1
|
Tchoukalova YD, Nair AA, Chen X, Zhang N, Myers CE, Badreldin A, Dudakovic A, Lott LG, Rebecca AM, Yi J, Cornella JL, van Wijnen AJ. Cell type specific differences in transcriptome profiles of adipose derived stem cells and vaginal fibroblasts in patients with pelvic organ prolapse. Gene 2025; 942:149230. [PMID: 39814191 DOI: 10.1016/j.gene.2025.149230] [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: 09/18/2024] [Revised: 12/16/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
This study examined the molecular phenotypes of adipose-derived stem cells (ASCs) and vaginal fibroblasts (VFBs) and assessed whether pelvic organ prolapse (POP) affects their biological properties. We performed RNA sequencing of paired ASCs and VFBs from six patients with POP and six controls (CTRL). The transcriptomes of POP and CTRL in either ASCs or VFBs were compared (DESeq2, false discovery rate (FDR) < 0.05) to identify differentially expressed genes (DEGs). The transcriptomes of VFBs were compared between POP and CTRL (non-adjusted p < 0.01) followed by Ingenuity Pathway Analysis on DEGs considering that pathways with FDR < 0.05 could be pathogenic. We also performed a pairwise comparison after combining the gene expression data of POP and CTRL for ASCs and VFBs to identify cell type specific DEGs and analyzed the functional associations among them (STRING platform). We found no DEGs between POP and CTRL in ASCs and VFBs. Less stringent statistical analysis of VFBs transcriptome showed 23 genes with higher and 29 genes with lower expression in POP compared to CTRL. Among the latter were five genes involved in the synaptogenesis pathway found to be significant. We were only able to validate POP related differences for very low density receptor (VLDLR). We found 508 DEGs with 4-fold difference between ASCs and VFBs (both POP and CTRL groups combined for each cell type) which formed cell type distinct functional networks including Homeobox transcription factors, extracellular matrix (ECM) related proteins, and growth factors. In summary, this study showed that the fibroblastic transcriptomes of VFBs and ASCs are different, and this cell type-specific difference is more prominent that the disease related effects of POP.
Collapse
Affiliation(s)
- Yourka D Tchoukalova
- Head and Neck Regenerative Medicine Laboratory, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Asha A Nair
- Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Xianfeng Chen
- Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Nan Zhang
- Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Cheryl E Myers
- Head and Neck Regenerative Medicine Laboratory, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Amr Badreldin
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Lott G Lott
- Head and Neck Regenerative Medicine Laboratory, Mayo Clinic Arizona, Scottsdale, AZ, USA; Department of Otolaryngology - Head and Neck Surgery, Division of Laryngology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Alanna M Rebecca
- Division of Plastic Surgery, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Johnny Yi
- Division of Gynecology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | | | | |
Collapse
|
2
|
Zhang T, Wang Y, Sun Y, Song M, Pang J, Wang M, Zhang Z, Yang P, Chen Y, Qi X, Zhou H, Han Z, Xing Y, Liu Y, Li B, Liu J, Yang J, Wang J. Proteome, Lysine Acetylome, and Succinylome Identify Posttranslational Modification of STAT1 as a Novel Drug Target in Silicosis. Mol Cell Proteomics 2024; 23:100770. [PMID: 38641226 PMCID: PMC11107463 DOI: 10.1016/j.mcpro.2024.100770] [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: 07/09/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024] Open
Abstract
Inhalation of crystalline silica dust induces incurable lung damage, silicosis, and pulmonary fibrosis. However, the mechanisms of the lung injury remain poorly understood, with limited therapeutic options aside from lung transplantation. Posttranslational modifications can regulate the function of proteins and play an important role in studying disease mechanisms. To investigate changes in posttranslational modifications of proteins in silicosis, combined quantitative proteome, acetylome, and succinylome analyses were performed with lung tissues from silica-injured and healthy mice using liquid chromatography-mass spectrometry. Combined analysis was applied to the three omics datasets to construct a protein landscape. The acetylation and succinylation of the key transcription factor STAT1 were found to play important roles in the silica-induced pathophysiological changes. Modulating the acetylation level of STAT1 with geranylgeranylacetone effectively inhibited the progression of silicosis. This report revealed a comprehensive landscape of posttranslational modifications in silica-injured mouse and presented a novel therapeutic strategy targeting the posttranslational level for silica-induced lung diseases.
