1
|
He X, Liu P, Luo Y, Fu X, Yang T. STATs, promising targets for the treatment of autoimmune and inflammatory diseases. Eur J Med Chem 2024; 277:116783. [PMID: 39180944 DOI: 10.1016/j.ejmech.2024.116783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Cytokines play a crucial role in the pathophysiology of autoimmune and inflammatory diseases, with over 50 cytokines undergoing signal transduction through the Signal Transducers and Activators of Transcription (STAT) signaling pathway. Recent studies have solidly confirmed the pivotal role of STATs in autoimmune and inflammatory diseases. Therefore, this review provides a detailed summary of the immunological functions of STATs, focusing on exploring their mechanisms in various autoimmune and inflammatory diseases. Additionally, with the rapid advancement of structural biology in the field of drug discovery, many STAT inhibitors have been identified using structure-based drug design strategies. In this review, we also examine the structures of STAT proteins and compile the latest research on STAT inhibitors currently being tested in animal models and clinical trials for the treatment of immunological diseases, which emphasizes the feasibility of STATs as promising therapeutic targets and provides insights into the design of the next generation of STAT inhibitors.
Collapse
Affiliation(s)
- Xinlian He
- Laboratory of Human Diseases and Immunotherapy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pingxian Liu
- Laboratory of Human Diseases and Immunotherapy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Youfu Luo
- Laboratory of Human Diseases and Immunotherapy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyuan Fu
- Laboratory of Human Diseases and Immunotherapy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Yang
- Laboratory of Human Diseases and Immunotherapy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Aghajani Mir M. Illuminating the pathogenic role of SARS-CoV-2: Insights into competing endogenous RNAs (ceRNAs) regulatory networks. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 122:105613. [PMID: 38844190 DOI: 10.1016/j.meegid.2024.105613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/20/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
The appearance of SARS-CoV-2 in 2019 triggered a significant economic and health crisis worldwide, with heterogeneous molecular mechanisms that contribute to its development are not yet fully understood. Although substantial progress has been made in elucidating the mechanisms behind SARS-CoV-2 infection and therapy, it continues to rank among the top three global causes of mortality due to infectious illnesses. Non-coding RNAs (ncRNAs), being integral components across nearly all biological processes, demonstrate effective importance in viral pathogenesis. Regarding viral infections, ncRNAs have demonstrated their ability to modulate host reactions, viral replication, and host-pathogen interactions. However, the complex interactions of different types of ncRNAs in the progression of COVID-19 remains understudied. In recent years, a novel mechanism of post-transcriptional gene regulation known as "competing endogenous RNA (ceRNA)" has been proposed. Long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and viral ncRNAs function as ceRNAs, influencing the expression of associated genes by sequestering shared microRNAs. Recent research on SARS-CoV-2 has revealed that disruptions in specific ceRNA regulatory networks (ceRNETs) contribute to the abnormal expression of key infection-related genes and the establishment of distinctive infection characteristics. These findings present new opportunities to delve deeper into the underlying mechanisms of SARS-CoV-2 pathogenesis, offering potential biomarkers and therapeutic targets. This progress paves the way for a more comprehensive understanding of ceRNETs, shedding light on the intricate mechanisms involved. Further exploration of these mechanisms holds promise for enhancing our ability to prevent viral infections and develop effective antiviral treatments.
Collapse
Affiliation(s)
- Mahsa Aghajani Mir
- Deputy of Research and Technology, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
3
|
Mutsaers SE, Miles T, Prêle CM, Hoyne GF. Emerging role of immune cells as drivers of pulmonary fibrosis. Pharmacol Ther 2023; 252:108562. [PMID: 37952904 DOI: 10.1016/j.pharmthera.2023.108562] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
The pathogenesis of pulmonary fibrosis, including idiopathic pulmonary fibrosis (IPF) and other forms of interstitial lung disease, involves a complex interplay of various factors including host genetics, environmental pollutants, infection, aberrant repair and dysregulated immune responses. Highly variable clinical outcomes of some ILDs, in particular IPF, have made it difficult to identify the precise mechanisms involved in disease pathogenesis and thus the development of a specific cure or treatment to halt and reverse the decline in patient health. With the advent of in-depth molecular diagnostics, it is becoming evident that the pathogenesis of IPF is unlikely to be the same for all patients and therefore will likely require different treatment approaches. Chronic inflammation is a cardinal feature of IPF and is driven by both innate and adaptive immune responses. Inflammatory cells and activated fibroblasts secrete various pro-inflammatory cytokines and chemokines that perpetuate the inflammatory response and contribute to the recruitment and activation of more immune cells and fibroblasts. The balance between pro-inflammatory and regulatory immune cell subsets, as well as the interactions between immune cell types and resident cells within the lung microenvironment, ultimately determines the extent of fibrosis and the potential for resolution. This review examines the role of the innate and adaptive immune responses in pulmonary fibrosis, with an emphasis on IPF. The role of different immune cell types is discussed as well as novel anti-inflammatory and immunotherapy approaches currently in clinical trial or in preclinical development.
Collapse
Affiliation(s)
- Steven E Mutsaers
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia.
| | - Tylah Miles
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia
| | - Cecilia M Prêle
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia; School of Medical, Molecular and Forensic Sciences, Murdoch University, WA, Australia
| | - Gerard F Hoyne
- Institute for Respiratory Health, The University of Western Australia, Nedlands, WA, Australia; The School of Health Sciences and Physiotherapy, University of Notre Dame Australia, Fremantle, WA, Australia
| |
Collapse
|
4
|
Curras-Alonso S, Soulier J, Defard T, Weber C, Heinrich S, Laporte H, Leboucher S, Lameiras S, Dutreix M, Favaudon V, Massip F, Walter T, Mueller F, Londoño-Vallejo JA, Fouillade C. An interactive murine single-cell atlas of the lung responses to radiation injury. Nat Commun 2023; 14:2445. [PMID: 37117166 PMCID: PMC10147670 DOI: 10.1038/s41467-023-38134-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 04/17/2023] [Indexed: 04/30/2023] Open
Abstract
Radiation Induced Lung Injury (RILI) is one of the main limiting factors of thorax irradiation, which can induce acute pneumonitis as well as pulmonary fibrosis, the latter being a life-threatening condition. The order of cellular and molecular events in the progression towards fibrosis is key to the physiopathogenesis of the disease, yet their coordination in space and time remains largely unexplored. Here, we present an interactive murine single cell atlas of the lung response to irradiation, generated from C57BL6/J female mice. This tool opens the door for exploration of the spatio-temporal dynamics of the mechanisms that lead to radiation-induced pulmonary fibrosis. It depicts with unprecedented detail cell type-specific radiation-induced responses associated with either lung regeneration or the failure thereof. A better understanding of the mechanisms leading to lung fibrosis will help finding new therapeutic options that could improve patients' quality of life.
Collapse
Affiliation(s)
- Sandra Curras-Alonso
- Institut Curie, CNRS UMR 3244, Sorbonne Universite, PSL University, 75005, Paris, France
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL University, Centre Universitaire, 91405, Orsay Cedex, France
| | - Juliette Soulier
- Institut Curie, CNRS UMR 3244, Sorbonne Universite, PSL University, 75005, Paris, France
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL University, Centre Universitaire, 91405, Orsay Cedex, France
| | - Thomas Defard
- Centre for Computational Biology (CBIO), Mines Paris, PSL University, 75006, Paris, France
- Institut Curie, PSL University, 75005, Paris, France
- INSERM, U900, 75005, Paris, France
- Imaging and Modeling Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Christian Weber
- Imaging and Modeling Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Sophie Heinrich
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL University, Centre Universitaire, 91405, Orsay Cedex, France
| | - Hugo Laporte
- Institut Curie, CNRS UMR 3244, Sorbonne Universite, PSL University, 75005, Paris, France
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL University, Centre Universitaire, 91405, Orsay Cedex, France
| | - Sophie Leboucher
- Institut Curie, CNRS UMR 3348, University Paris-Saclay, PSL University, Centre Universitaire, Orsay, France
| | - Sonia Lameiras
- Institut Curie Genomics of Excellence (ICGex) Platform, Paris, France
| | - Marie Dutreix
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL University, Centre Universitaire, 91405, Orsay Cedex, France
| | - Vincent Favaudon
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL University, Centre Universitaire, 91405, Orsay Cedex, France
| | - Florian Massip
- Centre for Computational Biology (CBIO), Mines Paris, PSL University, 75006, Paris, France
- Institut Curie, PSL University, 75005, Paris, France
- INSERM, U900, 75005, Paris, France
| | - Thomas Walter
- Centre for Computational Biology (CBIO), Mines Paris, PSL University, 75006, Paris, France
- Institut Curie, PSL University, 75005, Paris, France
- INSERM, U900, 75005, Paris, France
| | - Florian Mueller
- Imaging and Modeling Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - José-Arturo Londoño-Vallejo
- Institut Curie, CNRS UMR 3244, Sorbonne Universite, PSL University, 75005, Paris, France.
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL University, Centre Universitaire, 91405, Orsay Cedex, France.
| | - Charles Fouillade
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL University, Centre Universitaire, 91405, Orsay Cedex, France.
| |
Collapse
|
5
|
Li H, Zhao C, Muhetaer G, Guo L, Yao K, Zhang G, Ji Y, Xing S, Zhou J, Huang X. Integrated RNA-sequencing and network pharmacology approach reveals the protection of Yiqi Huoxue formula against idiopathic pulmonary fibrosis by interfering with core transcription factors. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154301. [PMID: 35792448 DOI: 10.1016/j.phymed.2022.154301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/13/2022] [Accepted: 06/26/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a refractory disease. Therefore, developing effective therapies for IPF is the need of the hour. PURPOSE Yiqi Huoxue Formula (YQHX) is an herbal formula comprising three herbal medicines: Ligusticum chuanxiong Hort. (Chuanxiong Rhizoma, CR), Panax notoginseng (Burk.) F. H. Chen (Notoginseng Radix Et Rhizoma, NR) and Panax ginseng C. A. Mey. (Ginseng Radix Et Rhizoma, GR). This study aims to determine the anti-pulmonary fibrosis effect of YQHX and explore its mechanism of action. STUDY Design and Methods: The chemical components in the GR, CR and NR extracts were identified by High Performance Liquid Chromatography. A TGF-β1-induced myofibroblast cell model was used to test the anti-fibrosis effect of GR, CR, NR and YQHX. RNA-sequencing was used to identify the differentially expressed genes (DEGs) after YQHX treatment. Subsequently, gene enrichment analysis and key transcription factors (TFs) prediction for YQHX-regulated DEGs was performed. The active constituents of GR, CR and NR were obtained from the Traditional Chinese Medicine Database and Analysis Platform. Targets of the active constituents were predicted using the similarity ensemble approach search server and Swiss Target Prediction tool. YQHX-targeted key TFs that transcribed the DEGs were screened out. Then, the effect of YQHX on the bleomycin-induced pulmonary fibrosis mouse model was studied. Finally, one of the predicted TFs, STAT3, was selected to validate the prediction accuracy. RESULTS Seven, two, and five compounds were identified in the GR, CR, and NR extracts, respectively. YQHX and its constituents-GR, CR and NR-inhibited the expression of fibrotic markers, including α -SMA and fibronectin, indicating that YQHX inhibited TGF-β1-induced myofibroblast activation. RNA-sequencing identified 291 genes that were up-regulated in the TGF-β1 group but down-regulated after YQHX treatment. In total, 55 key TFs that transcribed YQHX-regulated targets were predicted. A regulatory network of 24 active ingredients and 232 corresponding targets for YQHX was established. Among YQHX's predicted targets, 20 were TFs. On overlapping YQHX-targeted TFs and DEGs' key TFs, six key TFs, including HIF1A, STAT6, STAT3, PPARA, DDIT3 and AR, were identified as the targets of YQHX. Additionally, YQHX alleviated bleomycin-induced pulmonary fibrosis in a mouse model by inhibiting the phosphorylation of STAT3 in the lungs of pulmonary fibrosis mice. CONCLUSIONS This study provides pharmacological support for the use of YQHX in the treatment of IPF. The potential mechanism of action of YQHX is speculated to involve the modulation of core TFs and inhibition of pathogenetic gene expressions in IPF.
