinik für Allgemeine-, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität, Member of the DZL, Munich, Germany
| | - Michael Lindner
- 11 Asklepios Fachkliniken München-Gauting, Gauting, Germany; and
| | - Rudolf Hatz
- 10 Thoraxchirurgisches Zentrum, Klinik für Allgemeine-, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität, Member of the DZL, Munich, Germany
- 11 Asklepios Fachkliniken München-Gauting, Gauting, Germany; and
| | - Jürgen Behr
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- 11 Asklepios Fachkliniken München-Gauting, Gauting, Germany; and
- 12 Medizinische Klinik und Poliklinik V, Klinikum der Ludwig-Maximilians-Universität, Member of the DZL, Munich, Germany
| | - Katharina Heinzelmann
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Ali Ö Yildirim
- 3 Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the DZL, Neuherberg, Germany
| | - Elfriede Noessner
- 2 Institute of Molecular Immunology, Helmholtz Zentrum München, Munich, Germany
| | - Oliver Eickelberg
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Silke Meiners
- 1 Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
96
|
Kim KH, Park TS, Kim YS, Lee JS, Oh YM, Lee SD, Lee SW. Resolvin D1 prevents smoking-induced emphysema and promotes lung tissue regeneration. Int J Chron Obstruct Pulmon Dis 2016; 11:1119-28. [PMID: 27313451 PMCID: PMC4890694 DOI: 10.2147/copd.s100198] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Emphysema is an irreversible disease that is characterized by destruction of lung tissue as a result of inflammation caused by smoking. Resolvin D1 (RvD1), derived from docosahexaenoic acid, is a novel lipid that resolves inflammation. The present study tested whether RvD1 prevents smoking-induced emphysema and promotes lung tissue regeneration. Materials and methods C57BL/6 mice, 8 weeks of age, were randomly divided into four groups: control, RvD1 only, smoking only, and smoking with RvD1 administration. Four different protocols were used to induce emphysema and administer RvD1: mice were exposed to smoking for 4 weeks with poly(I:C) or to smoking only for 24 weeks, and RvD1 was injected within the smoking exposure period to prevent regeneration or after completion of smoking exposure to assess regeneration. The mean linear intercept and inflammation scores were measured in the lung tissue, and inflammatory cells and cytokines were measured in the bronchoalveolar lavage fluid. Results Measurements of mean linear intercept showed that RvD1 significantly attenuated smoking-induced lung destruction in all emphysema models. RvD1 also reduced smoking-induced inflammatory cell infiltration, which causes the structural derangements observed in emphysema. In the 4-week prevention model, RvD1 reduced the smoking-induced increase in eosinophils and interleukin-6 in the bronchoalveolar lavage fluid. In the 24-week prevention model, RvD1 also reduced the increased neutrophils and total cell counts induced by smoking. Conclusion RvD1 attenuated smoking-induced emphysema in vivo by reducing inflammation and promoting tissue regeneration. This result suggests that RvD1 may be useful in the prevention and treatment of emphysema.
Collapse
Affiliation(s)
- Kang-Hyun Kim
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tai Sun Park
- Department of Pulmonology and Critical Care Medicine, Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Pulmonology and Critical Care Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - You-Sun Kim
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Pulmonology and Critical Care Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae Seung Lee
- Department of Pulmonology and Critical Care Medicine, Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Pulmonology and Critical Care Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yeon-Mok Oh
- Department of Pulmonology and Critical Care Medicine, Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Pulmonology and Critical Care Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Do Lee
- Department of Pulmonology and Critical Care Medicine, Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Pulmonology and Critical Care Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sei Won Lee
- Department of Pulmonology and Critical Care Medicine, Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Pulmonology and Critical Care Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
97
|
Arellano-Orden E, Calero-Acuña C, Moreno-Mata N, Gómez-Izquierdo L, Sánchez-López V, López-Ramírez C, Tobar D, López-Villalobos JL, Gutiérrez C, Blanco-Orozco A, López-Campos JL. Cigarette Smoke Decreases the Maturation of Lung Myeloid Dendritic Cells. PLoS One 2016; 11:e0152737. [PMID: 27058955 PMCID: PMC4825972 DOI: 10.1371/journal.pone.0152737] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 02/29/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Conflicting data exist on the role of pulmonary dendritic cells (DCs) and their maturation in patients with chronic obstructive pulmonary disease (COPD). Herein, we investigated whether disease severity and smoking status could affect the distribution and maturation of DCs in lung tissues of patients undergoing elective pneumectomy or lobectomy for suspected primary lung cancer. MATERIALS AND METHODS A total of 75 consecutive patients were included. Spirometry testing was used to identify COPD. Lung parenchyma sections anatomically distant from the primary lesion were examined. We used flow cytometry to identify different DCs subtypes-including BDCA1-positive myeloid DCs (mDCs), BDCA3-positive mDCs, and plasmacytoid DCs (pDCs)-and determine their maturation markers (CD40, CD80, CD83, and CD86) in all participants. We also identified follicular DCs (fDCs), Langerhans DCs (LDCs), and pDCs in 42 patients by immunohistochemistry. RESULTS COPD was diagnosed in 43 patients (16 current smokers and 27 former smokers), whereas the remaining 32 subjects were classified as non-COPD (11 current smokers, 13 former smokers, and 8 never smokers). The number and maturation of DCs did not differ significantly between COPD and non-COPD patients. However, the results of flow cytometry indicated that maturation markers CD40 and CD83 of BDCA1-positive mDCs were significantly decreased in smokers than in non-smokers (P = 0.023 and 0.013, respectively). Immunohistochemistry also revealed a lower number of LDCs in COPD patients than in non-COPD subjects. CONCLUSIONS Cigarette smoke, rather than airflow limitation, is the main determinant of impaired DCs maturation in the lung.