Collapse
Affiliation(s)
- Tiantian Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yiyang Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Youliang Sun
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Institute of Basic Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Meiyue Song
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Junling Pang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Mingyao Wang
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Zhe Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yiling Chen
- Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Xianmei Qi
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Huan Zhou
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Zhenzong Han
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yanjiang Xing
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Ying Liu
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Baicun Li
- Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Jiangfeng Liu
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Juntao Yang
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Jing Wang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
Gefitinib and fostamatinib target EGFR and SYK to attenuate silicosis: a multi-omics study with drug exploration. Signal Transduct Target Ther 2022; 7:157. [PMID: 35551173 PMCID: PMC9098425 DOI: 10.1038/s41392-022-00959-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/21/2022] [Accepted: 02/27/2022] [Indexed: 02/06/2023] Open
Abstract
Silicosis is the most prevalent and fatal occupational disease with no effective therapeutics, and currently used drugs cannot reverse the disease progress. Worse still, there are still challenges to be addressed to fully decipher the intricated pathogenesis. Thus, specifying the essential mechanisms and targets in silicosis progression then exploring anti-silicosis pharmacuticals are desperately needed. In this work, multi-omics atlas was constructed to depict the pivotal abnormalities of silicosis and develop targeted agents. By utilizing an unbiased and time-resolved analysis of the transcriptome, proteome and phosphoproteome of a silicosis mouse model, we have verified the significant differences in transcript, protein, kinase activity and signaling pathway level during silicosis progression, in which the importance of essential biological processes such as macrophage activation, chemotaxis, immune cell recruitment and chronic inflammation were emphasized. Notably, the phosphorylation of EGFR (p-EGFR) and SYK (p-SYK) were identified as potential therapeutic targets in the progression of silicosis. To inhibit and validate these targets, we tested fostamatinib (targeting SYK) and Gefitinib (targeting EGFR), and both drugs effectively ameliorated pulmonary dysfunction and inhibited the progression of inflammation and fibrosis. Overall, our drug discovery with multi-omics approach provides novel and viable therapeutic strategies for the treatment of silicosis.
Collapse
|
4
|
Early Identification, Accurate Diagnosis, and Treatment of Silicosis. Can Respir J 2022; 2022:3769134. [PMID: 35509892 PMCID: PMC9061058 DOI: 10.1155/2022/3769134] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/05/2022] [Accepted: 04/15/2022] [Indexed: 12/04/2022] Open
Abstract
Silicosis is a global problem, and it has brought about great burdens to society and patients' families. The etiology of silicosis is clear, preventable, and controllable, but the onset is hidden and the duration is long. Thus, it is difficult to diagnose it early and treat it effectively, leaving workers unaware of the consequences of dust exposure. As such, a lack of details in the work history and a slow progression of lung disease contribute to the deterioration of patients until silicosis has advanced to fibrosis. These issues are the key factors impeding the diagnosis and the treatment of silicosis. This article reviews the literature on the early identification, diagnosis, and treatment of silicosis as well as analyzes the difficulties in the diagnosis and the treatment of silicosis and discusses its direction of future development.
