1
|
Milesi J, Gras D, Chanez P, Coiffard B. Airway epithelium in lung transplantation: a potential actor for post-transplant complications? Eur Respir Rev 2024; 33:240093. [PMID: 39603662 PMCID: PMC11600126 DOI: 10.1183/16000617.0093-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/20/2024] [Indexed: 11/29/2024] Open
Abstract
Lung transplantation, a critical intervention for end-stage lung diseases, is frequently challenged by post-transplant complications. Indeed, primary graft dysfunction, anastomotic complications, infections and acute and chronic rejections pose significant hurdles in lung transplantation. While evidence regarding the role of airway epithelium after lung transplantation is still emerging, its importance is becoming increasingly recognised. This review looks at the complex involvement of airway epithelium in various post-transplant complications, while emphasising the utility of airway epithelial culture as a research model. In summary, by elucidating the involvement of airway epithelium in each post-transplant complication and explaining these intricate processes, the review aims to guide specific future research efforts and therapeutic strategies aimed at improving lung transplant outcomes and enhancing patient care.
Collapse
Affiliation(s)
- Jules Milesi
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Delphine Gras
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Pascal Chanez
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Benjamin Coiffard
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| |
Collapse
|
2
|
Abd Elrazik NA, Helmy SA. Betanin protects against bleomycin-induced pulmonary fibrosis by regulating the NLRP3/IL-1β/TGF-β1 pathway-mediated epithelial-to-mesenchymal transition. Food Funct 2024; 15:284-294. [PMID: 38083874 DOI: 10.1039/d3fo03464j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a life-threatening disease that leads to dyspnea and progressive loss of lung function. This study aimed to investigate the protective effect of betanin (BET), the major pigment in red beetroot, on pulmonary fibrosis induced by bleomycin (BLM) in rats and to assess the underlying mechanisms. In this view, total and differential cell counts and LDH activity in bronchoalveolar lavage fluid were estimated. Furthermore, MDA and GSH contents in the lungs were colorimetrically measured, while hydroxyproline, NLRP3, ASC, caspase-1, TGF-β1, and vimentin levels in lung tissue were evaluated using the ELISA technique. Moreover, IL-1β, E-cadherin, and α-SMA expressions were analyzed by immunostaining of lung specimens. BET treatment protects against pulmonary fibrosis as indicated by the reduction in total and differential cell counts, LDH activity, hydroxyproline, NLRP3, ASC, caspase-1, IL-1β, and TGF-β1 levels. MDA content was also decreased following BET administration, while GSH content was elevated. Additionally, BET suppressed the EMT process as evidenced by an increase in E-cadherin expression besides the reduction in vimentin and α-SMA expressions. To conclude, these results revealed the protective effect of BET against pulmonary fibrosis that might be attributed to the attenuation of the NLRP3/IL-1β/TGF-β1 signaling pathway and EMT process.
Collapse
Affiliation(s)
- Nesma A Abd Elrazik
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Sahar A Helmy
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
3
|
Wang Z. Role of transforming growth factor-β in airway remodelling in bronchiolitis obliterans. Growth Factors 2023; 41:192-209. [PMID: 37487145 DOI: 10.1080/08977194.2023.2239356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 07/12/2023] [Indexed: 07/26/2023]
Abstract
Airway remodelling is the main pathological mechanism of bronchiolitis obliterans (BO). Several studies have found that transforming growth factor-β (TGF-β) expression is increased in BO during airway remodelling, where it plays an important role in various biological processes by binding to its receptor complex to activate multiple signalling proteins and pathways. This review examines the role of TGF-β in airway remodelling in BO and its potential as a therapeutic target, highlighting the mechanisms of TGF-β activation and signalling, cellular targets of TGF-β actions, and research progress in TGF-β signalling and TGF-β-mediated processes.
Collapse
Affiliation(s)
- Ziwei Wang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Delbrel E, Voituron N, Boncoeur E. HIF and ER stress are involved in TGFβ1-mediated wound closure of alveolar epithelial cells. Exp Lung Res 2023:1-9. [PMID: 36947400 DOI: 10.1080/01902148.2023.2183996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Purpose: Alveolar epithelium dysfunction is associated with a very large spectrum of disease and an abnormal repair capacity of the airway epithelium has been proposed to explain the pathogenesis of Idiopathic Pulmonary Fibrosis (IPF). Following epithelium insult, the damaged cells will activate pathways implicated in the repair process, including proliferation and acquisition of migratory capacities to cover the denuded basement membrane. Induction of Endoplasmic Reticulum stress may be implicated in this process. Interestingly, ER stress excessive activation has been proposed as a central event associated with aberrant repair process and cellular dysfunction observed in IPF. Methods: We study by wound healing assay the molecular targets associated with Alveolar Epithelial Cells (AEC) repair. Results: We demonstrate that the wound recovery of AEC is associated with TGF-β1 signaling and increased transcriptional activity of ER stress and HIF-dependent genes. We further demonstrated that inhibition of TGF-β1 signaling, CHOP expression or HIF-1 expression, limits AECs wound closure. Conclusion: the use of pharmacological drugs targeting the ER/HIF-1 axis could be an attractive approach to limit AEC dysregulation in pathological condition, and confirmed a critical role of theses factor in response to alveolar injury.
Collapse
Affiliation(s)
- Eva Delbrel
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, Bobigny, France
| | - Nicolas Voituron
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, Bobigny, France
| | - Emilie Boncoeur
- Laboratoire Hypoxie & Poumon, UMR INSERM U1272, Université Sorbonne Paris-Nord, Bobigny, France
| |
Collapse
|
5
|
Walters EH, Shukla S, Ward C. Broadening concepts of core pathobiology in various aspects of COPD development. Eur Respir J 2022; 60:2201531. [PMID: 36202414 DOI: 10.1183/13993003.01531-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/23/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Eugene H Walters
- School of Medicine and Menzies Institute, University of Tasmania, Hobart, Australia
| | - Shakti Shukla
- Graduate School of Pharmacy, University of Technology Sydney, Ultimo, Australia
| | - Chis Ward
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University Medical School, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
6
|
Tumor necrosis factor-α coordinates with transforming growth factor-β1 to induce epithelial-mesenchymal transition and migration via the NF-κB/NOX4 pathway in bronchial epithelial cells. Mol Biol Rep 2022; 49:9325-9333. [PMID: 35913579 DOI: 10.1007/s11033-022-07777-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 07/06/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Epithelial-to-mesenchymal transition (EMT) is the process by which epithelial cells transform into mesenchymal cells, which plays a significant role in lung fibrotic disease. Transforming growth factor-β1(TGF-β1) is considered to be the most effective EMT inducer. The purpose of this study was to investigate the effect of the proinflammatory cytokine tumor necrosis factor-α (TNF-α) on TGF-β1-induced EMT and the underlying mechanisms in the human bronchial epithelial cell line BEAS-2B. METHODS Human bronchial epithelial BEAS-2B cells were treated with TGF-β1 and TNF-α separately or in combination for 24 h, and qRT-PCR, western blotting, immunofluorescence staining, and migration assays were used to investigate the EMT process. Moreover, to further explore the effect of the NF-κB pathway on the EMT process, inhibitor assays (BAY-117082, NF-κB inhibitor), wound healing assays, and western blotting were performed. RESULTS The results showed that both cytokines enhanced the transformation of BEAS-2B cells from epithelial to mesenchymal cells. In addition, combined treatment with TNF-α and TGF-β1 further reduced E-cadherin expression, which conversely elevated α-SMA and vimentin mRNA and protein levels. Correspondingly, the migration rate of BEAS-2B cells was also increased. Furthermore, inhibiting the NF-κB signaling pathway blocked the expression of EMT-related markers and NOX4 induced by TGF-β1 and TNF-α, as well as cell migration. CONCLUSION Taken together, TNF-α and TGF-β1 cooperatively promoted EMT and cell migration in BEAS-2B cells through the NF-κB/NOX4 signaling pathway.
Collapse
|
7
|
α-Viniferin and ε-Viniferin Inhibited TGF-β1-Induced Epithelial-Mesenchymal Transition, Migration and Invasion in Lung Cancer Cells through Downregulation of Vimentin Expression. Nutrients 2022; 14:nu14112294. [PMID: 35684095 PMCID: PMC9182810 DOI: 10.3390/nu14112294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/22/2022] [Accepted: 05/26/2022] [Indexed: 12/13/2022] Open
Abstract
Resveratrol has well-known anticancer properties; however, its oligomers, including α-viniferin, ε-viniferin, and kobophenol A, have not yet been well investigated. This is the first study examining the anti-epithelial-mesenchymal transition (EMT) effects of α-viniferin and ε-viniferin on A549, NCI-H460, NCI-H520, MCF-7, HOS, and U2OS cells. The results showed that α-viniferin and ε-viniferin significantly inhibited EMT, invasion and migration in TGF-β1- or IL-1β-induced non-small cell lung cancer. α-Viniferin and ε-viniferin also reversed TGF-β1-induced reactive oxygen species (ROS), MMP2, vimentin, Zeb1, Snail, p-SMAD2, p-SMAD3, and ABCG2 expression in A549 cells. Furthermore, ε-viniferin was found to significantly inhibit lung metastasis in A549 cell xenograft metastatic mouse models. In view of these findings, α-viniferin and ε-viniferin may play an important role in the prevention of EMT and cancer metastasis in lung cancer.
Collapse
|
8
|
Zhang S, Fan Y, Qin L, Fang X, Zhang C, Yue J, Bai W, Wang G, Chen Z, Renz H, Skevaki C, Liu X, Xie M. IL-1β augments TGF-β inducing epithelial-mesenchymal transition of epithelial cells and associates with poor pulmonary function improvement in neutrophilic asthmatics. Respir Res 2021; 22:216. [PMID: 34344357 PMCID: PMC8336269 DOI: 10.1186/s12931-021-01808-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/23/2021] [Indexed: 02/08/2023] Open
Abstract
Background Neutrophilic asthmatics (NA) have less response to inhaled corticosteroids. We aimed to find out the predictor of treatment response in NA. Methods Asthmatics (n = 115) and healthy controls (n = 28) underwent clinical assessment during 6-month follow-up with standardized therapy. Asthmatics were categorized by sputum differential cell count. The mRNA expressions were measured by RT-qPCR for sputum cytokines (IFN-γ, IL-1β, IL-27, FOXP3, IL-17A, and IL-5). The protein of IL-1β in sputum supernatant was detected by ELISA. Reticular basement membranes (RBM) were measured in the biopsy samples. The role and signaling pathways of IL-1β mediating the epithelial-mesenchymal transition (EMT) process were explored through A549 cells. Results NA had increased baseline sputum cell IL-1β expression compared to eosinophilic asthmatics (EA). After follow-up, NA had less improvement in FEV1 compared to EA. For all asthmatics, sputum IL-1β mRNA was positively correlated with protein expression. Sputum IL-1β mRNA and protein levels were negatively correlated to FEV1 improvement. After subgrouping, the correlation between IL-1β mRNA and FEV1 improvement was significant in NA but not in EA. Thickness of RBM in asthmatics was greater than that of healthy controls and positively correlated with neutrophil percentage in bronchoalveolar lavage fluid. In vitro experiments, the process of IL-1β augmenting TGF-β1-induced EMT cannot be abrogated by glucocorticoid or montelukast sodium, but can be reversed by MAPK inhibitors. Conclusions IL-1β level in baseline sputum predicts the poor lung function improvement in NA. The potential mechanism may be related to IL-1β augmenting TGF-β1-induced steroid-resistant EMT through MAPK signaling pathways. Trial registration: This study was approved by the Ethics Committee of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (IRB ID: 20150406). Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01808-7.