Collapse
Affiliation(s)
- Hang Li
- Department of Respiratory Medicine, Central lab, Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Yu'an Second Road, No. 21, Shenzhen 518133, China.
| | - Caiping Zhao
- Department of Respiratory Medicine, Central lab, Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Yu'an Second Road, No. 21, Shenzhen 518133, China
| | - Gulizeba Muhetaer
- Department of Respiratory Medicine, Central lab, Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Yu'an Second Road, No. 21, Shenzhen 518133, China
| | - Longgang Guo
- Guangzhou Chromap Biotechnology Co., Ltd., Guangzhou 510700, China
| | - Kainan Yao
- Department of Respiratory Medicine, Central lab, Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Yu'an Second Road, No. 21, Shenzhen 518133, China
| | - Guiyu Zhang
- Department of Respiratory Medicine, Central lab, Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Yu'an Second Road, No. 21, Shenzhen 518133, China
| | - Yichun Ji
- Department of Respiratory Medicine, Central lab, Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Yu'an Second Road, No. 21, Shenzhen 518133, China
| | - Sizhong Xing
- Department of Respiratory Medicine, Central lab, Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Yu'an Second Road, No. 21, Shenzhen 518133, China
| | - Jihong Zhou
- Department of Respiratory Medicine, Central lab, Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Yu'an Second Road, No. 21, Shenzhen 518133, China.
| | - Xiufang Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Jichang Road, No. 12, Guangzhou 510405, China.
| |
Collapse
|
6
|
Wieczfinska J, Pawliczak R. Relaxin Affects Airway Remodeling Genes Expression through Various Signal Pathways Connected with Transcription Factors. Int J Mol Sci 2022; 23:ijms23158413. [PMID: 35955554 PMCID: PMC9368845 DOI: 10.3390/ijms23158413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/27/2023] Open
Abstract
Fibrosis is one of the parameters of lung tissue remodeling in asthma. Relaxin has emerged as a natural suppressor of fibrosis, showing efficacy in the prevention of a multiple models of fibrosis. Therefore, the aim of this study was to analyze the aptitudes of relaxin, in the context of its immunomodulatory properties, in the development of airway remodeling. WI-38 and HFL1 fibroblasts, as well as epithelial cells (NHBE), were incubated with relaxin. Additionally, remodeling conditions were induced with two serotypes of rhinovirus (HRV). The expression of the genes contributing to airway remodeling were determined. Moreover, NF-κB, c-Myc, and STAT3 were knocked down to analyze the pathways involved in airway remodeling. Relaxin decreased the mRNA expression of collagen I and TGF-β and increased the expression of MMP-9 (p < 0.05). Relaxin also decreased HRV-induced expression of collagen I and α-SMA (p < 0.05). Moreover, all the analyzed transcription factors—NF-κB, c-Myc, and STAT3—have shown its influence on the pathways connected with relaxin action. Though relaxin requires further study, our results suggest that this natural compound offers great potential for inhibition of the development, or even reversing, of factors related to airway remodeling. The presented contribution of the investigated transcription factors in this process additionally increases its potential possibilities through a variety of its activity pathways.
Collapse
|
7
|
Korfei M, Mahavadi P, Guenther A. Targeting Histone Deacetylases in Idiopathic Pulmonary Fibrosis: A Future Therapeutic Option. Cells 2022; 11:1626. [PMID: 35626663 PMCID: PMC9139813 DOI: 10.3390/cells11101626] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease with limited therapeutic options, and there is a huge unmet need for new therapies. A growing body of evidence suggests that the histone deacetylase (HDAC) family of transcriptional corepressors has emerged as crucial mediators of IPF pathogenesis. HDACs deacetylate histones and result in chromatin condensation and epigenetic repression of gene transcription. HDACs also catalyse the deacetylation of many non-histone proteins, including transcription factors, thus also leading to changes in the transcriptome and cellular signalling. Increased HDAC expression is associated with cell proliferation, cell growth and anti-apoptosis and is, thus, a salient feature of many cancers. In IPF, induction and abnormal upregulation of Class I and Class II HDAC enzymes in myofibroblast foci, as well as aberrant bronchiolar epithelium, is an eminent observation, whereas type-II alveolar epithelial cells (AECII) of IPF lungs indicate a significant depletion of many HDACs. We thus suggest that the significant imbalance of HDAC activity in IPF lungs, with a "cancer-like" increase in fibroblastic and bronchial cells versus a lack in AECII, promotes and perpetuates fibrosis. This review focuses on the mechanisms by which Class I and Class II HDACs mediate fibrogenesis and on the mechanisms by which various HDAC inhibitors reverse the deregulated epigenetic responses in IPF, supporting HDAC inhibition as promising IPF therapy.
Collapse
Affiliation(s)
- Martina Korfei
- Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, D-35392 Giessen, Germany; (P.M.); (A.G.)
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), D-35392 Giessen, Germany
| | - Poornima Mahavadi
- Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, D-35392 Giessen, Germany; (P.M.); (A.G.)
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), D-35392 Giessen, Germany
| | - Andreas Guenther
- Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, D-35392 Giessen, Germany; (P.M.); (A.G.)
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), D-35392 Giessen, Germany
- Lung Clinic, Evangelisches Krankenhaus Mittelhessen, D-35398 Giessen, Germany
- European IPF Registry and Biobank, D-35392 Giessen, Germany
| |
Collapse
|
8
|
Nie YJ, Wu SH, Xuan YH, Yan G. Role of IL-17 family cytokines in the progression of IPF from inflammation to fibrosis. Mil Med Res 2022; 9:21. [PMID: 35550651 PMCID: PMC9102601 DOI: 10.1186/s40779-022-00382-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 04/12/2022] [Indexed: 01/01/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal chronic interstitial lung disease with no established treatment and is characterized by progressive scarring of the lung tissue and an irreversible decline in lung function. Chronic inflammation has been demonstrated to be the pathological basis of fibrosis. Emerging studies have revealed that most interleukin-17 (IL-17) isoforms are essential for the mediation of acute and chronic inflammation via innate and adaptive immunity. Overexpression or aberrant expression of IL-17 cytokines contributes to various pathological outcomes, including the initiation and exacerbation of IPF. Here, we aim to provide an overview of IL-17 family members in the pathogenesis of IPF.
Collapse
Affiliation(s)
- Yun-Juan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, Jiangsu, China
| | - Shuo-Hua Wu
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, 515000, Shandong, China
| | - Ying-Hua Xuan
- Department of Basic Medicine, Xiamen Medical College, Xiamen, 361000, Fujian, China
| | - Gen Yan
- Department of Radiology, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, 361000, Fujian, China.
| |
Collapse
|
9
|
Luzina IG, Rus V, Lockatell V, Courneya JP, Hampton BS, Fishelevich R, Misharin AV, Todd NW, Badea TC, Rus H, Atamas SP. Regulator of Cell Cycle Protein (RGCC/RGC-32) Protects against Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2022; 66:146-157. [PMID: 34668840 PMCID: PMC8845131 DOI: 10.1165/rcmb.2021-0022oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Some previous studies in tissue fibrosis have suggested a profibrotic contribution from elevated expression of a protein termed either RGCC (regulator of cell cycle) or RGC-32 (response gene to complement 32 protein). Our analysis of public gene expression datasets, by contrast, revealed a consistent decrease in RGCC mRNA levels in association with pulmonary fibrosis. Consistent with this observation, we found that stimulating primary adult human lung fibroblasts with transforming growth factor (TGF)-β in cell cultures elevated collagen expression and simultaneously attenuated RGCC mRNA and protein levels. Moreover, overexpression of RGCC in cultured lung fibroblasts attenuated the stimulating effect of TGF-β on collagen levels. Similar to humans with pulmonary fibrosis, the levels of RGCC were also decreased in vivo in lung tissues of wild-type mice challenged with bleomycin in both acute and chronic models. Mice with constitutive RGCC gene deletion accumulated more collagen in their lungs in response to chronic bleomycin challenge than did wild-type mice. RNA-Seq analyses of lung fibroblasts revealed that RGCC overexpression alone had a modest transcriptomic effect, but in combination with TGF-β stimulation, induced notable transcriptomic changes that negated the effects of TGF-β, including on extracellular matrix-related genes. At the level of intracellular signaling, RGCC overexpression delayed early TGF-β-induced Smad2/3 phosphorylation, elevated the expression of total and phosphorylated antifibrotic mediator STAT1, and attenuated the expression of a profibrotic mediator STAT3. We conclude that RGCC plays a protective role in pulmonary fibrosis and that its decline permits collagen accumulation. Restoration of RGCC expression may have therapeutic potential in pulmonary fibrosis.
Collapse
Affiliation(s)
- Irina G. Luzina
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Violeta Rus
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Virginia Lockatell
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Jean-Paul Courneya
- Health Sciences and Human Services Library, University of Maryland–Baltimore, Baltimore, Maryland
| | | | - Rita Fishelevich
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Alexander V. Misharin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, Illinois
| | - Nevins W. Todd
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Tudor C. Badea
- Retinal Circuits Development and Genetics Unit, National Eye Institute, Bethesda, Maryland; and,Faculty of Medicine, Research and Development Institute, Transilvania University of Brașov, Brașov, Romania
| | - Horea Rus
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Sergei P. Atamas
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| |
Collapse
|
10
|
Deleterious Role of Th9 Cells in Pulmonary Fibrosis. Cells 2021; 10:cells10113209. [PMID: 34831433 PMCID: PMC8621886 DOI: 10.3390/cells10113209] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 12/05/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease of unknown etiology. Immune disorders play an important role in IPF pathogenesis. Here, we show that Th9 cells differentiate and activate in the lung tissue of patients with IPF and bleomycin (BLM)-induced lung fibrosis mice. Moreover, we found that Th9 cells promote pulmonary fibrosis in two ways. On the one hand, Th9 cells promote fibroblast differentiation, activation, and collagen secretion by secreting IL-9. On the other hand, they promote differentiation of Th0 cells into Th2 cells by secreting IL-4. Th9 cells and Th2 cells can promote each other, accelerating the Th1/Th2 imbalance and eventually forming a positive feedback of pulmonary fibrosis. In addition, we found that neutralizing IL-9 in both preventive and therapeutic settings ameliorates bleomycin-induced pulmonary fibrosis. Furthermore, we identified several critical signaling pathways involved in the effect of neutralizing IL-9 on pulmonary fibrosis by proteomics study. From an immunological perspective, we elucidated the novel role and underlying mechanism of Th9 cells in pulmonary fibrosis. Our study suggested that Th9-based immunotherapy may be employed as a treatment strategy for IPF.
Collapse
|
11
|
Varricchio L, Iancu-Rubin C, Upadhyaya B, Zingariello M, Martelli F, Verachi P, Clementelli C, Denis JF, Rahman AH, Tremblay G, Mascarenhas J, Mesa RA, O'Connor-McCourt M, Migliaccio AR, Hoffman R. TGFβ1 protein trap AVID200 beneficially affects hematopoiesis and bone marrow fibrosis in myelofibrosis. JCI Insight 2021; 6:e145651. [PMID: 34383713 PMCID: PMC8492354 DOI: 10.1172/jci.insight.145651] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 08/11/2021] [Indexed: 11/17/2022] Open
Abstract
Myelofibrosis (MF) is a progressive chronic myeloproliferative neoplasm characterized by hyperactivation of JAK/STAT signaling and dysregulation of the transcription factor GATA1 in megakaryocytes (MKs). TGF-β plays a pivotal role in the pathobiology of MF by promoting BM fibrosis and collagen deposition and by enhancing the dormancy of normal hematopoietic stem cells (HSCs). In this study, we show that MF-MKs elaborated significantly greater levels of TGF-β1 than TGF-β2 and TGF-β3 to a varying degree, and we evaluated the ability of AVID200, a potent TGF-β1/TGF-β3 protein trap, to block the excessive TGF-β signaling. Treatment of human mesenchymal stromal cells with AVID200 significantly reduced their proliferation, decreased phosphorylation of SMAD2, and interfered with the ability of TGF-β1 to induce collagen expression. Moreover, treatment of MF mononuclear cells with AVID200 led to increased numbers of progenitor cells (PCs) with WT JAK2 rather than mutated JAK2V617F. This effect of AVID200 on MF PCs was attributed to its ability to block TGF-β1–induced p57Kip2 expression and SMAD2 activation, thereby allowing normal rather than MF PCs to preferentially proliferate and form hematopoietic colonies. To assess the in vivo effects of AVID200, Gata1lo mice, a murine model of MF, were treated with AVID200, resulting in the reduction in BM fibrosis and an increase in BM cellularity. AVID200 treatment also increased the frequency and numbers of murine progenitor cells as well as short-term and long-term HSCs. Collectively, these data provide the rationale for TGF-β1 blockade, with AVID200 as a therapeutic strategy for patients with MF.