Collapse
|
5
|
Oxamate Attenuates Glycolysis and ER Stress in Silicotic Mice. Int J Mol Sci 2022; 23:ijms23063013. [PMID: 35328434 PMCID: PMC8953611 DOI: 10.3390/ijms23063013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Glycolysis and ER stress have been considered important drivers of pulmonary fibrosis. However, it is not clear whether glycolysis and ER stress are interconnected and if those interconnections regulate the development of pulmonary fibrosis. Our previous studies found that the expression of LDHA, a key enzyme involved in glycolysis, was increased in silica-induced macrophages and silicotic models, and it was closely related to silicosis fibrosis by participating in inflammatory response. However, whether pharmacological inhibition of LDHA is beneficial to the amelioration of silicosis fibrosis remains unclear. In this study, we investigated the effects of oxamate, a potent inhibitor of LDHA, on the regulation of glycolysis and ER stress in alveolar macrophages and silicotic mice. We found that silica induced the upregulation of glycolysis and the expression of key enzymes directly involved in ER stress in NR8383 macrophages. However, treatment of the macrophages and silicotic mice with oxamate attenuated glycolysis and ER stress by inhibiting LDHA, causing a decrease in the production of lactate. Therefore, oxamate demonstrated an anti-fibrotic role by reducing glycolysis and ER stress in silicotic mice.
Collapse
|
6
|
Mao N, Yang H, Yin J, Li Y, Jin F, Li T, Yang X, Sun Y, Liu H, Xu H, Yang F. Glycolytic Reprogramming in Silica-Induced Lung Macrophages and Silicosis Reversed by Ac-SDKP Treatment. Int J Mol Sci 2021; 22:ijms221810063. [PMID: 34576239 PMCID: PMC8465686 DOI: 10.3390/ijms221810063] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/15/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
Glycolytic reprogramming is an important metabolic feature in the development of pulmonary fibrosis. However, the specific mechanism of glycolysis in silicosis is still not clear. In this study, silicotic models and silica-induced macrophage were used to elucidate the mechanism of glycolysis induced by silica. Expression levels of the key enzymes in glycolysis and macrophage activation indicators were analyzed by Western blot, qRT-PCR, IHC, and IF analyses, and by using a lactate assay kit. We found that silica promotes the expression of the key glycolysis enzymes HK2, PKM2, LDHA, and macrophage activation factors iNOS, TNF-α, Arg-1, IL-10, and MCP1 in silicotic rats and silica-induced NR8383 macrophages. The enhancement of glycolysis and macrophage activation induced by silica was reduced by Ac-SDKP or siRNA-Ldha treatment. This study suggests that Ac-SDKP treatment can inhibit glycolytic reprogramming in silica-induced lung macrophages and silicosis.
Collapse
Affiliation(s)
- Na Mao
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (N.M.); (H.Y.); (J.Y.); (Y.L.); (F.J.); (H.L.)
| | - Honghao Yang
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (N.M.); (H.Y.); (J.Y.); (Y.L.); (F.J.); (H.L.)
| | - Jie Yin
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (N.M.); (H.Y.); (J.Y.); (Y.L.); (F.J.); (H.L.)
| | - Yaqian Li
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (N.M.); (H.Y.); (J.Y.); (Y.L.); (F.J.); (H.L.)
| | - Fuyu Jin
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (N.M.); (H.Y.); (J.Y.); (Y.L.); (F.J.); (H.L.)
| | - Tian Li
- Hebei Key Laboratory for Chronic Diseases, Basic Medical College, North China University of Science and Technology, Tangshan 063210, China; (T.L.); (X.Y.); (Y.S.)
| | - Xinyu Yang
- Hebei Key Laboratory for Chronic Diseases, Basic Medical College, North China University of Science and Technology, Tangshan 063210, China; (T.L.); (X.Y.); (Y.S.)
| | - Ying Sun
- Hebei Key Laboratory for Chronic Diseases, Basic Medical College, North China University of Science and Technology, Tangshan 063210, China; (T.L.); (X.Y.); (Y.S.)
| | - Heliang Liu
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (N.M.); (H.Y.); (J.Y.); (Y.L.); (F.J.); (H.L.)
| | - Hong Xu
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (N.M.); (H.Y.); (J.Y.); (Y.L.); (F.J.); (H.L.)
- Correspondence: (H.X.); (F.Y.); Tel.: +86-15133967479 (H.X.); +86-18832571018 (F.Y.); Fax: +86-315-8805522 (F.Y.)
| | - Fang Yang
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (N.M.); (H.Y.); (J.Y.); (Y.L.); (F.J.); (H.L.)