Collapse
Affiliation(s)
- Shengding Zhang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Yu Fan
- Department of Respiratory and Critical Care Medicine, Qiandongnanzhou People's Hospital, Kaili, China
| | - Lu Qin
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Xiaoyu Fang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Cong Zhang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Junqing Yue
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Wenxue Bai
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Harld Renz
- Institute of Laboratory Medicine, Philipps Universität Marburg, Marburg, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), and the German Center for Lung Research (DZL), Marburg, Germany
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine, Philipps Universität Marburg, Marburg, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), and the German Center for Lung Research (DZL), Marburg, Germany
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China. .,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China.
| | - Min Xie
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China. .,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China.
| |
Collapse
|
9
|
Ladak SS, Roebuck E, Powell J, Fisher AJ, Ward C, Ali S. The Role of miR-200b-3p in Modulating TGF-β1-induced Injury in Human Bronchial Epithelial Cells. Transplantation 2020; 103:2275-2286. [PMID: 31283671 DOI: 10.1097/tp.0000000000002845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Dysregulation of microRNAs (miRNAs) has been implicated in airway diseases where transforming growth factor-β (TGF-β)-induced epithelial-mesenchymal transition (EMT) may contribute to pathophysiology. Our study investigated the role of miRNA-200b in TGF-β1-induced EMT in human bronchial epithelial cells. METHODS NanoString nCounter miRNA assay was used to profile miRNA in control versus TGF-β1 (1, 4, and 24 h) stimulated BEAS-2B cells. Immortalized primary bronchial epithelial cell line (BEAS-2B cells), human primary bronchial epithelial cells (PBECs), and PBECs derived post-lung transplant were transfected with miR-200b-3p mimics and EMT marker expression was examined at RNA and protein level. miRNA target studies were performed and validated using computational tools and luciferase assay. In situ hybridization was done on normal lung tissue to localize miR-200b-3p in airway epithelium. RESULTS miR-200b-3p was downregulated post-TGF-β1 treatment compared with control in BEAS-2B. miR-200b-3p mimic transfection before TGF-β1 stimulation maintained epithelial marker expression and downregulated mesenchymal cell markers at RNA and protein level in BEAS-2B cells and PBECs. Furthermore, miR-200b-3p mimics reversed established TGF-β1-induced EMT in BEAS-2B cells. miR-200b-3p targets, ZNF532, and ZEB2 were validated as direct targets using luciferase assay. miR-200b-3p mimics suppress TGF-β1-induced EMT via inhibition of ZNF532 and ZEB2. In situ hybridization showed that miR-200b-3p is expressed in the normal lung epithelium. Additionally, miR-200b-3p mimics inhibit EMT in the presence of TGF-β1 in PBECs derived from lung allograft. CONCLUSIONS We provide proof of concept that miR-200b-3p protects airway epithelial cells from EMT. Manipulating miR-200b-3p expression may represent a novel therapeutic modulator in airway pathophysiology.
Collapse
Affiliation(s)
- Shameem S Ladak
- Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Eliott Roebuck
- Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jason Powell
- Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Otolaryngology, Head and Neck Surgery, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Andrew J Fisher
- Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom.,Institute of Transplantation, Newcastle Upon Tyne Hospitals, Newcastle upon Tyne, United Kingdom
| | - Chris Ward
- Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simi Ali
- Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
10
|
Abstract
Introduction: Lung transplantation remains an important treatment for patients with end stage lung disease. Chronic lung allograft dysfunction (CLAD) remains the greatest limiting factor for long term survival. As the diagnosis of CLAD is based on pulmonary function tests, significant lung injury is required before a diagnosis is feasible, likely when irreversible damage has already occurred. Therefore, research is ongoing for early CLAD recognition, with biomarkers making up a substantial amount of this research.Areas covered: The purpose of this review is to describe available biomarkers, focusing on those which aid in predicting CLAD and distinguishing between different CLAD phenotypes. We describe biomarkers presenting in bronchial alveolar lavage (BAL) as well as circulating in peripheral blood, both of which offer an appealing alternative to lung biopsy.Expert opinion: Development of CLAD involves complex, multiple immune and nonimmune mechanisms. Therefore, evaluation of potential CLAD biomarkers serves a dual purpose: clinically, the goal remains early detection and identification of patients at increased risk. Simultaneously, biomarkers offer insight into the different mechanisms involved in the pathophysiology of CLAD, leading to the development of possible interventions. The ultimate goal is the development of both preventive and early intervention strategies for CLAD to improve the overall survival of our lung transplant recipients.
Collapse
Affiliation(s)
- Osnat Shtraichman
- Division of Pulmonary, Allergy & Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Pulmonary institute, Rabin Medical Center, Petach Tikva, Israel; Sackler School of Medicine, Tel Aviv, Israel
| | - Joshua M Diamond
- Division of Pulmonary, Allergy & Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Renaud-Picard B, Vallière K, Toussaint J, Kreutter G, El-Habhab A, Kassem M, El-Ghazouani F, Olland A, Hirschi S, Porzio M, Chenard MP, Toti F, Kessler L, Kessler R. Epithelial-mesenchymal transition and membrane microparticles: Potential implications for bronchiolitis obliterans syndrome after lung transplantation. Transpl Immunol 2020; 59:101273. [PMID: 32097721 DOI: 10.1016/j.trim.2020.101273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Long term survival post lung transplantation (LTx) is limited by the occurrence of bronchiolitis obliterans syndrome (BOS). One mechanism involved is the epithelial-mesenchymal transition (EMT). Membrane microparticles (MPs) are known to be involved in some respiratory diseases and in other organs allograft rejection episodes. We hypothesized that leukocyte-derived MPs likely contribute to EMT. To emphasize this physiological concept, our objectives were to: (1) confirm the presence of EMT on explanted lungs from patients who underwent a second LTx for BOS; 2) characterize circulating MPs in transplanted patients, with or without BOS; (3) evaluate in vitro the effect of monocyte-derived MPs in EMT of human bronchial epithelial cells. Our IHC analysis on explanted graft lungs revealed significant pathological signs of EMT with an inhomogeneous destruction of the bronchial epithelium, with decreased expression of the epithelial protein E-cadherin and increased expression of the mesenchymal protein Vimentin. The immunophenotyping of MPs demonstrated that the concentration of MPs carrying E-cadherin was lower in patients affected by BOS (p = .007). In vitro, monocyte-derived MPs produced with LPS were associated with decreased E-cadherin expression (p < .05) along with significant morphological and functional cell modifications. MPs may play a role in EMT onset in bronchial epithelium following LTx.
Collapse
Affiliation(s)
- Benjamin Renaud-Picard
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France; INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France.
| | - Kevin Vallière
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Justine Toussaint
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Guillaume Kreutter
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Ali El-Habhab
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Mohamad Kassem
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Fatiha El-Ghazouani
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Anne Olland
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France; Department of Thoracic Surgery and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France
| | - Sandrine Hirschi
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France
| | - Michele Porzio
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France
| | | | - Florence Toti
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France
| | - Laurence Kessler
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France
| | - Romain Kessler
- Department of Respiratory Medicine and Strasbourg Lung Transplant Program, University Hospital of Strasbourg, France; INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, University of Strasbourg, France; Faculty of Medicine, Federation of Translational Medicine (FMTS), Strasbourg, France
| |
Collapse
|
12
|
Therkildsen JR, Christensen MG, Tingskov SJ, Wehmöller J, Nørregaard R, Praetorius HA. Lack of P2X 7 Receptors Protects against Renal Fibrosis after Pyelonephritis with α-Hemolysin-Producing Escherichia coli. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1201-1211. [PMID: 30926332 DOI: 10.1016/j.ajpath.2019.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 01/07/2023]
Abstract
Severe urinary tract infections are commonly caused by sub-strains of Escherichia coli secreting the pore-forming virulence factor α-hemolysin (HlyA). Repeated or severe cases of pyelonephritis can cause renal scarring that subsequently can lead to progressive failure. We have previously demonstrated that HlyA releases cellular ATP directly through its membrane pore and that acute HlyA-induced cell damage is completely prevented by blocking ATP signaling. Local ATP signaling and P2X7 receptor activation play a key role in the development of tissue fibrosis. This study investigated the effect of P2X7 receptors on infection-induced renal scarring in a murine model of pyelonephritis. Pyelonephritis was induced by injecting 100 million HlyA-producing, uropathogenic E. coli into the urinary bladder of BALB/cJ mice. A similar degree of pyelonephritis and mortality was confirmed at day 5 after infection in P2X7+/+ and P2X7-/- mice. Fibrosis was first observed 2 weeks after infection, and the data clearly demonstrated that P2X7-/- mice and mice exposed to the P2X7 antagonist, brillian blue G, show markedly less renal fibrosis 14 days after infection compared with controls (P < 0.001). Immunohistochemistry revealed comparable early neutrophil infiltration in the renal cortex from P2X7+/+ and P2X7-/- mice. Interestingly, lack of P2X7 receptors resulted in diminished macrophage infiltration and reduced neutrophil clearance in the cortex of P2X7-/- mice. Hence, this study suggests the P2X7 receptor to be an appealing antifibrotic target after renal infections.