Collapse
Affiliation(s)
- Lilian Varricchio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Camelia Iancu-Rubin
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Bhaskar Upadhyaya
- Human Immune Monitoring Core, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | | | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Verachi
- Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy
| | - Cara Clementelli
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | | | - Adeeb H Rahman
- Human Immune Monitoring Core, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | | | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Ruben A Mesa
- Hematology Oncology, Mays Cancer Center, San Antonio, United States of America
| | | | | | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| |
Collapse
|
12
|
Wang X, Ren R, Xu Z, Huang H, Jiang W, Ma J. Tirbanibulin Attenuates Pulmonary Fibrosis by Modulating Src/STAT3 Signaling. Front Pharmacol 2021; 12:693906. [PMID: 34349652 PMCID: PMC8326405 DOI: 10.3389/fphar.2021.693906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022] Open
Abstract
Tirbanibulin (KX-01) is the first clinical Src inhibitor of the novel peptidomimetic class that targets the peptide substrate site of Src providing more specificity toward the Src kinase. This study assessed the impact of KX-01 on cobalt chloride (CoCl2)-treated L929 cells and bleomycin (BLM)-induced pulmonary fibrosis in rats to evaluate the efficacy of this compound in vitro and in vivo, respectively. In CoCl2-treated L929 cells, KX-01 significantly reduced the expression of smooth muscle actin (α-SMA), collagen I, collagen III, hypoxia inducing factor (HIF-1α), signal transducers and transcriptional activators (p-STAT3), and p-Src. In BLM-induced pulmonary fibrosis rats, KX-01 reduced pathological scores, collagen deposition, α-SMA, collagen I, collagen III, p-Src, HIF-1α, and p-STAT3. Overall, these findings revealed that KX-01 can alleviate experimental pulmonary fibrosis via suppressing the p-SRC/p-STAT3 signaling pathways.
Collapse
Affiliation(s)
- Xin Wang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Rui Ren
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zehui Xu
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Haidi Huang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Wanglin Jiang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Jinbo Ma
- Medicine & Pharmacy Research Center, Binzhou Medical University, Yantai, China
| |
Collapse
|
13
|
Montero P, Milara J, Roger I, Cortijo J. Role of JAK/STAT in Interstitial Lung Diseases; Molecular and Cellular Mechanisms. Int J Mol Sci 2021; 22:6211. [PMID: 34207510 PMCID: PMC8226626 DOI: 10.3390/ijms22126211] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 02/07/2023] Open
Abstract
Interstitial lung diseases (ILDs) comprise different fibrotic lung disorders characterized by cellular proliferation, interstitial inflammation, and fibrosis. The JAK/STAT molecular pathway is activated under the interaction of a broad number of profibrotic/pro-inflammatory cytokines, such as IL-6, IL-11, and IL-13, among others, which are increased in different ILDs. Similarly, several growth factors over-expressed in ILDs, such as platelet-derived growth factor (PDGF), transforming growth factor β1 (TGF-β1), and fibroblast growth factor (FGF) activate JAK/STAT by canonical or non-canonical pathways, which indicates a predominant role of JAK/STAT in ILDs. Between the different JAK/STAT isoforms, it appears that JAK2/STAT3 are predominant, initiating cellular changes observed in ILDs. This review analyzes the expression and distribution of different JAK/STAT isoforms in ILDs lung tissue and different cell types related to ILDs, such as lung fibroblasts and alveolar epithelial type II cells and analyzes JAK/STAT activation. The effect of JAK/STAT phosphorylation on cellular fibrotic processes, such as proliferation, senescence, autophagy, endoplasmic reticulum stress, or epithelial/fibroblast to mesenchymal transition will be described. The small molecules directed to inhibit JAK/STAT activation were assayed in vitro and in in vivo models of pulmonary fibrosis, and different JAK inhibitors are currently approved for myeloproliferative disorders. Recent evidence indicates that JAK inhibitors or monoclonal antibodies directed to block IL-6 are used as compassionate use to attenuate the excessive inflammation and lung fibrosis related to SARS-CoV-2 virus. These altogether indicate that JAK/STAT pathway is an attractive target to be proven in future clinical trials of lung fibrotic disorders.
Collapse
Affiliation(s)
- Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (I.R.); (J.C.)
| | - Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (I.R.); (J.C.)
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029 Madrid, Spain
- Pharmacy Unit, University General Hospital Consortium of Valencia, 46014 Valencia, Spain
| | - Inés Roger
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (I.R.); (J.C.)
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029 Madrid, Spain
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (I.R.); (J.C.)
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029 Madrid, Spain
- Research and Teaching Unit, University General Hospital Consortium, 46014 Valencia, Spain
| |
Collapse
|
14
|
Kortekaas RK, Burgess JK, van Orsoy R, Lamb D, Webster M, Gosens R. Therapeutic Targeting of IL-11 for Chronic Lung Disease. Trends Pharmacol Sci 2021; 42:354-366. [PMID: 33612289 DOI: 10.1016/j.tips.2021.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/11/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
Interleukin (IL)-11 was originally recognized as an immunomodulatory and hematopoiesis-inducing cytokine. However, although IL-11 is typically not found in healthy individuals, it is now becoming evident that IL-11 may play a role in diverse pulmonary conditions, including IPF, asthma, and lung cancer. Additionally, experimental strategies targeting IL-11, such as humanized antibodies, have recently been developed, revealing the therapeutic potential of IL-11. Thus, further insight into the underlying mechanisms of IL-11 in lung disease may lead to the ability to interfere with pathological conditions that have a clear need for disease-modifying treatments, such as IPF. In this review, we outline the effects, expression, signaling, and crosstalk of IL-11 and focus on its role in lung disease and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Rosa K Kortekaas
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Janette K Burgess
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Medical Biology and Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Roël van Orsoy
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - David Lamb
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Megan Webster
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
15
|
Prêle CM, Iosifidis T, McAnulty RJ, Pearce DR, Badrian B, Miles T, Jamieson SE, Ernst M, Thompson PJ, Laurent GJ, Knight DA, Mutsaers SE. Reduced SOCS1 Expression in Lung Fibroblasts from Patients with IPF Is Not Mediated by Promoter Methylation or Mir155. Biomedicines 2021; 9:biomedicines9050498. [PMID: 33946612 PMCID: PMC8147237 DOI: 10.3390/biomedicines9050498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/12/2021] [Accepted: 04/23/2021] [Indexed: 01/16/2023] Open
Abstract
The interleukin (IL)-6 family of cytokines and exaggerated signal transducer and activator of transcription (STAT)3 signaling is implicated in idiopathic pulmonary fibrosis (IPF) pathogenesis, but the mechanisms regulating STAT3 expression and function are unknown. Suppressor of cytokine signaling (SOCS)1 and SOCS3 block STAT3, and low SOCS1 levels have been reported in IPF fibroblasts and shown to facilitate collagen production. Fibroblasts and lung tissue from IPF patients and controls were used to examine the mechanisms underlying SOCS1 down-regulation in IPF. A significant reduction in basal SOCS1 mRNA in IPF fibroblasts was confirmed. However, there was no difference in the kinetics of activation, and methylation of SOCS1 in control and IPF lung fibroblasts was low and unaffected by 5′-aza-2′-deoxycytidine’ treatment. SOCS1 is a target of microRNA-155 and although microRNA-155 levels were increased in IPF tissue, they were reduced in IPF fibroblasts. Therefore, SOCS1 is not regulated by SOCS1 gene methylation or microRNA155 in these cells. In conclusion, we confirmed that IPF fibroblasts had lower levels of SOCS1 mRNA compared with control fibroblasts, but we were unable to determine the mechanism. Furthermore, although SOCS1 may be important in the fibrotic process, we were unable to find a significant role for SOCS1 in regulating fibroblast function.
Collapse
Affiliation(s)
- Cecilia M. Prêle
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
- Centre for Respiratory Health and Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, University of Western Australia, Nedland, WA 6009, Australia
| | - Thomas Iosifidis
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
| | - Robin J. McAnulty
- Centre for Inflammation and Tissue Repair, Rayne Institute, Department of Medicine, University College London, London WC1E 6JJ, UK; (R.J.M.); (D.R.P.)
| | - David R. Pearce
- Centre for Inflammation and Tissue Repair, Rayne Institute, Department of Medicine, University College London, London WC1E 6JJ, UK; (R.J.M.); (D.R.P.)
| | - Bahareh Badrian
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
| | - Tylah Miles
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
| | - Sarra E. Jamieson
- Telethon Kids Institute and Centre for Child Health Research, University of Western Australia, Nedlands, WA 6009, Australia;
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
| | - Philip J. Thompson
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
| | - Geoffrey J. Laurent
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
- Centre for Respiratory Health and Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, University of Western Australia, Nedland, WA 6009, Australia
| | - Darryl A. Knight
- Faculty of Medicine, University of British Columbia (UBC), Vancouver, BC V6Z 1Y5, Canada;
| | - Steven E. Mutsaers
- Institute for Respiratory Health, Nedland, WA 6009, Australia; (C.M.P.); (T.I.); (B.B.); (T.M.); (P.J.T.); (G.J.L.)
- Centre for Respiratory Health and Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, University of Western Australia, Nedland, WA 6009, Australia
- Correspondence: ; Tel.: +61-(0)8-6151-0891; Fax: +61-(0)8-6151-1027
| |
Collapse
|
16
|
Faletti L, Ehl S, Heeg M. Germline STAT3 gain-of-function mutations in primary immunodeficiency: Impact on the cellular and clinical phenotype. Biomed J 2021; 44:412-421. [PMID: 34366294 PMCID: PMC8514798 DOI: 10.1016/j.bj.2021.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 12/25/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a key transcription factor involved in regulation of immune cell activation and differentiation. Recent discoveries highlight the role of germline activating STAT3 mutations in inborn errors of immunity characterized by early-onset multi-organ autoimmunity and lymphoproliferation. Much progress has been made in defining the clinical spectrum of STAT3 GOF disease and unraveling the molecular and cellular mechanisms underlying this disease. In this review, we summarize our current understanding of the disease and discuss the clinical phenotype, diagnostic approach, cellular and molecular effects of STAT3 GOF mutations and therapeutic concepts for these patients.
Collapse
Affiliation(s)
- Laura Faletti
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Heeg
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
17
|
miRNAs Potentially Involved in Post Lung Transplant-Obliterative Bronchiolitis: The Role of miR-21-5p. Cells 2021; 10:cells10030688. [PMID: 33804639 PMCID: PMC8003603 DOI: 10.3390/cells10030688] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 11/16/2022] Open
Abstract
Epigenetic changes, including miRNAs deregulation, have been suggested to play a significant role in development of obliterative bronchiolitis (OB) in transplanted lungs. Many studies have tried to identify ideal candidate miRNAs and the downstream pathways implicated in the bronchiolar fibro-obliterative process. Several candidate miRNAs, previously indicated as possibly being associated with OB, were analyzed by combining the quantitative real time-polymerase chain reaction (qRT-PCR) and in situ hybridization (ISH) of lung tissues of OB affected patients. Disease and OB-lesion-specific expression of miR-21-5p was confirmed and by computational analysis we were able to identify the network of genes most probably associated miR-21-5p in the context of OB fibrogenesis. Among all potentially associated genes, STAT3 had a very high probability score. Immunohistochemistry showed that STAT3/miR-21-5p were co-over expressed in OB lesions, thus, suggesting miR-21-5p could regulate STAT3 expression. However, miR-21-5p inhibition in cultures of bronchiolitis obliterans syndrome (BOS) derived myofibroblasts did not significantly affect STAT3 mRNA and protein expression levels. This study demonstrates the specificity of miR-21-5p over-expression in OB lesions and contributes to existing knowledge on the miR-21-5p downstream pathway. Activation of STAT3 is associated with miR-21-5p upregulation, however, STAT-3 network activation is most likely complex and miR-21-5p is not the sole regulator of STAT3.
Collapse
|
18
|
Crosstalk between alveolar macrophages and alveolar epithelial cells/fibroblasts contributes to the pulmonary toxicity of gefitinib. Toxicol Lett 2020; 338:1-9. [PMID: 33248157 DOI: 10.1016/j.toxlet.2020.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 01/02/2023]
Abstract
Gefitinib is an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor indicated for the first-line treatment of patients with metastatic or advanced non-small cell lung cancer (NSCLC) whose tumors have specific EGFR mutations. Pulmonary toxicity is one of the fatal adverse effects of gefitinib and the underlying mechanism remains unclear. Here we demonstrated that alveolar macrophages contributed to gefitinib-induced pulmonary toxicity through promoting alveolar epithelial cells to undergo epithelial to mesenchymal transition (EMT) and inducing activation and antiapoptotic effect in fibroblasts. Further, we found that alveolar macrophage-secreted MCP-1 worked as a key factor in the pathologic changes of these two cell types. Gefitinib increased Mcp-1 transcription level via the nuclear import of the transcription factor STAT3. In conclusion, our data uncovered the underlying mechanisms of macrophage-promoted pulmonary toxicity in the presence of gefitinib. MCP-1 antibody or inhibition of STAT3 activation may represent novel therapeutic strategies for preventing gefitinib-induced pulmonary toxicity.