- Correspondence: (H.X.); (F.Y.); Tel.: +86-15133967479 (H.X.); +86-18832571018 (F.Y.); Fax: +86-315-8805522 (F.Y.)
| |
Collapse
|
7
|
Zhang ZQ, Tian HT, Liu H, Xie R. The role of macrophage-derived TGF-β1 on SiO 2-induced pulmonary fibrosis: A review. Toxicol Ind Health 2021; 37:240-250. [PMID: 33588701 DOI: 10.1177/0748233721989896] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Silicosis is an occupational fibrotic lung disease caused by inhaling large amounts of crystalline silica dust. Transforming growth factor-β1 (TGF-β1), which is secreted from macrophages, has an important role in the development of this disease. Macrophages can recognize and capture silicon dust, undergo M2 polarization, synthesize TGF-β1 precursors, and secrete them out of the cell where they are activated. Activated TGF-β1 induces cells from different sources, transforming them into myofibroblasts through autocrine and paracrine mechanisms, ultimately causing silicosis. These processes involve complex molecular events, which are not yet fully understood. This systematic summary may further elucidate the location and development of pulmonary fibrosis in the formation of silicosis. In this review, we discussed the proposed cellular and molecular mechanisms of production, secretion, activation of TGF-β1, as well as the mechanisms through which TGF-β1 induces cells from three different sources into myofibroblasts during the pathogenesis of silicosis. This study furthers the medical understanding of the pathogenesis and theoretical basis for diagnosing silicosis, thereby promoting silicosis prevention and treatment.
Collapse
Affiliation(s)
- Zhao-Qiang Zhang
- Department of Public Health, 74496Jining Medical University, Jining, China
| | - Hai-Tao Tian
- Department of Public Health, 74496Jining Medical University, Jining, China.,Jining No. 1 People's Hospital, Jining, China
| | - Hu Liu
- Department of Public Health, 74496Jining Medical University, Jining, China
| | - Ruining Xie
- Department of Public Health, 74496Jining Medical University, Jining, China
| |
Collapse
|
8
|
Bo C, Geng X, Zhang J, Sai L, Zhang Y, Yu G, Zhang Z, Liu K, Du Z, Peng C, Jia Q, Shao H. Comparative proteomic analysis of silica-induced pulmonary fibrosis in rats based on tandem mass tag (TMT) quantitation technology. PLoS One 2020; 15:e0241310. [PMID: 33119648 PMCID: PMC7595299 DOI: 10.1371/journal.pone.0241310] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022] Open
Abstract
Silicosis is a systemic disease characterized by chronic persistent inflammation and incurable pulmonary fibrosis with the underlying molecular mechanisms to be fully elucidated. In this study, we employed tandem mass tag (TMT) based on quantitative proteomics technology to detect differentially expressed proteins (DEPs) in lung tissues of silica-exposed rats. A total of 285 DEPs (145 upregulated and 140 downregulated) were identified. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to predict the biological pathway and functional classification of the proteins. Results showed that these DEPs were mainly enriched in the phagosome, lysosome function, complement and the coagulation cascade, glutathione metabolism, focal adhesion and ECM-receptor interactions. To validate the proteomics data, we selected and analyzed the expression trends of six proteins including CD14, PSAP, GM2A, COL1A1, ITGA8 and CLDN5 using parallel reaction monitoring (PRM). The consistent result between PRM and TMT indicated the reliability of our proteomic data. These findings will help to reveal the pathogenesis of silicosis and provide potential therapeutic targets. Data are available via ProteomeXchange with identifier PXD020625.