Collapse
Affiliation(s)
| | | | - Stine J Tingskov
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Julia Wehmöller
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
13
|
Li Y, Zhu G, Zhai H, Jia J, Yang W, Li X, Liu L. Simultaneous stimulation with tumor necrosis factor-α and transforming growth factor-β1 induces epithelial-mesenchymal transition in colon cancer cells via the NF-κB pathway. Oncol Lett 2018; 15:6873-6880. [PMID: 29725419 PMCID: PMC5920468 DOI: 10.3892/ol.2018.8230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 12/19/2017] [Indexed: 12/11/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is critical in the progression of numerous types of carcinoma, and endows invasive and metastatic properties upon cancer cells. The tumor microenvironment facilitates tumor metastasis to distant organs. Various signaling pathways contribute to this process. In the present study, SW480 colon adenocarcinoma cells were treated with transforming growth factor-β1 (TGF-β1; 10 ng/ml) and tumor necrosis factor-α (TNF-α; 20 ng/ml), alone or in combination, for 72 h, and EMT was assessed using immunofluorescence, western blot analysis and migration assays. The functions of p38 mitogen-activated protein kinase, extracellular signal-regulated kinase (ERK) and nuclear factor-κB (NF-κB) pathways in EMT were examined. It was demonstrated that the cooperation of TGF-β1 and TNF-α signaling promoted the morphological conversion of the SW480 cells from an epithelial to a mesenchymal phenotype. Furthermore, simultaneous exposure to TNF-α and TGF-β1 downregulated the expression of E-cadherin (an epithelial marker) and increased the expression of N-cadherin and vimentin (mesenchymal markers). Additionally, the migratory capacity of the SW480 cells increased. The inhibition of p38 and ERK signaling exhibited no effect on EMT, whereas the inhibition of inhibitor of NF-κB kinase subunit β blocked the EMT induced by TGF-β1 and TNF-α. In conclusion, the results of the present study demonstrated that TNF-α and TGF-β1 synergistically promoted EMT in SW480 cells via the NF-κB pathway, independent of p38 activation and ERK1/2 signaling. These results suggest a novel function of TGF-β1 and TNF-α during EMT in colon carcinoma and, thus, provide insights into potential therapeutic interventions.
Collapse
Affiliation(s)
- Yuanfei Li
- Department of Oncology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Guoqiang Zhu
- Department of Burns and Plastic Surgery, The 264th Hospital of the PLA, Taiyuan, Shanxi 030001, P.R. China
| | - Huihong Zhai
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Junmei Jia
- Department of Oncology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Wenhui Yang
- Department of Oncology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xiaoqing Li
- Department of Oncology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Lixin Liu
- Department of Gastroenterology and Hepatology, The First Clinical Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
14
|
Wang A, Lu C, Ning Z, Gao W, Xie Y, Zhang N, Liang J, Abbasi FS, Yan Q, Liu J. Tumor-associated macrophages promote Ezrin phosphorylation-mediated epithelial-mesenchymal transition in lung adenocarcinoma through FUT4/LeY up-regulation. Oncotarget 2018; 8:28247-28259. [PMID: 28423676 PMCID: PMC5438647 DOI: 10.18632/oncotarget.16001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 02/27/2017] [Indexed: 12/28/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are key components of tumor microenvironment (TME) during tumorigenesis and progression. However, the role of TAMs in lung adenocarcinoma is still unclear. In this study, we aimed to clarify the mechanism underlying the crosstalk between TAMs and epithelial-mesenchymal transition (EMT) of lung adenocarcinoma. Fucosyltransferase IV (FUT4) and its synthetic cancer sugar antigen Lewis Y (LeY) was aberrantly elevated in various solid tumors, it plays critical role in the invasion and metastasis. Here, we found that in lung adenocarcinoma samples, the density of TAMs correlates with E-cadherin level and LeY level. In vitro assays, M2 macrophages promoted FUT4/LeY expression through the transforming growth factor-β1(TGF-β1)/Smad2/3 signaling pathway. FUT4/LeY was indispensable in M2 macrophages-mediated cytoskeletal remodeling and EMT. Furthermore, fucosylation of Ezrin mediated by FUT4/LeY can promote the phosphorylation of Ezrin, which was the critical mechanism of M2 macrophages-induced EMT. In vivo assays confirmed that M2 macrophages promoted EMT through the up-regulation of LeY and phosphorylated Ezrin. Together, our results revealed that TAMs promote Ezrin phosphorylation-mediated EMT in lung adenocarcinoma through FUT4/LeY- mediated fucosylation. Targeting this newly identified signaling may offer new possibilities for immunotherapy in lung adenocarcinoma.
Collapse
Affiliation(s)
- Aman Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Chang Lu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Zhen Ning
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Wei Gao
- City College, Zhejiang University, 310000, Hangzhou, China
| | - Yunpeng Xie
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Ningning Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Jinxiao Liang
- Department of Thoracic Surgery, Zhejiang Cancer Hospital, 310000, Hangzhou, China
| | - Faisal S Abbasi
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Qiu Yan
- Department of Biochemistry and Molecular Biology, Dalian Medical University, 116011, Dalian, China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| |
Collapse
|
15
|
Tang W, Huang S, Du L, Sun W, Yu Z, Zhou Y, Chen J, Li X, Li X, Yu B, Chen D. Expression of HMGB1 in maternal exposure to fine particulate air pollution induces lung injury in rat offspring assessed with micro-CT. Chem Biol Interact 2018; 280:64-69. [DOI: 10.1016/j.cbi.2017.12.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023]
|
16
|
Royer PJ, Henrio K, Pain M, Loy J, Roux A, Tissot A, Lacoste P, Pison C, Brouard S, Magnan A. TLR3 promotes MMP-9 production in primary human airway epithelial cells through Wnt/β-catenin signaling. Respir Res 2017; 18:208. [PMID: 29237464 PMCID: PMC5729411 DOI: 10.1186/s12931-017-0690-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/28/2017] [Indexed: 12/27/2022] Open
Abstract
Background Airway epithelial cells (AEC) act as the first line of defence in case of lung infections. They constitute a physical barrier against pathogens and they participate in the initiation of the immune response. Yet, the modalities of pathogen recognition by AEC and the consequences on the epithelial barrier remain poorly documented. Method We investigated the response of primary human AEC to viral (polyinosinic-polycytidylic acid, poly(I:C)) and bacterial (lipopolysaccharide, LPS) stimulations in combination with the lung remodeling factor Transforming Growth Factor-β (TGF-β). Results We showed a strong production of pro-inflammatory cytokines (Interleukin (IL)-6, Tumor Necrosis Factor α, TNFα) or chemokines (CCL2, CCL3, CCL4, CXCL10, CXCL11) by AEC stimulated with poly(I:C). Cytokine and chemokine production, except CXCL10, was Toll Like Receptor (TLR)-3 dependent and although they express TLR4, we found no cytokine production after LPS stimulation. Poly(I:C), but not LPS, synergised with TGF-β for the production of matrix metalloproteinase-9 (MMP-9) and fibronectin. Mechanistic analyses suggest the secretion of Wnt ligands by AEC along with a degradation of the cellular junctions after poly(I:C) exposure, leading to the release of β-catenin from the cell membrane and stimulation of the Wnt/β-catenin pathway. Conclusion Our results highlight the cross talk between TGF-β and TLR signaling in bronchial epithelium and its impact on the remodeling process. Electronic supplementary material The online version of this article (10.1186/s12931-017-0690-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- P-J Royer
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France.
| | - K Henrio
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - M Pain
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - J Loy
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - A Roux
- Hopital Foch, Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department,Suresnes, France, Université Versailles Saint-Quentin-en-Yvelines, UPRESS EA220, Montigny le Bretonneux, Grenoble, France
| | - A Tissot
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - P Lacoste
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - C Pison
- Clinique Universitaire de Pneumologie, Pôle Thorax et Vaisseaux, CHU Grenoble Alpes, Grenoble, France.,Université Grenoble Alpes, Grenoble, France.,Laboratoire de Bioénergétique Fondamentale et Appliquée, Inserm, 1055, Grenoble, France
| | - S Brouard
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,Faculté de Médecine, Université de Nantes, Nantes, France.,CIC Biotherapy, CHU Nantes, Nantes, France
| | - A Magnan
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | | |
Collapse
|
17
|
Inhibition of T Cell Alloreactivity by Bronchial Epithelium Is Impaired in Lung Transplant Recipients, Through Pathways Involving TGF-β, IL-10 and HLA-G. Transplantation 2017; 101:2192-2199. [PMID: 27820781 DOI: 10.1097/tp.0000000000001553] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Bronchiolitis obliterans syndrome (BOS) after lung transplantation (LTx) results from bronchial epithelial cell (BECs) damages, thought to be orchestrated by T cells primed by antigen-presenting cell presenting alloantigens. In this cell cross-talk, BECs are also suspected to play a pivotal immunosuppressive role in T cell alloreactivity. We studied the immunomodulating role of BECs in a human ex vivo model of allogeneic T cell response, both in healthy subjects and LTx recipients. METHODS BECs from 35 LTx recipients (n = 22 stable, n = 13 BOS) and healthy controls (n = 25) were cultured as primary cell cultures. Their inhibitory capacities through the involvement of tolerogenic molecules (HLA-G, TGF-β, and IL-10) were tested on a mixed lymphocyte reaction between antigen-presenting cells and recipient T cells. RESULTS Control BECs inhibited T cell alloproliferation by a mean of 53 ± 7%. This inhibitory effect of BECs was significantly reduced in the stable LTx group (24 ± 8%, P = 0.009), but not in the BOS TxP group (53 ± 10%, P = 0.97). Neutralization of HLA-G, TGF-β, and IL-10 partially restored T cell alloproliferation, arguing for their involvement in the immunosuppressive effect of BECs. BECs culture supernatant from stable LTx patients with impaired BEC properties showed a skewed Th2-type secretion profile (high IL-4/IFN-γ ratio). CONCLUSIONS The inhibitory properties of BECs are dysregulated in stable LTx recipients, which could suggest their instrumental role in the initiation of BOS process and potential targeted therapies.