Collapse
Affiliation(s)
- Elena Arellano-Orden
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
- * E-mail:
| | - Carmen Calero-Acuña
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - Nicolás Moreno-Mata
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Lourdes Gómez-Izquierdo
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - Verónica Sánchez-López
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - Cecilia López-Ramírez
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - Daniela Tobar
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - José Luis López-Villalobos
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Cesar Gutiérrez
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - Ana Blanco-Orozco
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
| | - José Luis López-Campos
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Seville, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
98
|
TAK-242 attenuates acute cigarette smoke-induced pulmonary inflammation in mouse via the TLR4/NF-κB signaling pathway. Biochem Biophys Res Commun 2016; 472:508-15. [PMID: 26944017 DOI: 10.1016/j.bbrc.2016.03.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/01/2016] [Indexed: 11/19/2022]
|
99
|
Khedoe PPSJ, Rensen PCN, Berbée JFP, Hiemstra PS. Murine models of cardiovascular comorbidity in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2016; 310:L1011-27. [PMID: 26993520 DOI: 10.1152/ajplung.00013.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/15/2016] [Indexed: 01/12/2023] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) have an increased risk for cardiovascular disease (CVD). Currently, COPD patients with atherosclerosis (i.e., the most important underlying cause of CVD) receive COPD therapy complemented with standard CVD therapy. This may, however, not be the most optimal treatment. To investigate the link between COPD and atherosclerosis and to develop specific therapeutic strategies for COPD patients with atherosclerosis, a substantial number of preclinical studies using murine models have been performed. In this review, we summarize the currently used murine models of COPD and atherosclerosis, both individually and combined, and discuss the relevance of these models for studying the pathogenesis and development of new treatments for COPD patients with atherosclerosis. Murine and clinical studies have provided complementary information showing a prominent role for systemic inflammation and oxidative stress in the link between COPD and atherosclerosis. These and other studies showed that murine models for COPD and atherosclerosis are useful tools and can provide important insights relevant to understanding the link between COPD and CVD. More importantly, murine studies provide good platforms for studying the potential of promising (new) therapeutic strategies for COPD patients with CVD.
Collapse
Affiliation(s)
- P Padmini S J Khedoe
- Department of Pulmonology, Leiden University Medical Center, the Netherlands; Department of Medicine, Division of Endocrinology, Leiden University Medical Center, the Netherlands; and
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, the Netherlands; and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands
| | - Jimmy F P Berbée
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, the Netherlands; and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, the Netherlands
| |
Collapse
|
100
|
Carpagnano GE, Ruggieri C, Scioscia G, Storto MML, Zoppo L, Foschino-Barbaro MP. Is the Exhaled Breath Temperature Sensitive to Cigarette Smoking? COPD 2016; 13:642-6. [PMID: 26934668 DOI: 10.3109/15412555.2016.1143458] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The smoking habit is accompanied by an acute inflammatory response which follows tissue injury. It would be desirable to find a non-invasive inflammatory marker that would simplify the task of studying and monitoring smokers more simply and allow us to identify populations at risk of contracting Chronic Obstructive Pulmonary Disease (COPD). Today's expectations regarding research focus on issues ranging from inflammatory markers to those of exhaled breath temperature (EBT) are considerable. That said, although the EBT has been largely studied in asthma and COPD, there have not been any studies thus far that have analysed the effect of cigarette smoking on the EBT. Bearing this in mind, in this longitudinal study we aim to analyse the EBT in current smokers, monitor the effects both of cigarette smoking on EBT and of what happens after smoking cessation. Twenty-five (25) smokers (59.5 ± 3.1 yrs, 12 M) who participated in a multi-disciplinary smoking cessation programme and 25 healthy never-smokers (58.7 ± 2.9, 13 M) underwent EBT measurement. EBT values were higher in smokers before smoking (T0) than in never-smokers [34.6 (34.2-35) vs 33.2 (32.4-33.7)°C, p < 0.001. The smokers repeated measurement 5 minutes after smoking a cigarette (T1) and 2 hours after (T2). They repeated EBC measurement after 1 week (T3) and then after 3 months (T4) from smoking cessation. EBT is higher in smokers compared to controls. EBT increases after cigarette smoking and progressively decreases with the increase of time from when the last cigarette was smoked. Thus, we can conclude that EBT is increased in smokers and also sensitive to the acute effect of cigarette smoke.
Collapse
Affiliation(s)
- Giovanna E Carpagnano
- a Institute of Respiratory Disease, Department of Medical and Surgical Sciences, University of Foggia , Italy
| | - Cinzia Ruggieri
- a Institute of Respiratory Disease, Department of Medical and Surgical Sciences, University of Foggia , Italy
| | - Giulia Scioscia
- a Institute of Respiratory Disease, Department of Medical and Surgical Sciences, University of Foggia , Italy
| | - Maria Maddalena Lo Storto
- a Institute of Respiratory Disease, Department of Medical and Surgical Sciences, University of Foggia , Italy
| | - Luigi Zoppo
- a Institute of Respiratory Disease, Department of Medical and Surgical Sciences, University of Foggia , Italy
| | - Maria P Foschino-Barbaro
- a Institute of Respiratory Disease, Department of Medical and Surgical Sciences, University of Foggia , Italy
| |
Collapse
|