Collapse
Affiliation(s)
- Cunxiang Bo
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Xiao Geng
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Juan Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Linlin Sai
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Yu Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Gongchang Yu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Zhenling Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Kai Liu
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Ji’nan, Shandong, China
| | - Zhongjun Du
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Cheng Peng
- Queensland Alliance for Environmental Health Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
- * E-mail: (QJ); (HS)
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
- * E-mail: (QJ); (HS)
| |
Collapse
|
9
|
Cong L, Li T, Wang H, Wu Y, Wang S, Zhao Y, Zhang G, Duan J. IL-17A-producing T cells exacerbate fine particulate matter-induced lung inflammation and fibrosis by inhibiting PI3K/Akt/mTOR-mediated autophagy. J Cell Mol Med 2020; 24:8532-8544. [PMID: 32643865 PMCID: PMC7412687 DOI: 10.1111/jcmm.15475] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/24/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022] Open
Abstract
Fine particulate matter (PM2.5) is the primary air pollutant that is able to induce airway injury. Compelling evidence has shown the involvement of IL-17A in lung injury, while its contribution to PM2.5-induced lung injury remains largely unknown. Here, we probed into the possible role of IL-17A in mouse models of PM2.5-induced lung injury. Mice were instilled with PM2.5 to construct a lung injury model. Flow cytometry was carried out to isolate γδT and Th17 cells. ELISA was adopted to detect the expression of inflammatory factors in the supernatant of lavage fluid. Primary bronchial epithelial cells (mBECs) were extracted, and the expression of TGF signalling pathway-, autophagy- and PI3K/Akt/mTOR signalling pathway-related proteins in mBECs was detected by immunofluorescence assay and Western blot analysis. The mitochondrial function was also evaluated. PM2.5 aggravated the inflammatory response through enhancing the secretion of IL-17A by γδT/Th17 cells. Meanwhile, PM2.5 activated the TGF signalling pathway and induced EMT progression in bronchial epithelial cells, thereby contributing to pulmonary fibrosis. Besides, PM2.5 suppressed autophagy of bronchial epithelial cells by up-regulating IL-17A, which in turn activated the PI3K/Akt/mTOR signalling pathway. Furthermore, IL-17A impaired the energy metabolism of airway epithelial cells in the PM2.5-induced models. This study suggested that PM2.5 could inhibit autophagy of bronchial epithelial cells and promote pulmonary inflammation and fibrosis by inducing the secretion of IL-17A in γδT and Th17 cells and regulating the PI3K/Akt/mTOR signalling pathway.
Collapse
Affiliation(s)
- Lu‐Hong Cong
- Department of EmergencyChina‐Japan Friendship HospitalBeijingChina
| | - Tao Li
- Surgical Intensive Care UnitChina‐Japan Friendship HospitalBeijingChina
| | - Hui Wang
- Surgical Intensive Care UnitChina‐Japan Friendship HospitalBeijingChina
| | - Yi‐Na Wu
- Surgical Intensive Care UnitChina‐Japan Friendship HospitalBeijingChina
| | - Shu‐Peng Wang
- Surgical Intensive Care UnitChina‐Japan Friendship HospitalBeijingChina
| | - Yu‐Yue Zhao
- Surgical Intensive Care UnitChina‐Japan Friendship HospitalBeijingChina
| | - Guo‐Qiang Zhang
- Department of EmergencyChina‐Japan Friendship HospitalBeijingChina
| | - Jun Duan
- Surgical Intensive Care UnitChina‐Japan Friendship HospitalBeijingChina
| |
Collapse
|
10
|
Gao X, Xu D, Li S, Wei Z, Li S, Cai W, Mao N, Jin F, Li Y, Yi X, Liu H, Xu H, Yang F. Pulmonary Silicosis Alters MicroRNA Expression in Rat Lung and miR-411-3p Exerts Anti-fibrotic Effects by Inhibiting MRTF-A/SRF Signaling. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:851-865. [PMID: 32464548 PMCID: PMC7256439 DOI: 10.1016/j.omtn.2020.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/15/2020] [Accepted: 05/06/2020] [Indexed: 02/08/2023]
Abstract
To identify potential therapeutic targets for pulmonary fibrosis induced by silica, we studied the effects of this disease on the expression of microRNAs (miRNAs) in the lung. Rattus norvegicus pulmonary silicosis models were used in conjunction with high-throughput screening of lung specimens to compare the expression of miRNAs in control and pulmonary silicosis tissues. A total of 70 miRNAs were found to be differentially expressed between control and pulmonary silicosis tissues. This included 41 miRNAs that were upregulated and 29 that were downregulated relative to controls. Among them, miR-292-5p, miR-155-3p, miR-1193-3p, miR-411-3p, miR-370-3p, and miR-409a-5p were found to be similarly altered in rat lung and transforming growth factor (TGF)-β1-induced cultured fibroblasts. Using miRNA mimics and inhibitors, we found that miR-1193-3p, miR-411-3p, and miR-370-3p exhibited potent anti-fibrotic effects, while miR-292-5p demonstrated pro-fibrotic effects in TGF-β1-stimulated lung fibroblasts. Moreover, we also found that miR-411-3p effectively reduced pulmonary silicosis in the mouse lung by regulating Mrtfa expression, as demonstrated using biochemical and histological assays. In conclusion, our findings indicate that miRNA expression is perturbed in pulmonary silicosis and suggest that therapeutic interventions targeting specific miRNAs might be effective in the treatment of this occupational disease.