Collapse
|
18
|
NLRP3 participates in the regulation of EMT in bleomycin-induced pulmonary fibrosis. Exp Cell Res 2017; 357:328-334. [DOI: 10.1016/j.yexcr.2017.05.028] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/18/2017] [Accepted: 05/31/2017] [Indexed: 12/23/2022]
|
19
|
Jolly MK, Ward C, Eapen MS, Myers S, Hallgren O, Levine H, Sohal SS. Epithelial-mesenchymal transition, a spectrum of states: Role in lung development, homeostasis, and disease. Dev Dyn 2017. [DOI: 10.1002/dvdy.24541] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Mohit Kumar Jolly
- Center for Theoretical Biological Physics; Rice University; Houston Texas
| | - Chris Ward
- Institute of Cellular Medicine; Newcastle University; Newcastle upon Tyne United Kingdom
| | - Mathew Suji Eapen
- School of Health Sciences; Faculty of Health, University of Tasmania, Launceston, University of Tasmania; Hobart Tasmania Australia
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease; University of Tasmania; Hobart Tasmania Australia
| | - Stephen Myers
- School of Health Sciences; Faculty of Health, University of Tasmania, Launceston, University of Tasmania; Hobart Tasmania Australia
| | - Oskar Hallgren
- Department of Experimental Medical Sciences; Department of Respiratory Medicine and Allergology, Lund University; Sweden
| | - Herbert Levine
- Center for Theoretical Biological Physics; Rice University; Houston Texas
| | - Sukhwinder Singh Sohal
- School of Health Sciences; Faculty of Health, University of Tasmania, Launceston, University of Tasmania; Hobart Tasmania Australia
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease; University of Tasmania; Hobart Tasmania Australia
| |
Collapse
|
20
|
Pain M, Royer PJ, Loy J, Girardeau A, Tissot A, Lacoste P, Roux A, Reynaud-Gaubert M, Kessler R, Mussot S, Dromer C, Brugière O, Mornex JF, Guillemain R, Dahan M, Knoop C, Botturi K, Pison C, Danger R, Brouard S, Magnan A. T Cells Promote Bronchial Epithelial Cell Secretion of Matrix Metalloproteinase-9 via a C-C Chemokine Receptor Type 2 Pathway: Implications for Chronic Lung Allograft Dysfunction. Am J Transplant 2017; 17:1502-1514. [PMID: 27982503 DOI: 10.1111/ajt.14166] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/30/2016] [Accepted: 12/04/2016] [Indexed: 01/25/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) is the major limitation of long-term survival after lung transplantation. CLAD manifests as bronchiolitis obliterans syndrome (BOS) or restrictive allograft syndrome (RAS). Alloimmune reactions and epithelial-to-mesenchymal transition have been suggested in BOS. However, little is known regarding the role of allogenicity in epithelial cell differentiation. Primary human bronchial epithelial cells (BECs) were treated with activated T cells in the presence or absence of transforming growth factor (TGF)-β. The expression of epithelial and mesenchymal markers was investigated. The secretion of inflammatory cytokines and matrix metalloproteinase (MMP)-9 was measured in culture supernatants and in plasma from lung transplant recipients (LTRs): 49 stable, 29 with BOS, and 16 with RAS. We demonstrated that C-C motif chemokine 2 secreted by T cells supports TGF-β-induced MMP-9 production by BECs after binding to C-C chemokine receptor type 2. Longitudinal investigation in LTRs revealed a rise in plasma MMP-9 before CLAD onset. Multivariate analysis showed that plasma MMP-9 was independently associated with BOS (odds ratio [OR] = 6.19, p = 0.002) or RAS (OR = 3.9, p = 0.024) and predicted the occurrence of CLAD 12 months before the functional diagnosis. Thus, immune cells support airway remodeling through the production of MMP-9. Plasma MMP-9 is a potential predictive biomarker of CLAD.
Collapse
Affiliation(s)
- M Pain
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - P-J Royer
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - J Loy
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - A Girardeau
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - A Tissot
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - P Lacoste
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - A Roux
- Hôpital Foch, Suresnes, Université Versailles Saint-Quentin-en-Yvelines, UPRES EA220, Versailles, France
| | | | - R Kessler
- CHU de Strasbourg, Strasbourg, France
| | - S Mussot
- Centre Chirurgical Marie Lannelongue, Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardiopulmonaire, Le Plessis Robinson, France
| | - C Dromer
- CHU de Bordeaux, Bordeaux, France
| | - O Brugière
- Hôpital Bichat, Service de Pneumologie et Transplantation Pulmonaire, Paris, France
| | - J-F Mornex
- Université de Lyon, INRA, UMR754, Lyon, Hospices Civils de Lyon, Lyon, France
| | | | - M Dahan
- CHU de Toulouse, Toulouse, France
| | - C Knoop
- Hôpital Erasme, Bruxelles, Belgique
| | - K Botturi
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - C Pison
- Clinique Universitaire Pneumologie, Pôle Thorax et Vaisseaux, CHU de Grenoble, Université de Grenoble, INSERM U1055, Grenoble, France
| | - R Danger
- Université de Nantes, INSERM U1064 and Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | - S Brouard
- Université de Nantes, INSERM U1064 and Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | - A Magnan
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | | |
Collapse
|
21
|
Zhang H, Luo J, Alcorn JF, Chen K, Fan S, Pilewski J, Liu A, Chen W, Kolls JK, Wang J. AIM2 Inflammasome Is Critical for Influenza-Induced Lung Injury and Mortality. THE JOURNAL OF IMMUNOLOGY 2017; 198:4383-4393. [PMID: 28424239 PMCID: PMC5439025 DOI: 10.4049/jimmunol.1600714] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 03/24/2017] [Indexed: 11/19/2022]
Abstract
The absent in melanoma 2 (AIM2) inflammasome plays an important role in many viral and bacterial infections, but very little is known about its role in RNA virus infection, including influenza A virus (IAV). In this study, we have designed in vivo and in vitro studies to determine the role of AIM2 in infections with lethal doses of IAVs A/PR8/34 and A/California/07/09. In wild-type mice, IAV infection enhanced AIM2 expression, induced dsDNA release, and stimulated caspase-1 activation and release of cleaved IL-1β in the lung, which was significantly reduced in AIM2-deficient mice. Interestingly, AIM2 deficiency did not affect the transcription of caspase-1 and IL-1β. In addition, AIM2-deficient mice exhibited attenuated lung injury and significantly improved survival against IAV challenges, but did not alter viral burden in the lung. However, AIM2 deficiency did not seem to affect adaptive immune response against IAV infections. Furthermore, experiments with AIM2-specific small interfering RNA-treated and AIM2-deficient human and mouse lung alveolar macrophages and type II cells indicated a macrophage-specific function of AIM2 in regulation of IAV-stimulated proinflammatory response. Collectively, our results demonstrate that influenza infection activates the AIM2 inflammasome, which plays a critical role in IAV-induced lung injury and mortality. AIM2 might serve as a therapeutic target for combating influenza-associated morbidity and mortality without compromising the host antiviral responses.
Collapse
Affiliation(s)
- Hongbo Zhang
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Jiadi Luo
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224.,Department of Pathology, Second Affiliated Xiangya Hospital, Central South University, Changsha 410078, China
| | - John F Alcorn
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Kong Chen
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Songqing Fan
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224.,Department of Pathology, Second Affiliated Xiangya Hospital, Central South University, Changsha 410078, China
| | - Joseph Pilewski
- Pulmonary, Allergy, and Critical Care Medicine Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224; and
| | - Aizhong Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Central South University, Changsha 410078, China
| | - Wei Chen
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Jay K Kolls
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Jieru Wang
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224;
| |
Collapse
|
22
|
Zhang SB, Sun X, Wu Q, Wu JP, Chen HY. Impaired Capacity of Fibroblasts to Support Airway Epithelial Progenitors in Bronchiolitis Obliterans Syndrome. Chin Med J (Engl) 2016; 129:2040-2044. [PMID: 27569228 PMCID: PMC5009585 DOI: 10.4103/0366-6999.189058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Bronchiolitis obliterans syndrome (BOS) often develops in transplant patients and results in injury to the respiratory and terminal airway epithelium. Owing to its rising incidence, the pathogenesis of BOS is currently an area of intensive research. Studies have shown that injury to the respiratory epithelium results in dysregulation of epithelial repair. Airway epithelial regeneration is supported by stromal cells, including fibroblasts. This study aimed to investigate whether the supportive role of lung fibroblasts is altered in BOS. METHODS Suspensions of lung cells were prepared by enzyme digestion. Lung progenitor cells (LPCs) were separated by fluorescence-activated cell sorting. Lung fibroblasts from patients with BOS or healthy controls were mixed with sorted mouse LPCs to compare the colony-forming efficiency of LPCs by counting the number of colonies with a diameter of ≥50 μm in each culture. Statistical analyses were performed using the SPSS 17.0 software (SPSS Inc., USA). The paired Student's t-test was used to test for statistical significance. RESULTS LPCs were isolated with the surface phenotype of CD31-CD34-CD45- EpCAM+Sca-1+. The colony-forming efficiency of LPCs was significantly reduced when co-cultured with fibroblasts isolated from patients with BOS. The addition of SB431542 increased the colony-forming efficiency of LPCs to 1.8%; however, it was still significantly less than that in co-culture with healthy control fibroblasts (P < 0.05). CONCLUSION The epithelial-supportive capacity of fibroblasts is impaired in the development of BOS and suggest that inefficient repair of airway epithelium could contribute to persistent airway inflammation in BOS.
Collapse
Affiliation(s)
- Su-Bei Zhang
- Haihe Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Xin Sun
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Qi Wu
- Tianjin Institute of Respiratory Diseases, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Jun-Ping Wu
- Department of Respiratory, Tianjin Haihe Hospital, Tianjin 300350, China
| | - Huai-Yong Chen
- Department of Basic Medicine, Tianjin Haihe Hospital, Tianjin 300350, China
| |
Collapse
|
23
|
|
24
|
Borthwick LA. The IL-1 cytokine family and its role in inflammation and fibrosis in the lung. Semin Immunopathol 2016; 38:517-34. [PMID: 27001429 PMCID: PMC4896974 DOI: 10.1007/s00281-016-0559-z] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/25/2016] [Indexed: 12/24/2022]
Abstract
The IL-1 cytokine family comprises 11 members (7 ligands with agonist activity, 3 receptor antagonists and 1 anti-inflammatory cytokine) and is recognised as a key mediator of inflammation and fibrosis in multiple tissues including the lung. IL-1 targeted therapies have been successfully employed to treat a range of inflammatory conditions such as rheumatoid arthritis and gouty arthritis. This review will introduce the members of the IL-1 cytokine family, briefly discuss the cellular origins and cellular targets and provide an overview of the role of these molecules in inflammation and fibrosis in the lung.
Collapse
Affiliation(s)
- L A Borthwick
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, 4th Floor, William Leech Building, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
25
|
|
26
|
Jonigk D, Izykowski N, Rische J, Braubach P, Kühnel M, Warnecke G, Lippmann T, Kreipe H, Haverich A, Welte T, Gottlieb J, Laenger F. Molecular Profiling in Lung Biopsies of Human Pulmonary Allografts to Predict Chronic Lung Allograft Dysfunction. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:3178-88. [PMID: 26476349 DOI: 10.1016/j.ajpath.2015.08.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/03/2015] [Accepted: 08/28/2015] [Indexed: 10/22/2022]
Abstract
Chronic lung allograft dysfunction (CLAD) is the main reason for poor long-term outcome of lung transplantation, with bronchiolitis obliterans (BO) representing the predominant pathological feature. BO is defined as a progressive fibrous obliteration of the small airways, thought to be triggered by a combination of nonimmune bronchial injury and alloimmune and autoimmune mechanisms. Because biopsy samples are too insensitive to reliably detect BO and a decline in lung function test results, which is clinically used to define CLAD, does not detect early stages, there is need for alternative biomarkers for early diagnosis. Herein, we analyzed the cellular composition and differential expression of 45 tissue remodeling-associated genes in transbronchial lung biopsy specimens from two cohorts with 18 patients each: patients who did not develop CLAD within 3 years after transplantation (48 biopsy specimens) and patients rapidly developing CLAD within the first 3 postoperative years (57 biopsy specimens). Integrating the mRNA expression levels of the five most significantly dysregulated genes from the transforming growth factor-β axis (BMP4, IL6, MMP1, SMAD1, and THBS1) into a score, patient groups could be confidently separated and the outcome predicted (P < 0.001). We conclude that overexpression of fibrosis-associated genes may be valuable as a tissue-based molecular biomarker to more accurately diagnose or predict the development of CLAD.