Collapse
|
19
|
Cheng J, Eroglu A. The Promising Effects of Astaxanthin on Lung Diseases. Adv Nutr 2020; 12:850-864. [PMID: 33179051 PMCID: PMC8166543 DOI: 10.1093/advances/nmaa143] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/25/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022] Open
Abstract
Astaxanthin (ASX) is a naturally occurring xanthophyll carotenoid. Both in vitro and in vivo studies have shown that it is a potent antioxidant with anti-inflammatory properties. Lung cancer is the leading cause of cancer death worldwide, whereas other lung diseases such as chronic obstructive pulmonary disease, emphysema, and asthma are of high prevalence. In the past decade, mounting evidence has suggested a protective role for ASX against lung diseases. This article reviews the potential role of ASX in protecting against lung diseases, including lung cancer. It also summarizes the underlying molecular mechanisms by which ASX protects against pulmonary diseases, including regulating the nuclear factor erythroid 2-related factor/heme oxygenase-1 pathway, NF-κB signaling, mitogen-activated protein kinase signaling, Janus kinase-signal transducers and activators of transcription-3 signaling, the phosphoinositide 3-kinase/Akt pathway, and modulating immune response. Several future directions are proposed in this review. However, most in vitro and in vivo studies have used ASX at concentrations that are not achievable by humans. Also, no clinical trials have been conducted and/or reported. Thus, preclinical studies with ASX treatment within physiological concentrations as well as human studies are required to examine the health benefits of ASX with respect to lung diseases.
Collapse
Affiliation(s)
- Junrui Cheng
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, USA
| | | |
Collapse
|
20
|
Kasam RK, Ghandikota S, Soundararajan D, Reddy GB, Huang SK, Jegga AG, Madala SK. Inhibition of Aurora Kinase B attenuates fibroblast activation and pulmonary fibrosis. EMBO Mol Med 2020; 12:e12131. [PMID: 32761869 PMCID: PMC7507328 DOI: 10.15252/emmm.202012131] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Fibroblast activation including proliferation, survival, and ECM production is central to initiation and maintenance of fibrotic lesions in idiopathic pulmonary fibrosis (IPF). However, druggable molecules that target fibroblast activation remain limited. In this study, we show that multiple pro‐fibrotic growth factors, including TGFα, CTGF, and IGF1, increase aurora kinase B (AURKB) expression and activity in fibroblasts. Mechanistically, we demonstrate that Wilms tumor 1 (WT1) is a key transcription factor that mediates TGFα‐driven AURKB upregulation in fibroblasts. Importantly, we found that inhibition of AURKB expression or activity is sufficient to attenuate fibroblast activation. We show that fibrosis induced by TGFα is highly dependent on AURKB expression and treating TGFα mice with barasertib, an AURKB inhibitor, reverses fibroblast activation, and pulmonary fibrosis. Barasertib similarly attenuated fibrosis in the bleomycin model of pulmonary fibrosis. Together, our preclinical studies provide important proof‐of‐concept that demonstrate barasertib as a possible intervention therapy for IPF.
Collapse
Affiliation(s)
- Rajesh K Kasam
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Biochemistry, National Institute of Nutrition, Hyderabad, India
| | - Sudhir Ghandikota
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Computer Science, University of Cincinnati College of Engineering, Cincinnati, OH, USA
| | | | - Geereddy B Reddy
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, India
| | - Steven K Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Computer Science, University of Cincinnati College of Engineering, Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Satish K Madala
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
21
|
Deng Z, Fear MW, Suk Choi Y, Wood FM, Allahham A, Mutsaers SE, Prêle CM. The extracellular matrix and mechanotransduction in pulmonary fibrosis. Int J Biochem Cell Biol 2020; 126:105802. [PMID: 32668329 DOI: 10.1016/j.biocel.2020.105802] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Abstract
Pulmonary fibrosis is characterised by excessive scarring in the lung which leads to compromised lung function, serious breathing problems and in some diseases, death. It includes several lung disorders with idiopathic pulmonary fibrosis (IPF) the most common and most severe. Pulmonary fibrosis is considered to be perpetuated by aberrant wound healing which leads to fibroblast accumulation, differentiation and activation, and deposition of excessive amounts of extracellular matrix (ECM) components, in particular, collagen. Recent studies have identified the importance of changes in the composition and structure of lung ECM during the development of pulmonary fibrosis and the interaction between ECM and lung cells. There is strong evidence that increased matrix stiffness induces changes in cell function including proliferation, migration, differentiation and activation. Understanding how changes in the ECM microenvironment influence cell behaviour during fibrogenesis, and the mechanisms regulating these changes, will provide insight for developing new treatments.
Collapse
Affiliation(s)
- Zhenjun Deng
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia; Institute for Respiratory Health, Nedlands, WA, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia; Burns Service of Western Australia, Perth Children's Hospital, Nedlands, WA, Australia; Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Amira Allahham
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia
| | - Steven E Mutsaers
- Institute for Respiratory Health, Nedlands, WA, Australia; Centre for Respiratory Health, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Cecilia M Prêle
- Institute for Respiratory Health, Nedlands, WA, Australia; Centre for Respiratory Health, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.
| |
Collapse
|
22
|
Galati D, Zanotta S, Polistina GE, Coppola A, Capitelli L, Bocchino M. Circulating dendritic cells are severely decreased in idiopathic pulmonary fibrosis with a potential value for prognosis prediction. Clin Immunol 2020; 215:108454. [DOI: 10.1016/j.clim.2020.108454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/15/2020] [Accepted: 04/29/2020] [Indexed: 01/13/2023]
|
23
|
Clynick B, Jo HE, Corte TJ, Glaspole IN, Grainge C, Hopkins PMA, Reynolds PN, Chapman S, Walters EH, Zappala C, Keir GJ, Cooper WA, Mahar AM, Ellis S, Goh NS, Baltic S, Ryan M, Tan DBA, Moodley YP. Circulating RNA differences between patients with stable and progressive idiopathic pulmonary fibrosis. Eur Respir J 2020; 56:13993003.02058-2019. [PMID: 32381494 DOI: 10.1183/13993003.02058-2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/08/2020] [Indexed: 11/05/2022]
Affiliation(s)
- Britt Clynick
- Institute for Respiratory Health, Nedlands, Australia.,University of Western Australia, Crawley, Australia
| | - Helen E Jo
- University of Sydney, Camperdown, Australia.,Royal Prince Alfred Hospital, Camperdown, Australia
| | - Tamera J Corte
- University of Sydney, Camperdown, Australia.,Royal Prince Alfred Hospital, Camperdown, Australia
| | - Ian N Glaspole
- Monash University, Clayton, Australia.,Alfred Hospital, Melbourne, Australia
| | - Christopher Grainge
- University of Newcastle, Callaghan, Australia.,John Hunter Hospital, New Lambton Heights, Australia
| | - Peter M A Hopkins
- University of Queensland, St Lucia, Australia.,Prince Charles Hospital, Chermside, Australia
| | - Paul N Reynolds
- University of Adelaide, Adelaide, Australia.,Royal Adelaide Hospital, Adelaide, Australia
| | | | - E Haydn Walters
- Alfred Hospital, Melbourne, Australia.,University of Tasmania, Hobart, Australia.,University of Melbourne, Parkville, Australia.,Royal Hobart Hospital, Hobart, Australia
| | | | | | - Wendy A Cooper
- University of Sydney, Camperdown, Australia.,Royal Prince Alfred Hospital, Camperdown, Australia.,Western Sydney University, Sydney, Australia
| | - Annabelle M Mahar
- University of Sydney, Camperdown, Australia.,Royal Prince Alfred Hospital, Camperdown, Australia
| | | | - Nicole S Goh
- Austin Hospital, Heidelberg, Australia.,Institute of Breathing and Sleep, Heidelberg, Australia
| | - Svetlana Baltic
- Institute for Respiratory Health, Nedlands, Australia.,University of Western Australia, Crawley, Australia
| | - Marisa Ryan
- Institute for Respiratory Health, Nedlands, Australia.,University of Western Australia, Crawley, Australia
| | - Dino B A Tan
- Institute for Respiratory Health, Nedlands, Australia.,University of Western Australia, Crawley, Australia
| | - Yuben P Moodley
- Institute for Respiratory Health, Nedlands, Australia.,University of Western Australia, Crawley, Australia.,Fiona Stanley Hospital, Murdoch, Australia
| |
Collapse
|
24
|
Carvalho JL, Miranda M, Fialho AK, Castro-Faria-Neto H, Anatriello E, Keller AC, Aimbire F. Oral feeding with probiotic Lactobacillus rhamnosus attenuates cigarette smoke-induced COPD in C57Bl/6 mice: Relevance to inflammatory markers in human bronchial epithelial cells. PLoS One 2020; 15:e0225560. [PMID: 32330145 PMCID: PMC7182195 DOI: 10.1371/journal.pone.0225560] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/03/2020] [Indexed: 01/10/2023] Open
Abstract
COPD is a prevalent lung disease with significant impacts on public health. Affected airways exhibit pulmonary neutrophilia and consequent secretion of pro-inflammatory cytokines and proteases, which result in lung emphysema. Probiotics act as nonspecific modulators of the innate immune system that improve several inflammatory responses. To investigate the effect of Lactobacillus rhamnosus (Lr) on cigarette smoke (CS)-induced COPD C57Bl/6 mice were treated with Lr during the week before COPD induction and three times/week until euthanasia. For in vitro assays, murine bronchial epithelial cells as well as human bronchial epithelial cells exposed to cigarette smoke extract during 24 hours were treated with Lr 1 hour before CSE addition. Lr treatment attenuated the inflammatory response both in the airways and lung parenchyma, reducing inflammatory cells infiltration and the production of pro-inflammatory cytokines and chemokines. Also, Lr-treated mice presented with lower metalloproteases in lung tissue and lung remodeling. In parallel to the reduction in the expression of TLR2, TLR4, TLR9, STAT3, and NF-κB in lung tissue, Lr increased the levels of IL-10 as well as SOCS3 and TIMP1/2, indicating the induction of an anti-inflammatory environment. Similarly, murine bronchial epithelial cells as well as human bronchial epithelial cells (BEAS) exposed to CSE produced pro-inflammatory cytokines and chemokines, which were inhibited by Lr treatment in association with the production of anti-inflammatory molecules. Moreover, the presence of Lr also modulated the expression of COPD-associated transcription found into BALF of COPD mice group, i.e., Lr downregulated expression of NF-κB and STAT3, and inversely upregulated increased expression of SOCS3. Thus, our findings indicate that Lr modulates the balance between pro- and anti-inflammatory cytokines in human bronchial epithelial cells upon CS exposure and it can be a useful tool to improve the lung inflammatory response associated with COPD.
Collapse
Affiliation(s)
- J. L. Carvalho
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, São Paulo, Brazil
| | - M. Miranda
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, São Paulo, Brazil
| | - A. K. Fialho
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, São Paulo, Brazil
| | | | - E. Anatriello
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, São Paulo, Brazil
| | - A. C. Keller
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - F. Aimbire
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, São Paulo, Brazil
| |
Collapse
|
25
|
Shen X, Zhao L, Chen P, Gong Y, Liu D, Zhang X, Dai L, Sun Q, Lou J, Jin Z, Zhang B, Niu D, Chen C, Qi X, Jia D. A thiazole-derived oridonin analogue exhibits antitumor activity by directly and allosterically inhibiting STAT3. J Biol Chem 2019; 294:17471-17486. [PMID: 31594861 DOI: 10.1074/jbc.ra119.009801] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/25/2019] [Indexed: 12/25/2022] Open
Abstract
Constitutive activation of signal transducer and activator of transcription 3 (STAT3) occurs in ∼70% of human cancers, and STAT3 is regarded as one of the most promising targets for cancer therapy. However, specific direct STAT3 inhibitors remain to be developed. Oridonin is an ent-kaurane plant-derived diterpenoid with anti-cancer and anti-inflammatory activities. Here, using an array of cell-based and biochemical approaches, including cell proliferation and apoptosis assays, pulldown and reporter gene assays, site-directed mutagenesis, and molecular dynamics analyses, we report that a thiazole-derived oridonin analogue, CYD0618, potently and directly inhibits STAT3. We found that CYD0618 covalently binds to Cys-542 in STAT3 and suppresses its activity through an allosteric effect, effectively reducing STAT3 dimerization and nuclear translocation, as well as decreasing expression of STAT3-targeted oncogenes. Remarkably, CYD0618 not only strongly inhibited growth of multiple cancer cell lines that harbor constitutive STAT3 activation, but it also suppressed in vivo tumor growth via STAT3 inhibition. Taken together, our findings suggest Cys-542 as a druggable site for selectively inhibiting STAT3 and indicate that CYD0618 represents a promising lead compound for developing therapeutic agents against STAT3-driven diseases.