Collapse
Affiliation(s)
- Danny Jonigk
- Institute of Pathology, Hanover Medical School, Hanover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany.
| | - Nicole Izykowski
- Institute of Pathology, Hanover Medical School, Hanover, Germany
| | - Johanna Rische
- Institute of Pathology, Hanover Medical School, Hanover, Germany
| | - Peter Braubach
- Institute of Pathology, Hanover Medical School, Hanover, Germany
| | - Mark Kühnel
- Institute of Functional and Applied Anatomy, Hanover Medical School, Hanover, Germany
| | - Gregor Warnecke
- Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany; Department of Thoracic Surgery, Hanover Medical School, Hanover, Germany
| | - Torsten Lippmann
- Institute of Pathology, Hanover Medical School, Hanover, Germany
| | - Hans Kreipe
- Institute of Pathology, Hanover Medical School, Hanover, Germany
| | - Axel Haverich
- Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany; Department of Thoracic Surgery, Hanover Medical School, Hanover, Germany
| | - Tobias Welte
- Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany; Department of Respiratory Medicine, Hanover Medical School, Hanover, Germany
| | - Jens Gottlieb
- Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany; Department of Respiratory Medicine, Hanover Medical School, Hanover, Germany
| | - Florian Laenger
- Institute of Pathology, Hanover Medical School, Hanover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease, the Hanover Division of the German Center for Lung Research, Gießen, Germany
| |
Collapse
|
27
|
Leukocytes: The Double-Edged Sword in Fibrosis. Mediators Inflamm 2015; 2015:652035. [PMID: 26568664 PMCID: PMC4629055 DOI: 10.1155/2015/652035] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/25/2015] [Indexed: 02/06/2023] Open
Abstract
Skin tissue scar formation and fibrosis are often characterized by the increased production and deposition of extracellular matrix components, accompanied by the accumulation of a vast number of myofibroblasts. Scaring is strongly associated with inflammation and wound healing to regain tissue integrity in response to skin tissue injury. However, increased and uncontrolled inflammation, repetitive injury, and individual predisposition might lead to fibrosis, a severe disorder resulting in the formation of dense and stiff tissue that loses the physical properties and physiological functions of normal tissue. Fibrosis is an extremely complicated and multistage process in which bone marrow-derived leukocytes act as both pro- and antifibrotic agents, and therefore, few, if any, effective therapies are available for the most severe and lethal forms of fibrosis. Herein, we discuss the current knowledge on the multidimensional impact of leukocytes on the induction of fibrosis, focusing on skin fibrosis.
Collapse
|
28
|
Lee DD, Schwarz MA. Adapted approach to profile genes while reconciling Vegf-a mRNA expression in the developing and injured lung. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1202-11. [PMID: 25868151 DOI: 10.1152/ajplung.00053.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/09/2015] [Indexed: 12/12/2022] Open
Abstract
During lung development and injury, messenger RNA (mRNA) transcript levels of genes fluctuate over both space and time. Quantitative PCR (qPCR) is a highly sensitive, widely used technique to measure the mRNA levels. The sensitivity of this technique can be disadvantageous and errors amplified when each qPCR assay is not validated. In contrast to other organs, lungs have high RNase activity, resulting in less than optimal RNA integrity. We implemented a strategy to address these limitations in developing and injured lungs. Parameters were established and a filter designed that optimized amplicon length and included or excluded samples based on RNA integrity. This approach was illustrated and validated by measuring mRNA levels including Vegf-a in newborn mouse lungs that were injured by 85% oxygen (hyperoxia) for 12 days and compared with control (normoxia). We demonstrate that, in contrast to contradictory Vegf-a expression when normalized to the least suitable housekeeping genes, application of this filter and normalization to most suitable three housekeeping genes, Hprt, Eef2, and Rpl13a, gave reproducible Vegf-a expression, thus corroborating the sample filter. Accordingly, both short amplicon length and proper normalization to ranked, evaluated genes minimized erroneous fluctuation and qPCR amplification issues associated with nonideal RNA integrity in injured and developing lungs. Furthermore, our work uncovers how RNA integrity, purity, amplicon length, and discovery of stable candidate reference genes enhance precision of qPCR results and utilizes the advantages of qPCR in developmental studies.
Collapse
Affiliation(s)
- Daniel D Lee
- Department of Pediatrics, Indiana University School of Medicine, South Bend, Indiana
| | - Margaret A Schwarz
- Department of Pediatrics, Indiana University School of Medicine, South Bend, Indiana
| |
Collapse
|
29
|
Huang X, Yan X, Zhang Z, Li X. Seeding of recipient-originated epithelial cells attenuates epithelial to mesenchymal transition in rat tracheal allotransplantation. Otolaryngol Head Neck Surg 2015; 152:1068-74. [PMID: 25820583 DOI: 10.1177/0194599815577102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 02/20/2015] [Indexed: 11/15/2022]
Abstract
OBJECTIVE The specific role and mechanism of epithelium in the progression of obliterative airway disease (OAD) after tracheal allotransplantation remain poorly understood. In this study, we used rat heterotopic tracheal transplantation to investigate the mechanism of epithelial cell seeding during the process of OAD. STUDY DESIGN Prospective, basic science. SETTING Research laboratory. SUBJECTS AND METHODS In total, 120 Sprague Dawley (SD) rats and 90 Wistar rats were used. Tracheas from SD rats were implanted into SD rats (syngeneic, n = 30) or Wistar rats (allogeneic, n = 30), and SD rat tracheas (seeded with Wistar rat-derived epithelial cells 6 days after transplantation) were transplanted into Wistar rats (seeded allogeneic, n = 30). Grafts were harvested at 7, 14, or 30 days after transplantation for histologic, quantitative reverse transcriptional polymerase chain reaction or Western blot analyses. RESULTS Syngrafts retained normal histologic structures, while the corresponding allografts demonstrated less ciliated epithelia and more lumenal occlusion. Seeding of epithelial cells ameliorated the histologic changes, reduced the expression of epithelial to mesenchymal transition (EMT)-related transcriptional factors and mesenchymal markers, and dampened the expression of transforming growth factor β1 (TGF-β1) and phosphorylation of smad3. CONCLUSION Seeding of recipient epithelial cells inhibits the progression of OAD by attenuating EMT via TGF-β-Smad signaling in rat heterotopic tracheal allografts. Clinically, the injection of recipient-originated epithelial cells might provide new insights into the treatment for OAD after tracheal allotransplantation.
Collapse
Affiliation(s)
- Xun Huang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhipei Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
30
|
Hupin C, Gohy S, Bouzin C, Lecocq M, Polette M, Pilette C. Features of mesenchymal transition in the airway epithelium from chronic rhinosinusitis. Allergy 2014; 69:1540-9. [PMID: 25104359 DOI: 10.1111/all.12503] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) defines a group of disorders characterized by persistent inflammation of the sinonasal tract. Epithelial changes and structural remodelling are present, but whether epithelial differentiation is altered remains uncertain. METHODS To evaluate the differentiation state of the sinonasal epithelium in CRS, sinonasal biopsies from patients with CRS with nasal polyps (CRSwNP) or CRS without nasal polyps (CRSsNP), or with allergic rhinitis (AR), as compared to controls, were processed by immunohistochemistry and RT-qPCR for terminal differentiation (E-cadherin, high molecular weight cytokeratins (Hmw CK) and CK5, vimentin) and lineage differentiation (ß-tubulin IV+ ciliated cells, MUC5AC+ goblet cells, p63 + basal cells). Findings were correlated with subepithelial fibrosis and clinical CT score. RESULTS Expression of E-cadherin was decreased at protein and mRNA levels in CRSwNP and CRSsNP, as compared to controls. Staining for Hmw CKs was also reduced in CRSwNP and CRSsNP, and CK5 mRNA was decreased in CRSwNP. These features were not due to changes in lineage specification, but associated with increases in vimentin-expressing epithelial cells. In addition, vimentin expression correlated with the basement membrane thickening and with CT score, as well as with tissue eosinophils. CONCLUSION Features of epithelial dedifferentiation towards a mesenchymal phenotype are observed in CRSwNP and CRSsNP and correlate with airway fibrosis and inflammation.
Collapse
Affiliation(s)
- C. Hupin
- Pole de Pneumologie, ORL & Dermatologie; Institut de Recherche Expérimentale et Clinique (IREC); Université catholique de Louvain (UCL); Brussels Belgium
| | - S. Gohy
- Pole de Pneumologie, ORL & Dermatologie; Institut de Recherche Expérimentale et Clinique (IREC); Université catholique de Louvain (UCL); Brussels Belgium
- Service de Pneumologie; Cliniques Universitaires St-Luc; Brussels Belgium
| | - C. Bouzin
- Imaging Platform; Institut de Recherche Expérimentale et Clinique (IREC); Université catholique de Louvain (UCL); Brussels Belgium
| | - M. Lecocq
- Pole de Pneumologie, ORL & Dermatologie; Institut de Recherche Expérimentale et Clinique (IREC); Université catholique de Louvain (UCL); Brussels Belgium
| | - M. Polette
- SFR CAP-Santé; INSERM UMR-S 903; University of Reims-Champagne-Ardenne; Reims France
| | - C. Pilette
- Pole de Pneumologie, ORL & Dermatologie; Institut de Recherche Expérimentale et Clinique (IREC); Université catholique de Louvain (UCL); Brussels Belgium
- Service de Pneumologie; Cliniques Universitaires St-Luc; Brussels Belgium
- Institute for Walloon Excellence in Lifesciences & Biotechnology (WELBIO); Wavre Belgium
| |
Collapse
|
31
|
Epithelial-mesenchymal transition of A549 cells is enhanced by co-cultured with THP-1 macrophages under hypoxic conditions. Biochem Biophys Res Commun 2014; 453:804-9. [PMID: 25445593 DOI: 10.1016/j.bbrc.2014.10.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 10/07/2014] [Indexed: 12/11/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is associated with pulmonary fibrosis, including idiopathic pulmonary fibrosis (IPF). In this study, we investigated EMT of human pulmonary epithelial-derived cells (A549). A549 cells was either cultured by itself or co-cultured with THP-1 macrophages under normoxic (21% O2) and hypoxic (2% O2) conditions. We evaluated the presence of EMT by determining the expression of EMT markers, E-cadherin, vimentin, and fibronectin. To determine the role of TGF-β1 and IL-1β in EMT of the A549 cells, we analyzed the effects of blocking their activity with TGF-β1 inhibitor or IL-1β neutralizing antibody respectively. The A549 cells presented EMT when they were co-cultured with THP-1 macrophages. The EMT of the A549 cells co-cultured with THP-1 macrophages was exacerbated under hypoxia. In addition, the EMT were prevented by the addition of TGF-β1 type I receptor kinase inhibitor. The hypoxic condition increased the mRNA levels of TGF-β1 in A549 cells and THP-1 macrophages and that of IL-1β in THP-1 macrophages when each cells were co-cultured. Anti-IL-1β neutralizing antibody attenuated TGF-β1 secretion in co-culture media under hypoxic conditions. Thus, the IL-1β from THP-1 macrophages up-regulated the TGF-β1 from A549 cells and THP-1 macrophages, and then the TGF-β1 from both cells induced and promoted the EMT of A549 cells when they were co-cultured under hypoxia. Together, these results demonstrate that the interaction between type II pneumocytes and macrophages under hypoxia is necessary for the development of pulmonary fibrosis.
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW To examine the recent literature on the role of innate cells in immunity to transplanted tissue. It specifically addresses the impact of monocytes/macrophages, neutrophils, natural killer cells, and platelets. RECENT FINDINGS Current research indicates that innate immunity plays a dual role in response to transplanted tissue with the ability to either facilitate rejection or promote tolerance. Intriguingly, some of these cells are even capable of reacting to allogeneic cells, a feature usually only attributed to cells of the adaptive immune system. SUMMARY This review highlights the new therapeutic targets in the innate immune system that may be useful in the treatment of transplant recipients. It also emphasizes the need to use caution in exploring these new therapeutics.
Collapse
|
33
|
Gardner A, Borthwick LA, Fisher AJ. Lung epithelial wound healing in health and disease. Expert Rev Respir Med 2014; 4:647-60. [DOI: 10.1586/ers.10.62] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
34
|
Adam D, Perotin JM, Lebargy F, Birembaut P, Deslée G, Coraux C. [Regeneration of airway epithelium]. Rev Mal Respir 2013; 31:300-11. [PMID: 24750950 DOI: 10.1016/j.rmr.2013.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 10/04/2013] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Epithelial regeneration is a complex process. It can lead to the remodeling of the airway epithelium as in asthma, COPD or cystic fibrosis. BACKGROUND The development of in vivo and in vitro models has allowed the analysis of remodeling mechanisms and showed the role of components of extracellular matrix, proteases, cytokines and growth factors. Airway epithelial progenitors and stems cells have been studied in these models. However, their identification remains difficult. CONCLUSION Identification and characterization of airway epithelial progenitor/stem-cells, and a better knowledge of the regeneration process may allow the development of new therapeutic strategies for airway epithelial reconstitution.
Collapse
Affiliation(s)
- D Adam
- Inserm UMRS 903, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France
| | - J-M Perotin
- Inserm UMRS 903, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France; Service des maladies respiratoires, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - F Lebargy
- Service des maladies respiratoires, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - P Birembaut
- Inserm UMRS 903, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France; Laboratoire d'histologie Pol Bouin, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France
| | - G Deslée
- Inserm UMRS 903, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France; Service des maladies respiratoires, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France.
| | - C Coraux
- Inserm UMRS 903, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France
| |
Collapse
|
35
|
Borthwick LA, Wynn TA, Fisher AJ. Cytokine mediated tissue fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1832:1049-60. [PMID: 23046809 PMCID: PMC3787896 DOI: 10.1016/j.bbadis.2012.09.014] [Citation(s) in RCA: 284] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 09/28/2012] [Accepted: 09/29/2012] [Indexed: 12/20/2022]
Abstract
Acute inflammation is a recognised part of normal wound healing. However, when inflammation fails to resolve and a chronic inflammatory response is established this process can become dysregulated resulting in pathological wound repair, accumulation of permanent fibrotic scar tissue at the site of injury and the failure to return the tissue to normal function. Fibrosis can affect any organ including the lung, skin, heart, kidney and liver and it is estimated that 45% of deaths in the western world can now be attributed to diseases where fibrosis plays a major aetiological role. In this review we examine the evidence that cytokines play a vital role in the acute and chronic inflammatory responses that drive fibrosis in injured tissues. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Lee A Borthwick
- Tissue Fibrosis and Repair Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK; Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | | | | |
Collapse
|
36
|
Jungraithmayr W, Jang JH, Schrepfer S, Inci I, Weder W. Small Animal Models of Experimental Obliterative Bronchiolitis. Am J Respir Cell Mol Biol 2013; 48:675-84. [DOI: 10.1165/rcmb.2012-0379tr] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
37
|
Weigt SS, DerHovanessian A, Wallace WD, Lynch JP, Belperio JA. Bronchiolitis obliterans syndrome: the Achilles' heel of lung transplantation. Semin Respir Crit Care Med 2013; 34:336-51. [PMID: 23821508 PMCID: PMC4768744 DOI: 10.1055/s-0033-1348467] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Lung transplantation is a therapeutic option for patients with end-stage pulmonary disorders. Unfortunately, chronic lung allograft dysfunction (CLAD), most commonly manifest as bronchiolitis obliterans syndrome (BOS), continues to be highly prevalent and is the major limitation to long-term survival. The pathogenesis of BOS is complex and involves alloimmune and nonalloimmune pathways. Clinically, BOS manifests as airway obstruction and dyspnea that are classically progressive and ultimately fatal; however, the course is highly variable, and distinguishable phenotypes may exist. There are few controlled studies assessing treatment efficacy, but only a minority of patients respond to current treatment modalities. Ultimately, preventive strategies may prove more effective at prolonging survival after lung transplantation, but their remains considerable debate and little data regarding the best strategies to prevent BOS. A better understanding of the risk factors and their relationship to the pathological mechanisms of chronic lung allograft rejection should lead to better pharmacological targets to prevent or treat this syndrome.
Collapse
Affiliation(s)
- S Samuel Weigt
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA.
| | | | | | | | | |
Collapse
|
38
|
McCormack N, Molloy EL, O'Dea S. Bone morphogenetic proteins enhance an epithelial-mesenchymal transition in normal airway epithelial cells during restitution of a disrupted epithelium. Respir Res 2013; 14:36. [PMID: 23509993 PMCID: PMC3607850 DOI: 10.1186/1465-9921-14-36] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 03/07/2013] [Indexed: 02/22/2023] Open
Abstract
Background Mechanisms of airway repair are poorly understood. It has been proposed that, following injury, progenitor populations such as club cells (Clara) become undifferentiated, proliferate and re-differentiate to re-epithelialise the airway. The exact phenotype of such cells during repair is unknown however. We hypothesised that airway epithelial cells (AECs) undergo some degree of epithelial-mesenchymal transition (EMT) in order to migrate over a denuded airway and effect re-epithelialisation. Furthermore, based on our previous findings that BMP signalling is an early event in AECs following injury in vivo and that BMP4 down-regulates E-cadherin expression and enhances migration in AECs in vitro, we hypothesised that BMPs could play a role in inducing such a phenotypic switch. Methods Normal AECs were isolated from mouse lungs and analysed in a model of a disrupted epithelium. EMT marker expression and BMP signalling were examined by immunofluorescence, Western blotting and RT-PCR. Results Following generation of a wound area, AECs at the wound edge migrated and acquired a mesenchymal-like morphology. E-cadherin expression was reduced in migrating cells while vimentin and α-smooth muscle actin (α-SMA) expression was increased. Re-expression of membrane E-cadherin was subsequently observed in some cells in the wound area following re-establishment of the monolayer. A transient increase in the incidence of nuclear phosphorylated Smad1/5/8 was observed in migrating cells compared with confluent cells, indicating active BMP signalling during migration. BMP antagonists noggin and gremlin inhibited cell migration, confirming the involvement of BMP signalling in migration and indicating autocrine signalling, possibly involving BMP7 or BMP4 which were expressed in AECs. Exogenous BMP2, BMP4 and BMP7 induced a mesenchymal-like morphology in AECs, enhanced the rate of cell migration and increased α-SMA protein expression in AECs. Conclusions Following disruption of an intact epithelium, migrating AECs at the wound edge acquire an EMT-like phenotype involving altered expression of E-cadherin, vimentin and α-SMA. BMP signalling is involved in AEC migration and is likely to mediate the switch towards an EMT-like phenotype by altering protein expression to facilitate cell migration and wound closure. We propose therefore that acquisition of an EMT-like phenotype by AECs is a normal aspect of wound repair. Furthermore, we suggest that diseases involving fibrosis may arise because the EMT phase of repair is prolonged by chronic injury/inflammation, rather than being caused by it, as is the current paradigm.
Collapse
Affiliation(s)
- Natasha McCormack
- Institute of Immunology, Biology Department, National University of Ireland Maynooth, Maynooth, Co, Kildare, Ireland
| | | | | |
Collapse
|
39
|
Wise SK, Den Beste KA, Hoddeson EK, Parkos CA, Nusrat A. Sinonasal epithelial wound resealing in an in vitro model: inhibition of wound closure with IL-4 exposure. Int Forum Allergy Rhinol 2013; 3:439-49. [PMID: 23468432 DOI: 10.1002/alr.21158] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 12/11/2012] [Accepted: 01/10/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND Prolonged healing and persistent inflammation following surgery for rhinosinusitis impacts patient satisfaction and healthcare resources. Cytokines interleukin (IL)-4, IL-5, and IL-13 are important mediators in T-helper 2 (Th2) inflammatory rhinosinusitis. Decreased wound healing has been demonstrated with Th2 cytokine exposure, but this has not been extensively studied in sinonasal epithelium. We hypothesized that in vitro exposure of primary sinonasal epithelial cell cultures to Th2 inflammatory cytokine IL-4 and IL-13 would impair wound resealing and decrease expression of annexin A2 at the wound edge. METHODS Following 24-hour exposure to IL-4, IL-5, or IL-13 vs controls, sterile linear mechanical wounds were created in primary sinonasal epithelial cultures (n = 12 wounds per condition). Wounds were followed for 36 hours or until complete closure, and residual wound areas were calculated by image analysis. Group differences in annexin A2 were assessed by immunofluorescence labeling, confocal microscopy, and Western blots. RESULTS Significant wound closure differences were identified across cytokine exposure groups (p < 0.001). Mean percentage wound closure at the completion of the 36-hour time course was 98.41% ± 3.43% for control wounds vs 85.02% ± 18.46% for IL-4 exposed wounds. IL-13 did not significantly impair sinonasal epithelial wound resealing in vitro. Annexin A2 protein levels were decreased in IL-4 treated wounds when compared to control wounds (p < 0.01). CONCLUSION Th2 cytokine IL-4 decreases sinonasal epithelial wound closure in vitro. Annexin A2 is also diminished with IL-4 exposure. This supports the hypothesis that IL-4 exposure impairs sinonasal epithelial wound healing and may contribute to prolonged healing in Th2 inflammatory rhinosinusitis.