Collapse
Affiliation(s)
- Xiaofei Shen
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Lin Zhao
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Peihao Chen
- School of Life Science, Peking University, Beijing 100084, China.,National Institute of Biological Sciences (NIBS), Beijing 102206, China
| | - Yanqiu Gong
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Dingdong Liu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Xia Zhang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Lunzhi Dai
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Qingxiang Sun
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Jizhong Lou
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhong Jin
- Computer Network Information Center and Center of Scientific Computing Applications and Research, Chinese Academy of Sciences, Beijing 100190, China
| | - Baohua Zhang
- Computer Network Information Center and Center of Scientific Computing Applications and Research, Chinese Academy of Sciences, Beijing 100190, China
| | - Dawen Niu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming 650223, China
| | - Xiangbing Qi
- National Institute of Biological Sciences (NIBS), Beijing 102206, China
| | - Da Jia
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| |
Collapse
|
26
|
CXCL16 Induces the Progression of Pulmonary Fibrosis through Promoting the Phosphorylation of STAT3. Can Respir J 2019; 2019:2697376. [PMID: 31379980 PMCID: PMC6652085 DOI: 10.1155/2019/2697376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/18/2019] [Accepted: 03/26/2019] [Indexed: 01/21/2023] Open
Abstract
Aim The transmembrane chemokine (C-X-C motif) ligand 16 (CXCL16) plays a vital role in the pathogenesis of organ fibrosis, including the liver and kidney. However, the detailed biological function of CXCL16 is still not fully understood in the progression of pulmonary fibrosis (PF). The aim of present study is to examine the function of CXCL16 in PF. Materials and Methods In this study, we constructed the PF model on mouse by using bleomycin and analyzed the effect of the mouse recombinant protein CXCL16 on mouse lung fibroblast L929 (LF) as well. To further examine the connection between CXCL16 and STAT3 in mouse LF cells, the STAT3 inhibitor AG490 was utilized to inhibit the expression of STAT3. Meanwhile, lipopolysaccharide was used to enhance the phosphorylation of STAT3 (p-STAT3) in mouse LF cells. Results Our results indicated that the level of CXCL16/CXCR6 was significantly upregulated in the mouse PF model. Moreover, the level of p-STAT3 was also promoted. In addition, the mouse recombinant protein CXCL16 not only contributed to the proliferation of mouse LF cells but also induced the expression of p-STAT3 in LF cells. However, the effect of CXCL16 was deeply abolished by the STAT3 inhibitor AG490 in LF cells. Meanwhile, the antibody of CXCL16 deeply reduced the phosphorylation of STAT3 in lipopolysaccharide (LPS) cultured cells. Conclusions All these results demonstrated that CXCL16 promoted the phosphorylation of STAT3 and further demonstrated that STAT3 was a critical component in CXCL16/CXCR6 signaling pathway. This research not only enhanced the comprehension of CXCL16 but also indicated its potential value as a target in the treatment for human PF.
Collapse
|
27
|
Xu Q, Liu Y, Pan H, Xu T, Li Y, Yuan J, Li P, Yao W, Yan W, Ni C. Aberrant expression of miR-125a-3p promotes fibroblast activation via Fyn/STAT3 pathway during silica-induced pulmonary fibrosis. Toxicology 2019; 414:57-67. [PMID: 30658076 DOI: 10.1016/j.tox.2019.01.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 12/11/2022]
Abstract
Various miRNAs are dysregulated during initiation and progression of pulmonary fibrosis. However, their function remains limited in silicosis. Here, we observed that miR-125a-3p was downregulated in silica-induced fibrotic murine lung tissues. Ectopic miR-125a-3p expression with chemotherapy attenuated silica-induced pulmonary fibrosis. Further in vitro experiments revealed that TGF-β1 effectively decreased miR-125a-3p expression in fibroblast lines (NIH/3T3 and MRC-5). Overexpression of miR-125a-3p blocked fibroblast activation stimulated by TGF-β1. Mechanistically, miR-125a-3p could bind to the 3'-untranslated region of Fyn and inhibit its expression in both mRNA and protein levels, thus causing inactivation of Fyn downstream effector STAT3. Fyn and p-STAT3, as opposed to miR-125a-3p expression, were elevated in silica-induced fibrotic murine lung tissues and TGF-β1-treated fibroblast lines. Furthermore, Fyn knockdown or p-STAT3 suppression effectively attenuated fibroblast activation and ECM production. Taken together, miR-125a-3p is involved in fibrosis pathogenesis by fibroblast activation, suggesting that targeting miR-125a-3p/Fyn/STAT3 signaling pathway could be a potential therapeutic approach for pulmonary fibrosis.
Collapse
Affiliation(s)
- Qi Xu
- Department of Occupational Medical and Environmental Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Yi Liu
- Department of Occupational Medical and Environmental Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Honghong Pan
- Department of Occupational Medical and Environmental Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Tiantian Xu
- Department of Occupational Medical and Environmental Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Yan Li
- Department of Occupational Medical and Environmental Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Jiali Yuan
- Department of Occupational Medical and Environmental Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Ping Li
- Department of Occupational Medical and Environmental Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Wenxi Yao
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| | - Weiwen Yan
- Department of Occupational Medical and Environmental Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Chunhui Ni
- Department of Occupational Medical and Environmental Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
28
|
Pterostilbene, a bioactive component of blueberries, alleviates renal fibrosis in a severe mouse model of hyperuricemic nephropathy. Biomed Pharmacother 2019; 109:1802-1808. [DOI: 10.1016/j.biopha.2018.11.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/23/2018] [Accepted: 11/06/2018] [Indexed: 11/20/2022] Open
|
29
|
Song DG, Kim D, Jung JW, Nam SH, Kim JE, Kim HJ, Kim JH, Pan CH, Kim S, Lee JW. Glutamyl-Prolyl-tRNA Synthetase Regulates Epithelial Expression of Mesenchymal Markers and Extracellular Matrix Proteins: Implications for Idiopathic Pulmonary Fibrosis. Front Pharmacol 2018; 9:1337. [PMID: 30524284 PMCID: PMC6256097 DOI: 10.3389/fphar.2018.01337] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/30/2018] [Indexed: 01/01/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), a chronic disease of unknown cause, is characterized by abnormal accumulation of extracellular matrix (ECM) in fibrotic foci in the lung. Previous studies have shown that the transforming growth factor β1 (TGFβ1) and signal transducers and activators of transcription (STAT) pathways play roles in IPF pathogenesis. Glutamyl-prolyl-tRNA-synthetase (EPRS) has been identified as a target for anti-fibrosis therapy, but the link between EPRS and TGFβ1-mediated IPF pathogenesis remains unknown. Here, we studied the role of EPRS in the development of fibrotic phenotypes in A549 alveolar epithelial cells and bleomycin-treated animal models. We found that EPRS knockdown inhibited the TGFβ1-mediated upregulation of fibronectin and collagen I and the mesenchymal proteins α-smooth muscle actin (α-SMA) and snail 1. TGFβ1-mediated transcription of collagen I-α1 and laminin γ2 in A549 cells was also down-regulated by EPRS suppression, indicating that EPRS is required for ECM protein transcriptions. Activation of STAT signaling in TGFβ1-induced ECM expression was dependent on EPRS. TGFβ1 treatment resulted in EPRS-dependent in vitro formation of a multi-protein complex consisting of the TGFβ1 receptor, EPRS, Janus tyrosine kinases (JAKs), and STATs. In vivo lung tissue from bleomycin-treated mice showed EPRS-dependent STAT6 phosphorylation and ECM production. Our results suggest that epithelial EPRS regulates the expression of mesenchymal markers and ECM proteins via the TGFβ1/STAT signaling pathway. Therefore, epithelial EPRS can be used as a potential target to develop anti-IPF treatments.
Collapse
Affiliation(s)
- Dae-Geun Song
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea.,Systems Biotechnology Research Center, Korea Institute of Science and Technology (KIST), Gangneung-si, South Korea
| | - Doyeun Kim
- Medicinal Bioconvergence Research Center, Seoul National University, Seoul, South Korea
| | - Jae Woo Jung
- Interdisciplinary Program in Genetic Engineering, Seoul National University, Seoul, South Korea
| | - Seo Hee Nam
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Ji Eon Kim
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Hye-Jin Kim
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jong Hyun Kim
- Medicinal Bioconvergence Research Center, Seoul National University, Seoul, South Korea
| | - Cheol-Ho Pan
- Systems Biotechnology Research Center, Korea Institute of Science and Technology (KIST), Gangneung-si, South Korea
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, Seoul National University, Seoul, South Korea
| | - Jung Weon Lee
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea.,Medicinal Bioconvergence Research Center, Seoul National University, Seoul, South Korea
| |
Collapse
|
30
|
Oh RS, Haak AJ, Smith KMJ, Ligresti G, Choi KM, Xie T, Wang S, Walters PR, Thompson MA, Freeman MR, Manlove LJ, Chu VM, Feghali-Bostwick C, Roden AC, Schymeinsky J, Pabelick CM, Prakash YS, Vassallo R, Tschumperlin DJ. RNAi screening identifies a mechanosensitive ROCK-JAK2-STAT3 network central to myofibroblast activation. J Cell Sci 2018; 131:jcs.209932. [PMID: 29678906 DOI: 10.1242/jcs.209932] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
Myofibroblasts play key roles in wound healing and pathological fibrosis. Here, we used an RNAi screen to characterize myofibroblast regulatory genes, using a high-content imaging approach to quantify α-smooth muscle actin stress fibers in cultured human fibroblasts. Screen hits were validated on physiological compliance hydrogels, and selected hits tested in primary fibroblasts from patients with idiopathic pulmonary fibrosis. Our RNAi screen led to the identification of STAT3 as an essential mediator of myofibroblast activation and function. Strikingly, we found that STAT3 phosphorylation, while responsive to exogenous ligands on both soft and stiff matrices, is innately active on a stiff matrix in a ligand/receptor-independent, but ROCK- and JAK2-dependent fashion. These results demonstrate how a cytokine-inducible signal can become persistently activated by pathological matrix stiffening. Consistent with a pivotal role for this pathway in driving persistent fibrosis, a STAT3 inhibitor attenuated murine pulmonary fibrosis when administered in a therapeutic fashion after bleomycin injury. Our results identify novel genes essential for the myofibroblast phenotype, and point to STAT3 as an important target in pulmonary fibrosis and other fibrotic diseases.
Collapse
Affiliation(s)
- Raymond S Oh
- Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115, USA
| | - Andrew J Haak
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Karry M J Smith
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Giovanni Ligresti
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Kyoung Moo Choi
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tiao Xie
- Image and Data Analysis Core, Harvard Medical School, Boston, MA 02115, USA
| | - Shaohua Wang
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Paula R Walters
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael A Thompson
- Departments of Anesthesiology and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Michelle R Freeman
- Departments of Anesthesiology and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Logan J Manlove
- Departments of Anesthesiology and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Vivian M Chu
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Carol Feghali-Bostwick
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jürgen Schymeinsky
- Department of Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - Christina M Pabelick
- Departments of Anesthesiology and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Y S Prakash
- Departments of Anesthesiology and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert Vassallo
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel J Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
31
|
Abstract
Activation of TGF-β1 initiates a program of temporary collagen accumulation important to wound repair in many organs. However, the outcome of temporary extracellular matrix strengthening all too frequently morphs into progressive fibrosis, contributing to morbidity and mortality worldwide. To avoid this maladaptive outcome, TGF-β1 signaling is regulated at numerous levels and intimately connected to feedback signals that limit accumulation. Here, we examine the current understanding of the core functions of TGF-β1 in promoting collagen accumulation, parallel pathways that promote physiological repair, and pathological triggers that tip the balance toward progressive fibrosis. Implicit in better understanding of these processes is the identification of therapeutic opportunities that will need to be further advanced to limit or reverse organ fibrosis.
Collapse
Affiliation(s)
- Kevin K Kim
- Department of Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan 48109
| | - Dean Sheppard
- Department of Medicine, Cardiovascular Research Institute, and Lung Biology Center, University of California, San Francisco, San Francisco, California 94143
| | - Harold A Chapman
- Department of Medicine, Cardiovascular Research Institute, and Lung Biology Center, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
32
|
Milara J, Ballester B, Morell A, Ortiz JL, Escrivá J, Fernández E, Perez-Vizcaino F, Cogolludo A, Pastor E, Artigues E, Morcillo E, Cortijo J. JAK2 mediates lung fibrosis, pulmonary vascular remodelling and hypertension in idiopathic pulmonary fibrosis: an experimental study. Thorax 2018; 73:519-529. [PMID: 29440315 DOI: 10.1136/thoraxjnl-2017-210728] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 01/11/2018] [Accepted: 01/22/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a common disorder in patients with idiopathic pulmonary fibrosis (IPF) and portends a poor prognosis. Recent studies using vasodilators approved for PH have failed in improving IPF mainly due to ventilation (V)/perfusion (Q) mismatching and oxygen desaturation. Janus kinase type 2 (JAK2) is a non-receptor tyrosine kinase activated by a broad spectrum of profibrotic and vasoactive mediators, but its role in PH associated to PH is unknown. OBJECTIVE The study of JAK2 as potential target to treat PH in IPF. METHODS AND RESULTS JAK2 expression was increased in pulmonary arteries (PAs) from IPF (n=10; 1.93-fold; P=0.0011) and IPF+PH (n=9; 2.65-fold; P<0.0001) compared with PA from control subjects (n=10). PA remodelling was evaluated in human pulmonary artery endothelial cells (HPAECs) and human pulmonary artery smooth muscle cells (HPASMCs) from patients with IPF in vitro treated with the JAK2 inhibitor JSI-124 or siRNA-JAK2 and stimulated with transforming growth factor beta. Both JSI-124 and siRNA-JAK2 inhibited the HPAEC to mesenchymal transition and the HPASMCs to myofibroblast transition and proliferation. JAK2 inhibition induced small PA relaxation in precision-cut lung slice experiments. PA relaxation was dependent of the large conductance calcium-activated potassium channel (BKCa). JAK2 inhibition activated BKCa channels and reduced intracellular Ca2+. JSI-124 1 mg/kg/day, reduced bleomycin-induced lung fibrosis, PA remodelling, right ventricular hypertrophy, PA hypertension and V/Q mismatching in rats. The animal studies followed the ARRIVE guidelines. CONCLUSIONS JAK2 participates in PA remodelling and tension and may be an attractive target to treat IPF associated to PH.
Collapse
Affiliation(s)
- Javier Milara
- Department of Pharmacology, Faculty of Medicine, Jaume I University, Castellón de la Plana, Spain.,Pharmacy Unit, University General Hospital Consortium, Valencia, Spain.,CIBERES, Health Institute Carlos III, Valencia, Spain
| | - Beatriz Ballester
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Anselm Morell
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - José L Ortiz
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Juan Escrivá
- Thoracic Surgery Unit, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Estrella Fernández
- Respiratory Unit, University General Hospital Consortium, Valencia, Spain
| | - Francisco Perez-Vizcaino
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Angel Cogolludo
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Enrique Pastor
- Department of Thoracic Surgery, University General Hospital Consortium, Valencia, Spain
| | - Enrique Artigues
- Surgery Unit, University General Hospital Consortium, Valencia, Spain
| | - Esteban Morcillo
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain.,Health Research Institute INCLIVA, Valencia, Spain
| | - Julio Cortijo
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Research and teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
33
|
Milara J, Hernandez G, Ballester B, Morell A, Roger I, Montero P, Escrivá J, Lloris JM, Molina-Molina M, Morcillo E, Cortijo J. The JAK2 pathway is activated in idiopathic pulmonary fibrosis. Respir Res 2018; 19:24. [PMID: 29409529 PMCID: PMC5801676 DOI: 10.1186/s12931-018-0728-9] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 01/25/2018] [Indexed: 02/01/2023] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is the most rapidly progressive and fatal fibrotic disorder, with no curative therapies. The signal transducer and activator of transcription 3 (STAT3) protein is activated in lung fibroblasts and alveolar type II cells (ATII), thereby contributing to lung fibrosis in IPF. Although activation of Janus kinase 2 (JAK2) has been implicated in proliferative disorders, its role in IPF is unknown. The aim of this study was to analyze JAK2 activation in IPF, and to determine whether JAK2/STAT3 inhibition is a potential therapeutic strategy for this disease. Methods and results JAK2/p-JAK2 and STAT3/pSTAT3 expression was evaluated using quantitative real time-PCR, western blotting, and immunohistochemistry. Compared to human healthy lung tissue (n = 10) both proteins were upregulated in the lung tissue of IPF patients (n = 12). Stimulating primary ATII and lung fibroblasts with transforming growth factor beta 1 or interleukin (IL)-6/IL-13 activated JAK2 and STAT3, inducing epithelial to mesenchymal and fibroblast to myofibroblast transitions. Dual p-JAK2/p-STAT3 inhibition with JSI-124 or silencing of JAK2 and STAT3 genes suppressed ATII and the fibroblast to myofibroblast transition, with greater effects than the sum of those obtained using JAK2 or STAT3 inhibitors individually. Dual rather than single inhibition was also more effective for inhibiting fibroblast migration, preventing increases in fibroblast senescence and Bcl-2 expression, and ameliorating impaired autophagy. In rats administered JSI-124, a dual inhibitor of p-JAK2/p-STAT3, at a dose of 1 mg/kg/day, bleomycin-induced lung fibrosis was reduced and collagen deposition in the lung was inhibited, as were JAK2 and STAT3 activation and several markers of fibrosis, autophagy, senescence, and anti-apoptosis. Conclusions JAK2 and STAT3 are activated in IPF, and their dual inhibition may be an attractive strategy for treating this disease. Electronic supplementary material The online version of this article (10.1186/s12931-018-0728-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Javier Milara
- Department of Pharmacology, Faculty of Medicine, Jaume I University, Castellon de la Plana, Spain. .,Pharmacy Unit, University General Hospital Consortium, Valencia, Spain. .,CIBERES, Health Institute Carlos III, Valencia, Spain. .,Unidad de Investigación Clínica, Consorcio Hospital General Universitario, Avenida Tres Cruces s/n, E-46014, Valencia, Spain.
| | - Gracia Hernandez
- Departmnt of Biotechnology, Valencia Polytechnic University of Valencia, Valencia, Spain
| | - Beatriz Ballester
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Anselm Morell
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Inés Roger
- CIBERES, Health Institute Carlos III, Valencia, Spain
| | - P Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Juan Escrivá
- Thoracic Surgery Unit, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - José M Lloris
- Department of Medicine, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Maria Molina-Molina
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Unidad Funcional de Intersticio Pulmonar (UFIP), Servicio de Neumología, Hospital Universitario de Bellvitge, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Esteban Morcillo
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Departmnt of Biotechnology, Valencia Polytechnic University of Valencia, Valencia, Spain.,Health Research Institute INCLIVA, Valencia, Spain
| | - Julio Cortijo
- CIBERES, Health Institute Carlos III, Valencia, Spain.,Departmnt of Biotechnology, Valencia Polytechnic University of Valencia, Valencia, Spain.,Research and Teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
34
|
Endothelial fibrosis induced by suppressed STAT3 expression mediated by signaling involving the TGF-β1/ALK5/Smad pathway. J Transl Med 2017; 97:1033-1046. [PMID: 28737766 DOI: 10.1038/labinvest.2017.61] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 12/22/2022] Open
Abstract
During systemic inflammatory pathologies, mediators of inflammation circulate in the bloodstream and interact with endothelial cells (ECs), resulting in endothelial dysfunction that maintains and enhances the pathological condition. Inflammatory mediators change the protein expression profile of ECs, which become activated fibroblasts via endothelial-to-mesenchymal transition. This process is characterized by downregulated endothelial proteins and strongly upregulated fibrotic-specific genes and extracellular matrix-forming proteins. The main inductor of endothelial fibrosis is transforming growth factor-β1 (TGF-β1), which acts through the TGF-β1/activin receptor-like kinase 5 (ALK5)/Smads intracellular signaling pathway. The signal transducer and activator of transcription 3 (STAT3) is also involved in fibrosis in several tissues (e.g. heart and vascular system), where STAT3 signaling decreases TGF-β1-induced responses by directly interacting with Smad proteins, suggesting that decreased STAT3 could induce TGF-β1-mediated fibrosis. However, it is unknown if suppressed STAT3 expression induces EC fibrosis through a mechanism involving the TGF-β signaling pathway. The present study evaluated the fibrotic actions of STAT3 suppression in ECs and investigated TGF-β1/ALK5/Smad4 signaling pathway participation. Suppressed STAT3 expression stimulated fibrotic conversion in ECs, as mediated by protein expression reprograming that decreased endothelial marker expression and increased fibrotic and extracellular matrix protein levels. The potential mechanism underlying these changes was dependent on TGF-β1 secretion, the ALK5 activation pathway, and Smad4 translocation into the nucleus. We conclude that suppressed STAT3 expression converts ECs into activated fibroblasts via TGF-β1/ALK5/Smad4 signaling pathway involvement.
Collapse
|
35
|
Yue L, Bartenstein M, Zhao W, Ho WT, Han Y, Murdun C, Mailloux AW, Zhang L, Wang X, Budhathoki A, Pradhan K, Rapaport F, Wang H, Shao Z, Ren X, Steidl U, Levine RL, Zhao ZJ, Verma A, Epling-Burnette PK. Efficacy of ALK5 inhibition in myelofibrosis. JCI Insight 2017; 2:e90932. [PMID: 28405618 DOI: 10.1172/jci.insight.90932] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Myelofibrosis (MF) is a bone marrow disorder characterized by clonal myeloproliferation, aberrant cytokine production, extramedullary hematopoiesis, and bone marrow fibrosis. Although somatic mutations in JAK2, MPL, and CALR have been identified in the pathogenesis of these diseases, inhibitors of the Jak2 pathway have not demonstrated efficacy in ameliorating MF in patients. TGF-β family members are profibrotic cytokines and we observed significant TGF-β1 isoform overexpression in a large cohort of primary MF patient samples. Significant overexpression of TGF-β1 was also observed in murine clonal MPLW515L megakaryocytic cells. TGF-β1 stimulated the deposition of excessive collagen by mesenchymal stromal cells (MSCs) by activating the TGF-β receptor I kinase (ALK5)/Smad3 pathway. MSCs derived from MPLW515L mice demonstrated sustained overproduction of both collagen I and collagen III, effects that were abrogated by ALK5 inhibition in vitro and in vivo. Importantly, use of galunisertib, a clinically active ALK5 inhibitor, significantly improved MF in both MPLW515L and JAK2V617F mouse models. These data demonstrate the role of malignant hematopoietic stem cell (HSC)/TGF-β/MSC axis in the pathogenesis of MF, and provide a preclinical rationale for ALK5 blockade as a therapeutic strategy in MF.
Collapse
Affiliation(s)
- Lanzhu Yue
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Matthias Bartenstein
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Wanke Zhao
- Department of Pathology, Peggy and Stephenson Cancer Center, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Wanting Tina Ho
- Department of Pathology, Peggy and Stephenson Cancer Center, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Ying Han
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Cem Murdun
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Adam W Mailloux
- Translational Science, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Ling Zhang
- Department of Hematopathology and Laboratory Medicine
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Anjali Budhathoki
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Kith Pradhan
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Franck Rapaport
- Leukemia Center, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Huaquan Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiubao Ren
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Ulrich Steidl
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Ross L Levine
- Leukemia Center, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Zhizhuang Joe Zhao
- Department of Pathology, Peggy and Stephenson Cancer Center, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Amit Verma
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | | |
Collapse
|
36
|
Diabetic Microvascular Disease and Pulmonary Fibrosis: The Contribution of Platelets and Systemic Inflammation. Int J Mol Sci 2016; 17:ijms17111853. [PMID: 27834824 PMCID: PMC5133853 DOI: 10.3390/ijms17111853] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 12/29/2022] Open
Abstract
Diabetes is strongly associated with systemic inflammation and oxidative stress, but its effect on pulmonary vascular disease and lung function has often been disregarded. Several studies identified restrictive lung disease and fibrotic changes in diabetic patients and in animal models of diabetes. While microvascular dysfunction is a well-known complication of diabetes, the mechanisms leading to diabetes-induced lung injury have largely been disregarded. We described the potential involvement of diabetes-induced platelet-endothelial interactions in perpetuating vascular inflammation and oxidative injury leading to fibrotic changes in the lung. Changes in nitric oxide synthase (NOS) activation and decreased NO bioavailability in the diabetic lung increase platelet activation and vascular injury and may account for platelet hyperreactivity reported in diabetic patients. Additionally, the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway has been reported to mediate pancreatic islet damage, and is implicated in the onset of diabetes, inflammation and vascular injury. Many growth factors and diabetes-induced agonists act via the JAK/STAT pathway. Other studies reported the contribution of the JAK/STAT pathway to the regulation of the pulmonary fibrotic process but the role of this pathway in the development of diabetic lung fibrosis has not been considered. These observations may open new therapeutic perspectives for modulating multiple pathways to mitigate diabetes onset or its pulmonary consequences.