Collapse
Affiliation(s)
- Sarah K Wise
- Department of Otolaryngology-Head and Neck Surgery, Emory University, Atlanta, GA 30308, USA.
| | | | | | | | | |
Collapse
|
40
|
Borthwick LA, Corris PA, Mahida R, Walker A, Gardner A, Suwara M, Johnson GE, Moisey EJ, Brodlie M, Ward C, Perry JD, De Soyza A, Mann DA, Fisher AJ. TNFα from classically activated macrophages accentuates epithelial to mesenchymal transition in obliterative bronchiolitis. Am J Transplant 2013; 13:621-33. [PMID: 23331923 DOI: 10.1111/ajt.12065] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 10/04/2012] [Accepted: 10/08/2012] [Indexed: 01/25/2023]
Abstract
Bronchiolitis obliterans syndrome is characterized by fibrotic obliteration of small airways which severely impairs graft function and survival after lung transplantation. Bronchial epithelial cells from the transplanted lung can undergo epithelial to mesenchymal transition and this can be accentuated by activated macrophages. Macrophages demonstrate significant plasticity and change phenotype in response to their microenvironment. In this study we aimed to identify secretory products from macrophages that might be therapeutic targets for limiting the inflammatory accentuation of epithelial to mesenchymal transition in bronchiolitis obliterans syndrome. TNFα, IL-1β and IL-8 are elevated in bronchoalveolar lavage from lung transplant patients prior to diagnosis of bronchiolitis obliterans syndrome. Classically activated macrophages secrete more TNFα and IL-1β than alternatively activated macrophages and dramatically accentuate TGF-β1-driven epithelial to mesenchymal transition in bronchial epithelial cells isolated from lung transplant patients. Blocking TNFα, but not IL-1β, inhibits the accentuation of epithelial to mesenchymal transition. In a pilot unblinded therapeutic intervention in five patients with progressive bronchiolitis obliterans syndrome, anti-TNFα treatment improved forced expiratory volume in 1 second and 6-min walk distances in four patients. Our data identify TNFα as a potential new therapeutic target in bronchiolitis obliterans syndrome deserving of a randomized placebo controlled clinical trial.
Collapse
Affiliation(s)
- L A Borthwick
- Tissue Fibrosis and Repair Group, Institute of Cellular Medicine, Newcastle University, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Vittal R, Fan L, Greenspan DS, Mickler EA, Gopalakrishnan B, Gu H, Benson HL, Zhang C, Burlingham W, Cummings OW, Wilkes DS. IL-17 induces type V collagen overexpression and EMT via TGF-β-dependent pathways in obliterative bronchiolitis. Am J Physiol Lung Cell Mol Physiol 2012; 304:L401-14. [PMID: 23262228 DOI: 10.1152/ajplung.00080.2012] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Obliterative bronchiolitis (OB), a fibrotic airway lesion, is the leading cause of death after lung transplantation. Type V collagen [col(V)] overexpression and IL-17-mediated anti-col(V) immunity are key contributors to OB pathogenesis. Here, we report a previously undefined role of IL-17 in inducing col(V) overexpression, leading to epithelial mesenchymal transition (EMT) and subsequent OB. We observed IL-17-mediated induction of col(V) α1 chains [α1 (V)] in normal airway epithelial cells in vitro and detected α1 (V)-specific antibodies in bronchoalveolar lavage fluid of lung transplant patients. Overexpression of IL-17 and col(V) was detected in OB lesions in patient lung biopsies and in a murine OB model. IL-17 is shown to induce EMT, TGF-β mRNA expression, and SMAD3 activation, whereas downregulating SMAD7 expression in vitro. Pharmacological inhibition of TGF-βRI tyrosine kinase, p38 MAPK, or focal adhesion kinase prevented col(V) overexpression and EMT. In murine orthotopic lung transplants, neutralizing IL-17 significantly decreased TGF-β mRNA and protein expression and prevented epithelial repair/OB. Our findings highlight a feed-forward loop between IL-17 and TGF-β, leading to induction of col(V) and associated epithelial repair, thus providing one possible link between autoimmunity and OB after lung transplantation.
Collapse
Affiliation(s)
- Ragini Vittal
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Banerjee B, Musk M, Sutanto EN, Yerkovich ST, Hopkins P, Knight DA, Lindsey-Temple S, Stick SM, Kicic A, Chambers DC. Regional differences in susceptibiity of bronchial epithelium to mesenchymal transition and inhibition by the macrolide antibiotic azithromycin. PLoS One 2012; 7:e52309. [PMID: 23284981 PMCID: PMC3528745 DOI: 10.1371/journal.pone.0052309] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Accepted: 11/16/2012] [Indexed: 12/11/2022] Open
Abstract
Objective Dysregulated repair following epithelial injury is a key forerunner of disease in many organs, and the acquisition of a mesenchymal phenotype by the injured epithelial cells (epithelial to mesenchymal transition, EMT) may serve as a source of fibrosis. The macrolide antibiotic azithromycin and the DNA synthesis inhibitor mycophenolate are in clinical use but their mechanism of action remains unknown in post-transplant bronchiolitis obliterans syndrome (BOS). Here we determined if regional variation in the EMT response to TGFβ1 underlies the bronchiolocentric fibrosis leading to BOS and whether EMT could be inhibited by azithromycin or mycophenolate. Methods/Results We found that small and large airway epithelial cells from stable lung transplant patients underwent EMT when stimulated with TGFβ1, however mesenchymal protein expression was higher and loss of epithelial protein expression more complete in small airway epithelial cells. This regional difference was not mediated by changes in expression of the TGFβRII or Smad3 activation. Azithromycin potentially inhibited EMT in both small and large airway epithelial cells by inhibiting Smad3 expression, but not activation. Conclusion Collectively, these observations provide a biologic basis for a previously unexplained but widely observed clinical phenomena, and a platform for the development of new approaches to fibrotic diseases.
Collapse
Affiliation(s)
- Balarka Banerjee
- School of Paediatrics and Child Health, the University of Western Australia, Nedlands, Western Australia, Australia
- School of Medicine and Pharmacology, the University of Western Australia, Nedlands, Western Australia, Australia
- Western Australia Lung Transplant Program, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Michael Musk
- Western Australia Lung Transplant Program, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Erika N. Sutanto
- Telethon Institute for Child Health Research, Centre for Child Health Research, the University of Western Australia, Subiaco, Western Australia, Australia
| | | | - Peter Hopkins
- School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Darryl A. Knight
- University of British Columbia, James Hogg Research Centre for Cardiovascular and Pulmonary Research, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Suzanna Lindsey-Temple
- Centre for Asthma and Allergy Research Institute (CAARR) The Lung Institute of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Stephen M. Stick
- School of Paediatrics and Child Health, the University of Western Australia, Nedlands, Western Australia, Australia
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
- Telethon Institute for Child Health Research, Centre for Child Health Research, the University of Western Australia, Subiaco, Western Australia, Australia
| | - Anthony Kicic
- School of Paediatrics and Child Health, the University of Western Australia, Nedlands, Western Australia, Australia
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
- Telethon Institute for Child Health Research, Centre for Child Health Research, the University of Western Australia, Subiaco, Western Australia, Australia
| | - Daniel C. Chambers
- School of Medicine, The University of Queensland, Herston, Queensland, Australia
- Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
43
|
Anti-inflammatory and immunomodulatory properties of azithromycin involved in treatment and prevention of chronic lung allograft rejection. Transplantation 2012; 94:101-9. [PMID: 22461039 DOI: 10.1097/tp.0b013e31824db9da] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Chronic lung allograft rejection is the single most important cause of death in lung transplant recipients after the first postoperative year, resulting in a 5-year survival rate of approximately 50%, which is far behind that of other solid organ transplantations. Spirometry is routinely used as a clinical marker for assessing pulmonary allograft function and diagnosing chronic lung allograft rejection after lung transplantation (LTx). As such, a progressive obstructive decline in pulmonary allograft function (forced expiratory volume in 1 sec [FEV1]) in absence of all other causes (currently defined as bronchiolitis obliterans syndrome [BOS]) is considered to reflect the evolution of chronic lung allograft rejection. BOS has a 5-year prevalence of approximately 45% and is thought to be the final common endpoint of various alloimmunologic and nonalloimmunologic injuries to the pulmonary allograft, triggering different innate and adaptive immune responses. Most preventive and therapeutic strategies for this complex process have thus far been largely unsuccessful. However, the introduction of the neomacrolide antibiotic azithromycin (AZI) in the field of LTx as of 2003 made it clear that some patients with established BOS might in fact benefit from such therapy due to its various antiinflammatory and immunomodulatory properties, as summarized in this review. Particularly in patients with an increased bronchoalveolar lavage neutrophilia (i.e., 15%-20% or more), AZI treatment could result in an increase in FEV1 of at least 10%. More recently, it has become clear that prophylactic therapy with AZI actually may prevent BOS and improve FEV1 after LTx, most likely through its interactions with the innate immune system. However, one should always be aware of possible adverse effects related to AZI when implementing this drug as prophylactic or long-term treatment. Even so, AZI therapy after LTx can generally be considered as safe.
Collapse
|
44
|
Epithelial-mesenchymal transition in the pathophysiology of airway remodelling in asthma. Curr Opin Allergy Clin Immunol 2012; 12:53-9. [PMID: 22217512 DOI: 10.1097/aci.0b013e32834ec6eb] [Citation(s) in RCA: 156] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW We currently understand little about the mechanisms that lead to airway remodeling in asthma. The origin of the mesenchymal cells that contribute to fibrosis of the airway is poorly understood. However, emerging evidence suggests that the airway epithelium could contribute to airway remodeling through the process of epithelial-mesenchymal transition (EMT) following environmental challenge. In this review, we will discuss the mechanistic features of EMT and highlight recent descriptions of EMT in the airway to further define the role of the airway epithelium in the pathogenesis of asthma. RECENT FINDINGS Growth factors, inflammatory mediators, and matricellular proteins expressed following exposure to environmental insults are known to induce downregulation of epithelial cell-cell adhesions and promote mesenchymal gene expression programs both in vitro and in vivo. These results demonstrate that the plastic and dynamic airway epithelium may contribute to airway remodeling via EMT in asthma. SUMMARY It is becoming increasingly clear that the airway epithelium orchestrates inflammatory and remodeling responses of the airway. Understanding the regulatory mechanisms involved in epithelial plasticity will be crucial to determine effective therapies to halt the progression of airway remodeling in asthma.