Collapse
|
37
|
Chen NY, D Collum S, Luo F, Weng T, Le TT, M Hernandez A, Philip K, Molina JG, Garcia-Morales LJ, Cao Y, Ko TC, Amione-Guerra J, Al-Jabbari O, Bunge RR, Youker K, Bruckner BA, Hamid R, Davies J, Sinha N, Karmouty-Quintana H. Macrophage bone morphogenic protein receptor 2 depletion in idiopathic pulmonary fibrosis and Group III pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2016; 311:L238-54. [PMID: 27317687 DOI: 10.1152/ajplung.00142.2016] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/10/2016] [Indexed: 01/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease of unknown etiology. The development of pulmonary hypertension (PH) is considered the single most significant predictor of mortality in patients with chronic lung diseases. The processes that govern the progression and development of fibroproliferative and vascular lesions in IPF are not fully understood. Using human lung explant samples from patients with IPF with or without a diagnosis of PH as well as normal control tissue, we report reduced BMPR2 expression in patients with IPF or IPF+PH. These changes were consistent with dampened P-SMAD 1/5/8 and elevated P-SMAD 2/3, demonstrating reduced BMPR2 signaling and elevated TGF-β activity in IPF. In the bleomycin (BLM) model of lung fibrosis and PH, we also report decreased BMPR2 expression compared with control animals that correlated with vascular remodeling and PH. We show that genetic abrogation or pharmacological inhibition of interleukin-6 leads to diminished markers of fibrosis and PH consistent with elevated levels of BMPR2 and reduced levels of a collection of microRNAs (miRs) that are able to degrade BMPR2. We also demonstrate that isolated bone marrow-derived macrophages from BLM-exposed mice show reduced BMPR2 levels upon exposure with IL6 or the IL6+IL6R complex that are consistent with immunohistochemistry showing reduced BMPR2 in CD206 expressing macrophages from lung sections from IPF and IPF+PH patients. In conclusion, our data suggest that depletion of BMPR2 mediated by a collection of miRs induced by IL6 and subsequent STAT3 phosphorylation as a novel mechanism participating to fibroproliferative and vascular injuries in IPF.
Collapse
Affiliation(s)
- Ning-Yuan Chen
- Department of Biochemistry and Molecular Biology, UTHealth, Houston, Texas
| | - Scott D Collum
- Department of Biochemistry and Molecular Biology, UTHealth, Houston, Texas
| | - Fayong Luo
- Department of Biochemistry and Molecular Biology, UTHealth, Houston, Texas
| | - Tingting Weng
- Department of Biochemistry and Molecular Biology, UTHealth, Houston, Texas
| | - Thuy-Trahn Le
- Department of Biochemistry and Molecular Biology, UTHealth, Houston, Texas
| | | | - Kemly Philip
- Department of Biochemistry and Molecular Biology, UTHealth, Houston, Texas
| | - Jose G Molina
- Department of Biochemistry and Molecular Biology, UTHealth, Houston, Texas
| | | | - Yanna Cao
- Department of Surgery, UTHealth, Houston, Texas
| | - Tien C Ko
- Department of Surgery, UTHealth, Houston, Texas
| | | | - Odeaa Al-Jabbari
- Debakey Heart & Vascular Center, Houston Methodist Hospital, Houston, Texas
| | - Raquel R Bunge
- Debakey Heart & Vascular Center, Houston Methodist Hospital, Houston, Texas
| | - Keith Youker
- Debakey Heart & Vascular Center, Houston Methodist Hospital, Houston, Texas
| | - Brian A Bruckner
- Debakey Heart & Vascular Center, Houston Methodist Hospital, Houston, Texas
| | - Rizwan Hamid
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Jonathan Davies
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Neeraj Sinha
- Debakey Heart & Vascular Center, Houston Methodist Hospital, Houston, Texas
| | | |
Collapse
|
38
|
Halwani R, Sultana A, Al-Kufaidy R, Jamhawi A, Vazquez-Tello A, Al-Muhsen S. Th-17 regulatory cytokines inhibit corticosteroid induced airway structural cells apoptosis. Respir Res 2016; 17:6. [PMID: 26772733 PMCID: PMC4715361 DOI: 10.1186/s12931-015-0307-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 12/04/2015] [Indexed: 12/22/2022] Open
Abstract
Background Although corticosteroid is a powerful anti-inflammatory drug that is used widely to control asthma, still severe asthmatics can develop steroid resistance. Airway fibroblasts are quite resistant to steroids during Idiopathic pulmonary fibrosis (IPF) and fibrosis in asthmatic lungs is not always controlled. Th-17 regulatory cytokine which are elevated in lung tissues of asthmatics were shown to enhance the survival of various types of cells. STAT factors are central to this anti-apoptotic function. However, it is not yet clear whether these cytokines contribute to steroid hypo-responsiveness in asthma. Therefore, in this study, we investigated the ability of Th-17 regulatory cytokines, specifically IL-21, IL22 and IL23, to protect structural airway cells against dexamethasone-induced apoptosis. Methods Primary human fibroblasts, ASM cells, and lung endothelial cells line were treated with IL-21, IL-22, and IL-23 cytokines before incubation with dexamethasone and the level of apoptosis was determined by measuring cellular Annexin-V using Flow cytometry. Results Our data indicated that treatment with Th-17 regulatory cytokines was effective in inhibiting induced apoptosis for both fibroblasts and endothelial cells but not ASM cells. STAT3 phosphorylation levels were also upregulated in fibroblasts and endothelial upon treatment with these cytokines. Interestingly, inhibiting STAT3 phosphorylation abrogated IL-21, IL-22, and IL-23 anti-apoptotic effect on fibroblasts and endothelial cells. Conclusions This data suggest that Th-17 regulatory cytokines may play a critical role in regulating the survival of fibroblasts during asthma, IPF as well as other chronic lung inflammatory diseases leading to enhanced fibrosis. Accordingly, findings of this paper may pave the way for more extensive research on the role of these regulatory cytokines in fibrosis development in various chronic inflammatory diseases.
Collapse
Affiliation(s)
- Rabih Halwani
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia.
| | - Asma Sultana
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia.,Prince Naif Health Research Center, King Saud University, Riyadh, Saudi Arabia
| | - Roua Al-Kufaidy
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia
| | - Amer Jamhawi
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia
| | - Alejandro Vazquez-Tello
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia
| | - Saleh Al-Muhsen
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, P. O. Box 2925, Postal Code 11461, Riyadh, Saudi Arabia
| |
Collapse
|
39
|
Delayed Administration of WP1066, an STAT3 Inhibitor, Ameliorates Radiation-Induced Lung Injury in Mice. Lung 2015; 194:67-74. [PMID: 26563331 DOI: 10.1007/s00408-015-9821-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/30/2015] [Indexed: 12/19/2022]
Abstract
PURPOSE The present study was designed to investigate the effects of WP1066, a specific inhibitor of STAT3 signaling, on radiation-induced lung injury in mice. METHODS C57BL/6J mice were subjected to a single thoracic irradiation of 15 Gy X-ray and WP1066 was administrated through intraperitoneal injection. The early and delayed treatment groups were treated with WP1066 during the first 2 weeks and the second 2 weeks, respectively. The therapeutic effects of WP1066 were evaluated by survival analysis, histological examination, and measurement of inflammatory parameters and collagen deposition. The activation of STAT3 pathway was also estimated by immunohistochemical staining and Western blotting. RESULTS Delayed treatment of WP1066, but not early treatment, prolonged survival time and prevented the development of radiation pneumonitis and the subsequent lung fibrosis in mice. WP1066 treatment also significantly suppressed the activation of STAT3 signaling in the irradiated lung tissues. CONCLUSIONS The activation of STAT3 pathway might play an important part in the pathogenesis of radiation-induced lung injury. The protective effects of delayed treatment of WP1066 suggested STAT3 signaling could be a therapeutic target for radiation pneumonitis.
Collapse
|
40
|
Guzy RD, Stoilov I, Elton TJ, Mecham RP, Ornitz DM. Fibroblast growth factor 2 is required for epithelial recovery, but not for pulmonary fibrosis, in response to bleomycin. Am J Respir Cell Mol Biol 2015; 52:116-28. [PMID: 24988442 DOI: 10.1165/rcmb.2014-0184oc] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The pathogenesis of pulmonary fibrosis involves lung epithelial injury and aberrant proliferation of fibroblasts, and results in progressive pulmonary scarring and declining lung function. In vitro, fibroblast growth factor (FGF) 2 promotes myofibroblast differentiation and proliferation in cooperation with the profibrotic growth factor, transforming growth factor-β1, but the in vivo requirement for FGF2 in the development of pulmonary fibrosis is not known. The bleomycin model of lung injury and pulmonary fibrosis was applied to Fgf2 knockout (Fgf2(-/-)) and littermate control mice. Weight loss, mortality, pulmonary fibrosis, and histology were analyzed after a single intranasal dose of bleomycin. Inflammation was evaluated in bronchoalveolar lavage (BAL) fluid, and epithelial barrier integrity was assessed by measuring BAL protein and Evans Blue dye permeability. Fgf2 is expressed in mouse and human lung epithelial and inflammatory cells, and, in response to bleomycin, Fgf2(-/-) mice have significantly increased mortality and weight loss. Analysis of BAL fluid and histology show that pulmonary fibrosis is unaltered, but Fgf2(-/-) mice fail to efficiently resolve inflammation, have increased BAL cellularity, and, importantly, deficient recovery of epithelial integrity. Fgf2(-/-) mice similarly have deficient recovery of club cell secretory protein(+) bronchial epithelium in response to naphthalene. We conclude that FGF2 is not required for bleomycin-induced pulmonary fibrosis, but rather is essential for epithelial repair and maintaining epithelial integrity after bleomycin-induced lung injury in mice. These data identify that FGF2 acts as a protective growth factor after lung epithelial injury, and call into question the role of FGF2 as a profibrotic growth factor in vivo.
Collapse
Affiliation(s)
- Robert D Guzy
- Departments of 1 Internal Medicine, Division of Pulmonary and Critical Care Medicine
| | | | | | | | | |
Collapse
|
41
|
Amara S, Lopez K, Banan B, Brown SK, Whalen M, Myles E, Ivy MT, Johnson T, Schey KL, Tiriveedhi V. Synergistic effect of pro-inflammatory TNFα and IL-17 in periostin mediated collagen deposition: potential role in liver fibrosis. Mol Immunol 2014; 64:26-35. [PMID: 25467797 DOI: 10.1016/j.molimm.2014.10.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/19/2014] [Accepted: 10/25/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND The pro-inflammatory cytokines, tumor necrosis factor (TNF)-α, and interleukin (IL)-17, have been implicated in the pathogenesis of liver fibrosis. In this study, we investigated the role of TNFα and IL-17 toward induction of profibrotic factor, periostin. METHODS HepG2 cells were cultured and treated with inflammatory cytokines, TNFα and IL-17. Computational promoter sequence analysis of the periostin promoter was performed to define the putative binding sites for transcription factors. Transcription factors were analyzed by Western blot and Chromatin Immunoprecipitation. Periostin and transcription factor expression analysis was performed by RT-PCR, Western blot, and fluorescence microscopy. Type I collagen expression from fibroblast cultures was analyzed by Western blot and Sircol soluble collagen assay. RESULTS Activation of HepG2 Cells with TNFα and IL-17 enhanced the expression of periostin (3.5 and 4.4 fold, respectively p<0.05) compared to untreated cells. However, combined treatment with both TNFα and IL-17 at similar concentration demonstrated a 13.3 fold increase in periostin (p<0.01), thus suggesting a synergistic role of these cytokines. Periostin promoter analysis and specific siRNA knock-down revealed that TNFα induces periostin through cJun, while IL-17 induced periostin via STAT-3 signaling mechanisms. Treatment of the supernatant from the cytokine activated HepG2 cells on fibroblast cultures induced enhanced expression of type I collagen (>9.1 fold, p<0.01), indicative of a direct fibrogenic effect of TNFα and IL-17. CONCLUSION TNFα and IL-17 induced fibrogenesis through cJun and STAT-3 mediated expression of profibrotic biomarker, periostin. Therefore, periostin might serve as a novel biomarker in early diagnosis of liver fibrosis.