Collapse
|
45
|
Gardner A, Fisher AJ, Richter C, Johnson GE, Moisey EJ, Brodlie M, Ward C, Krippner-Heidenreich A, Mann DA, Borthwick LA. The critical role of TAK1 in accentuated epithelial to mesenchymal transition in obliterative bronchiolitis after lung transplantation. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:2293-308. [PMID: 22525462 DOI: 10.1016/j.ajpath.2012.02.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 01/20/2012] [Accepted: 02/07/2012] [Indexed: 01/13/2023]
Abstract
Therapies to limit or reverse fibrosis have proven unsuccessful, highlighting the need for a greater understanding of basic mechanisms that drive fibrosis and, in particular, the link between fibrosis and inflammation. It has been shown that pro-fibrotic transforming growth factor β1 (TGF-β1)-driven epithelial-to-mesenchymal transition (EMT) can be accentuated by tumor necrosis factor α (TNF-α). TGF-β-activated kinase 1 (TAK1) is activated by both TGF-β1 and TNF-α, activating both nuclear factor kappa-light-chain-enhancer of activated B cells and mitogen-activated protein kinase signaling pathways. In this study, we evaluated the potential for TAK1 to modulate the synergistic effect between TGF-β1 and TNF-α in driving EMT. Co-stimulation with TGF-β1 and TNF-α induced an accentuated and extended phosphorylation of TAK1 compared to either alone. TAK1 signaled downstream via nuclear factor kappa-light-chain-enhancer of activated B cells, and Jun N-terminal kinase-2, but independent of Jun N-terminal kinase-1, extracellular signal-regulated kinase-1/2, or p38 mitogen-activated protein kinase signaling to drive EMT in bronchial epithelial cells. Blocking either TAK1 or Jun N-terminal kinase-2 inhibited EMT. TAK1 phosphorylation was increased in the airway epithelium of patients with fibrotic airway disease. These data identify factors leading to and affected by accentuated and extended TAK1 phosphorylations potential novel therapeutic targets in inflammation-driven fibrotic diseases.
Collapse
Affiliation(s)
- Aaron Gardner
- Tissue Fibrosis & Repair Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gorowiec MR, Borthwick LA, Parker SM, Kirby JA, Saretzki GC, Fisher AJ. Free radical generation induces epithelial-to-mesenchymal transition in lung epithelium via a TGF-β1-dependent mechanism. Free Radic Biol Med 2012; 52:1024-32. [PMID: 22240154 DOI: 10.1016/j.freeradbiomed.2011.12.020] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 12/20/2011] [Accepted: 12/21/2011] [Indexed: 01/14/2023]
Abstract
Fibrotic remodelling of lung parenchymal and airway compartments is the major contributor to life-threatening organ dysfunction in chronic lung diseases such as idiopathic pulmonary fibrosis (IPF) and Chronic Obstructive Pulmonary Disease (COPD). Since transforming growth factor-β1 (TGF-β1) is believed to play a key role in disease pathogenesis and markers of oxidative stress are also commonly detected in bronchoalveolar lavage (BAL) from such patients we sought to investigate whether both factors might be interrelated. Here we investigated the hypothesis that oxidative stress to the lung epithelium promotes fibrotic repair by driving epithelial-to-mesenchymal transition (EMT) via the augmentation of TGF-β1. We show that in response to 400μM hydrogen peroxide (H(2)O(2)) A549 cells, used a model for alveolar epithelium, and human primary bronchial epithelial cells (PBECs) undergo EMT displaying morphology changes, decreased expression of epithelial markers (E-cadherin and ZO-1), increased expression of mesenchymal markers (vimentin and α-smooth muscle actin) as well as increased secretion of extracelluar matrix components. The same oxidative stress also promotes expression of TGF-β1. Inhibition of TGF-β1 signalling as well as treatment with antioxidants such as phenyl tert-butylnitrone (PBN) and superoxide dismutase 3 (SOD3) prevent the oxidative stress driven EMT-like changes described above. Interventions also inhibited EMT-like changes. This study identifies a link between oxidative stress, TGF-β1 and EMT in lung epithelium and highlights the potential for antioxidant therapies to limit EMT and its potential contribution to chronic lung disease.
Collapse
Affiliation(s)
- Marta R Gorowiec
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Newcastle University, UK
| | | | | | | | | | | |
Collapse
|
47
|
Kawata M, Koinuma D, Ogami T, Umezawa K, Iwata C, Watabe T, Miyazono K. TGF-β-induced epithelial-mesenchymal transition of A549 lung adenocarcinoma cells is enhanced by pro-inflammatory cytokines derived from RAW 264.7 macrophage cells. J Biochem 2011; 151:205-16. [PMID: 22161143 DOI: 10.1093/jb/mvr136] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cancer cells undergo epithelial-mesenchymal transition (EMT) during invasion and metastasis. Although transforming growth factor-β (TGF-β) and pro-inflammatory cytokines have been implicated in EMT, the underlying molecular mechanisms remain to be elucidated. Here, we studied the effects of proinflammatory cytokines derived from the mouse macrophage cell line RAW 264.7 on TGF-β-induced EMT in A549 lung cancer cells. Co-culture and treatment with conditioned medium of RAW 264.7 cells enhanced a subset of TGF-β-induced EMT phenotypes in A549 cells, including changes in cell morphology and induction of mesenchymal marker expression. These effects were increased by the treatment of RAW 264.7 cells with lipopolysaccharide, which also induced the expression of various proinflammatory cytokines, including TNF-α and IL-1β. The effects of conditioned medium of RAW 264.7 cells were partially inhibited by a TNF-α neutralizing antibody. Dehydroxy methyl epoxyquinomicin, a selective inhibitor of NFκB, partially inhibited the enhancement of fibronectin expression by TGF-β, TNF-α, and IL-1β, but not of N-cadherin expression. Effects of other pharmacological inhibitors also suggested complex regulatory mechanisms of the TGF-β-induced EMT phenotype by TNF-α stimulation. These findings provide direct evidence of the effects of RAW 264.7-derived TNF-α on TGF-β-induced EMT in A549 cells, which is transduced in part by NFκB signalling.
Collapse
Affiliation(s)
- Mikiko Kawata
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Transforming Growth Factor-β1 (TGF-β1) Driven Epithelial to Mesenchymal Transition (EMT) is Accentuated by Tumour Necrosis Factor α (TNFα) via Crosstalk Between the SMAD and NF-κB Pathways. CANCER MICROENVIRONMENT 2011; 5:45-57. [PMID: 21792635 DOI: 10.1007/s12307-011-0080-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 07/18/2011] [Indexed: 12/15/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is a process by which an epithelial cell alters its phenotype to that of a mesenchymal cell and plays a critical role in embryonic development, tumour invasion and metastasis and tissue fibrosis. Transforming growth factor-β1 (TGF-β1) continues to be regarded as the key growth factor involved in driving EMT however recently tumour necrosis factor α (TNFα) has been demonstrated to accentuate TGF-β1 driven EMT. In this study we investigate how various signalling pathways contribute to this accentuated effect. A549 cells were treated with TGF-β1 (10 ng/ml), TNFα (20 ng/ml) or a combination of both for 72 h and EMT assessed. The effect of selective inhibition of the SMAD, MAPK and NF-κB pathways on EMT was assessed. A549 cells treated with TGF-β1 downregulate the expression of epithelial markers, increase the expression of mesenchymal markers, secrete matrix-metalloproteinases and become invasive. Significantly, TGF-β1 driven EMT is accentuated by co-treatment with TNFα. SMAD 3 inhibition attenuated TGF-β1 driven EMT but has no effect on the accentuation effect of TNFα. However, inhibiting IKKβ blocked both TGF-β1 driven EMT and the accentuating action of TNFα. Inhibiting p38 and ERK signalling had no effect on EMT. TNFα accentuates TGF-β1 driven EMT in A549 cells via a SMAD 2/3 independent mechanism involving the NF-κB pathway independent of p38 and ERK 1/2 activation.
Collapse
|
49
|
Ornellas DS, Maron-Gutierrez T, Ornellas FM, Cruz FF, Oliveira GP, Lucas IH, Fujisaki L, Oliveira MG, Teodoro WR, Capelozzi VL, Pelosi P, Morales MM, Rocco PRM. Early and late effects of bone marrow-derived mononuclear cell therapy on lung and distal organs in experimental sepsis. Respir Physiol Neurobiol 2011; 178:304-14. [PMID: 21763473 DOI: 10.1016/j.resp.2011.06.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 06/23/2011] [Accepted: 06/30/2011] [Indexed: 01/01/2023]
Abstract
We tested the hypothesis that bone marrow-derived mononuclear cells (BMDMCs) at an early phase of cecal ligation and puncture (CLP)-induced sepsis may have lasting effects on: (1) lung mechanics and histology, (2) the structural remodelling of lung parenchyma, (3) lung, kidney, and liver cell apoptosis, and (4) pro- and anti-inflammatory cytokines and growth factors. At day 1, BMDMC significantly reduced mortality, as well as caspase-3, interleukin (IL)-6 and IL-1β, vascular endothelial growth factor, platelet-derived growth factor, hepatocyte growth factor, and transforming growth factor-β, but increased IL-10 mRNA expression in lung tissue in septic mice contributing to endothelium and epithelium alveolar repair and improvement of lung mechanics. BMDMC also prevented the increase of apoptotic cells in lung, liver, and kidney. At day 7, these early functional and morphological effects were preserved or further improved. In conclusion, in the present model of sepsis, the beneficial effects of early administration of BMDMCs on lung and distal organs were preserved, possibly by paracrine mechanisms.
Collapse
Affiliation(s)
- Debora S Ornellas
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Floreth T, Stern E, Tu Y, Stern R, Garrity ER, Bhorade SM, White SR. Differentiated transplant derived airway epithelial cell cytokine secretion is not regulated by cyclosporine. Respir Res 2011; 12:44. [PMID: 21477368 PMCID: PMC3079624 DOI: 10.1186/1465-9921-12-44] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 04/10/2011] [Indexed: 11/25/2022] Open
Abstract
Background While lung transplantation is an increasingly utilized therapy for advanced lung diseases, chronic rejection in the form of Bronchiolitis Obliterans Syndrome (BOS) continues to result in significant allograft dysfunction and patient mortality. Despite correlation of clinical events with eventual development of BOS, the causative pathophysiology remains unknown. Airway epithelial cells within the region of inflammation and fibrosis associated with BOS may have a participatory role. Methods Transplant derived airway epithelial cells differentiated in air liquid interface culture were treated with IL-1β and/or cyclosporine, after which secretion of cytokines and growth factor and gene expression for markers of epithelial to mesenchymal transition were analyzed. Results Secretion of IL-6, IL-8, and TNF-α, but not TGF-β1, was increased by IL-1β stimulation. In contrast to previous studies using epithelial cells grown in submersion culture, treatment of differentiated cells in ALI culture with cyclosporine did not elicit cytokine or growth factor secretion, and did not alter IL-6, IL-8, or TNF-α production in response to IL-1β treatment. Neither IL-1β nor cyclosporine elicited expression of markers of the epithelial to mesenchymal transition E-cadherin, EDN-fibronectin, and α-smooth muscle actin. Conclusion Transplant derived differentiated airway epithelial cell IL-6, IL-8, and TNF-α secretion is not regulated by cyclosporine in vitro; these cells thus may participate in local inflammatory responses in the setting of immunosuppression. Further, treatment with IL-1β did not elicit gene expression of markers of epithelial to mesenchymal transition. These data present a model of differentiated airway epithelial cells that may be useful in understanding epithelial participation in airway inflammation and allograft rejection in lung transplantation.
Collapse
Affiliation(s)
- Timothy Floreth
- Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | |
Collapse
|