Collapse
Affiliation(s)
- Suneetha Amara
- Department of Medicine, Mercy Hospital, St Louis, MO, USA
| | - Karina Lopez
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Babak Banan
- Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Sade-Kemi Brown
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Margaret Whalen
- Department of Chemistry, Tennessee State University, Nashville, TN, USA
| | - Elbert Myles
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Michael T Ivy
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Terrance Johnson
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Kevin L Schey
- Department of Biochemistry, Mass Spectrometry and Proteomics Research Core, Vanderbilt University, Nashville, TN, USA
| | | |
Collapse
|
42
|
Le TTT, Karmouty-Quintana H, Melicoff E, Le TTT, Weng T, Chen NY, Pedroza M, Zhou Y, Davies J, Philip K, Molina J, Luo F, George AT, Garcia-Morales LJ, Bunge RR, Bruckner BA, Loebe M, Seethamraju H, Agarwal SK, Blackburn MR. Blockade of IL-6 Trans signaling attenuates pulmonary fibrosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:3755-68. [PMID: 25172494 PMCID: PMC4169999 DOI: 10.4049/jimmunol.1302470] [Citation(s) in RCA: 231] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 07/31/2014] [Indexed: 12/20/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease with progressive fibrosis and death within 2-3 y of diagnosis. IPF incidence and prevalence rates are increasing annually with few effective treatments available. Inhibition of IL-6 results in the attenuation of pulmonary fibrosis in mice. It is unclear whether this is due to blockade of classical signaling, mediated by membrane-bound IL-6Rα, or trans signaling, mediated by soluble IL-6Rα (sIL-6Rα). Our study assessed the role of sIL-6Rα in IPF. We demonstrated elevations of sIL-6Rα in IPF patients and in mice during the onset and progression of fibrosis. We demonstrated that protease-mediated cleavage from lung macrophages was important in production of sIL-6Rα. In vivo neutralization of sIL-6Rα attenuated pulmonary fibrosis in mice as seen by reductions in myofibroblasts, fibronectin, and collagen in the lung. In vitro activation of IL-6 trans signaling enhanced fibroblast proliferation and extracellular matrix protein production, effects relevant in the progression of pulmonary fibrosis. Taken together, these findings demonstrate that the production of sIL-6Rα from macrophages in the diseased lung contributes to IL-6 trans signaling that in turn influences events crucial in pulmonary fibrosis.
Collapse
Affiliation(s)
- Thanh-Thuy T Le
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030; University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030
| | | | - Thanh-Truc T Le
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030
| | - Tingting Weng
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030
| | - Ning-Yuan Chen
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030
| | - Mesias Pedroza
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030; University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030; Biology of Inflammation Center, Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Yang Zhou
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030
| | - Jonathan Davies
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Kemly Philip
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030
| | - Jose Molina
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030
| | - Fayong Luo
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030
| | - Anuh T George
- Biology of Inflammation Center, Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Luis J Garcia-Morales
- Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX 77030; and
| | - Raquel R Bunge
- Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX 77030; and
| | - Brian A Bruckner
- Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX 77030; and Methodist J.C. Walter Jr. Transplant Center, The Methodist Hospital, Houston, TX 77030
| | - Matthias Loebe
- Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX 77030; and Methodist J.C. Walter Jr. Transplant Center, The Methodist Hospital, Houston, TX 77030
| | - Harish Seethamraju
- Methodist J.C. Walter Jr. Transplant Center, The Methodist Hospital, Houston, TX 77030
| | - Sandeep K Agarwal
- Biology of Inflammation Center, Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Michael R Blackburn
- Department of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030; University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030;
| |
Collapse
|
43
|
Richards CD. The enigmatic cytokine oncostatin m and roles in disease. ISRN INFLAMMATION 2013; 2013:512103. [PMID: 24381786 PMCID: PMC3870656 DOI: 10.1155/2013/512103] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 09/29/2013] [Indexed: 12/11/2022]
Abstract
Oncostatin M is a secreted cytokine involved in homeostasis and in diseases involving chronic inflammation. It is a member of the gp130 family of cytokines that have pleiotropic functions in differentiation, cell proliferation, and hematopoetic, immunologic, and inflammatory networks. However, Oncostatin M also has activities novel to mediators of this cytokine family and others and may have fundamental roles in mechanisms of inflammation in pathology. Studies have explored Oncostatin M functions in cancer, bone metabolism, liver regeneration, and conditions with chronic inflammation including rheumatoid arthritis, lung and skin inflammatory disease, atherosclerosis, and cardiovascular disease. This paper will review Oncostatin M biology in a historical fashion and focus on its unique activities, in vitro and in vivo, that differentiate it from other cytokines and inspire further study or consideration in therapeutic approaches.
Collapse
Affiliation(s)
- Carl D. Richards
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street, West, Hamilton, ON, Canada L8S 4K1
| |
Collapse
|
44
|
Obeidat M, Miller S, Probert K, Billington CK, Henry AP, Hodge E, Nelson CP, Stewart CE, Swan C, Wain LV, Artigas MS, Melén E, Ushey K, Hao K, Lamontagne M, Bossé Y, Postma DS, Tobin MD, Sayers I, Hall IP. GSTCD and INTS12 regulation and expression in the human lung. PLoS One 2013; 8:e74630. [PMID: 24058608 PMCID: PMC3776747 DOI: 10.1371/journal.pone.0074630] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/05/2013] [Indexed: 12/29/2022] Open
Abstract
Genome-Wide Association Study (GWAS) meta-analyses have identified a strong association signal for lung function, which maps to a region on 4q24 containing two oppositely transcribed genes: glutathione S-transferase, C-terminal domain containing (GSTCD) and integrator complex subunit 12 (INTS12). Both genes were found to be expressed in a range of human airway cell types. The promoter regions and transcription start sites were determined in mRNA from human lung and a novel splice variant was identified for each gene. We obtained the following evidence for GSTCD and INTS12 co-regulation and expression: (i) correlated mRNA expression was observed both via Q-PCR and in a lung expression quantitative trait loci (eQTL) study, (ii) induction of both GSTCD and INTS12 mRNA expression in human airway smooth muscle cells was seen in response to TGFβ1, (iii) a lung eQTL study revealed that both GSTCD and INTS12 mRNA levels positively correlate with percent predicted FEV1, and (iv) FEV1 GWAS associated SNPs in 4q24 were found to act as an eQTL for INTS12 in a number of tissues. In fixed sections of human lung tissue, GSTCD protein expression was ubiquitous, whereas INTS12 expression was predominantly in epithelial cells and pneumocytes. During human fetal lung development, GSTCD protein expression was observed to be highest at the earlier pseudoglandular stage (10-12 weeks) compared with the later canalicular stage (17-19 weeks), whereas INTS12 expression levels did not alter throughout these stages. Knowledge of the transcriptional and translational regulation and expression of GSTCD and INTS12 provides important insights into the potential role of these genes in determining lung function. Future work is warranted to fully define the functions of INTS12 and GSTCD.
Collapse
Affiliation(s)
- Ma’en Obeidat
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
- James Hogg Research Centre, Institute for Heart and Lung Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Suzanne Miller
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Kelly Probert
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Charlotte K. Billington
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Amanda P. Henry
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Emily Hodge
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Carl P. Nelson
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Ceri E. Stewart
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Caroline Swan
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Louise V. Wain
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - María Soler Artigas
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet and Sachs’ Children’s Hospital, Stockholm, Sweden
| | - Kevin Ushey
- James Hogg Research Centre, Institute for Heart and Lung Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Maxime Lamontagne
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Québec City, Canada
| | - Yohan Bossé
- Department of Molecular Medicine, Laval University, Québec City, Canada
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Québec City, Canada
| | - Dirkje S. Postma
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin D. Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health Research (NIHR) Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | - Ian Sayers
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Ian P. Hall
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| |
Collapse
|
45
|
Aumiller V, Balsara N, Wilhelm J, Günther A, Königshoff M. WNT/β-catenin signaling induces IL-1β expression by alveolar epithelial cells in pulmonary fibrosis. Am J Respir Cell Mol Biol 2013; 49:96-104. [PMID: 23526221 DOI: 10.1165/rcmb.2012-0524oc] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease of unknown etiology. It is characterized by alterations of the alveolar epithelium, myofibroblast activation, and increased extracellular matrix deposition. Recently, reactivation of the developmental WNT/β-catenin pathway has been linked with pulmonary fibrosis. The cell-specific mechanisms and mediators of WNT/β-catenin signaling in the lung, however, remain elusive. Here, we applied an unbiased gene expression screen to identify epithelial cell-specific mediators of WNT/β-catenin signaling. We found the proinflammatory cytokine IL-1β to be one of the most up-regulated genes in primary murine alveolar epithelial Type II (ATII) cells after WNT3a treatment. Increased transcript and protein expression of IL-1β upon WNT3a treatment was further detected in primary ATII cells by quantitative RT-PCR (log fold change, 2.0 ± 0.5) and ELISA (1.8-fold increase). We observed significant up-regulation of IL-1β and IL-6 in bronchoalveolar lavage fluid (BALF) in bleomycin-induced lung fibrosis in vivo. Importantly, primary fibrotic ATII cells isolated from lungs subjected to bleomycin secreted enhanced IL-1β and IL-6 in vitro. Furthermore, the orotracheal application of recombinant WNT protein in the Tcf optimal promoter (TOP)-β-galactosidase reporter animals led to WNT/β-catenin activation in epithelial cells, along with significant increases in IL-1β and IL-6 in vivo (2.7-fold and 6.0-fold increases, respectively). Finally, we found increased WNT3a protein in fibrotic alveolar epithelia, accompanied by enhanced IL-1β and IL-6 concentrations in BALF from patients with IPF. Taken together, our findings reveal that the alveolar epithelium is a relevant source of proinflammatory cytokines induced by active WNT/β-catenin in pulmonary fibrosis. Thus, WNT/interleukin signaling represents a novel link between developmental pathway reactivation and inflammation in the development of pulmonary fibrosis.
Collapse
Affiliation(s)
- Verena Aumiller
- Comprehensive Pneumology Center, Ludwig Maximilians University, University Hospital Grosshadern, and Helmholtz Zentrum München, Munich, Germany
| | | | | | | | | |
Collapse
|
46
|
Hügle T, O'Reilly S, Simpson R, Kraaij MD, Bigley V, Collin M, Krippner-Heidenreich A, van Laar JM. Tumor necrosis factor-costimulated T lymphocytes from patients with systemic sclerosis trigger collagen production in fibroblasts. ACTA ACUST UNITED AC 2013; 65:481-91. [PMID: 23045159 DOI: 10.1002/art.37738] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 10/02/2012] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The role of tumor necrosis factor (TNF) in systemic sclerosis (SSc) remains controversial. The present study was undertaken to investigate the influence of TNF receptor (TNFR)-costimulated lymphocytes on collagen expression in fibroblasts. METHODS TNFR expression on mononuclear cells from the dermis and blood of SSc patients was assessed by flow cytometry. Peripheral blood CD3+ lymphocytes were activated with CD3/CD28 beads and costimulated with TNFR-selective variants. Expression of interleukin-6 (IL-6), soluble IL-6 receptor (sIL-6R), IL-10, and IL-13 was detected by enzyme-linked immunosorbent assay or quantitative reverse transcription-polymerase chain reaction. Healthy fibroblasts were incubated with conditioned media from TNFR-costimulated T lymphocytes, and type I collagen expression was quantified. RESULTS TNFRI and TNFRII were up-regulated on dermal T lymphocytes from patients with diffuse cutaneous SSc. TNFRII expression correlated with skin thickening. After CD3/CD28 activation, peripheral blood lymphocytes from SSc patients produced more IL-6, sIL-6R, and IL-13 compared to healthy lymphocytes. Costimulation with TNFRI-selective ligands and soluble TNF further increased IL-6 expression, whereas costimulation with TNFRII led to greater release of sIL-6R. IL-10 expression, which normally occurs after TNFRII costimulation, was impaired in SSc T cells. Supernatants of TNF-costimulated SSc lymphocytes induced higher type I collagen expression in fibroblasts, which was partially reversible by dual inhibition of IL-6 and IL-13. Expression of TNFR and IL-6 in the dermis was reversible in a patient who received lymphoablative therapy prior to autologous hematopoietic stem cell transplantation. CONCLUSION TNF-costimulated T lymphocytes from SSc patients have a propensity to secrete profibrotic cytokines, while the ability to produce IL-10 is weakened. These results suggest that T lymphocytes in SSc support fibrosis, but might lack the capacity to resolve inflammation.
Collapse
|