Collapse
Affiliation(s)
- Xuemin Gao
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 Hebei, China
| | - Dingjie Xu
- Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, 063210 Hebei, China
| | - Shumin Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 Hebei, China
| | - Zhongqiu Wei
- Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, 063210 Hebei, China
| | - Shifeng Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 Hebei, China
| | - Wenchen Cai
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 Hebei, China
| | - Na Mao
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 Hebei, China
| | - Fuyu Jin
- Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, 063210 Hebei, China
| | - Yaqian Li
- Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, 063210 Hebei, China
| | - Xue Yi
- Department of Basic Medicine, Fujian Collaborative Innovation Center for Accurate Medicine of Respiratory Diseases, Xiamen Medical College, Xiamen, 361023 Fujian, China
| | - Heliang Liu
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 Hebei, China
| | - Hong Xu
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 Hebei, China.
| | - Fang Yang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 Hebei, China.
| |
Collapse
|
11
|
Zhu Y, Yao J, Duan Y, Xu H, Cheng Q, Gao X, Li S, Yang F, Liu H, Yuan J. Protein Expression Profile in Rat Silicosis Model Reveals Upregulation of PTPN2 and Its Inhibitory Effect on Epithelial-Mesenchymal Transition by Dephosphorylation of STAT3. Int J Mol Sci 2020; 21:ijms21041189. [PMID: 32054021 PMCID: PMC7072761 DOI: 10.3390/ijms21041189] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/07/2020] [Accepted: 02/09/2020] [Indexed: 12/16/2022] Open
Abstract
Silicosis is a chronic occupational lung disease caused by long-term inhalation of crystalline silica particulates. We created a rat model that closely approximates the exposure and development of silicosis in humans. Isobaric tags for relative and absolute quantitation (iTRAQ) technologies we used to identify proteins differentially expressed in activated rat lung tissue. We constructed three lentiviral knockdown vectors and an overexpression vector for the protein tyrosine phosphatase non-receptor type 2 (PTPN2) gene to achieve stable long-term expression. A total of 471 proteins were differentially expressed in the silicosis group compared with controls. Twenty upregulated, and eight downregulated proteins exhibited a ≥1.5-fold change relative to controls. We next found that the PTPN2, Factor B, and VRK1 concentrations in silicotic rats silicosis and SiO2-stimulated MLE-12 cells were significantly higher than control groups. More importantly, we found that overexpression of PTPN2 simultaneously decreased the expression of phospho–signal transducer and activator of transcription 3 (p-STAT3) and Vimentin, while increasing E-cadherin expression. The opposite pattern was observed for PTPN2-gene silencing. We identified three proteins with substantially enhanced expression in silicosis. Our study also showed that PTPN2 can inhibit epithelial-mesenchymal transition by dephosphorylating STAT3 in silicosis fibrosis.
Collapse
|
12
|
Inhibitory effects of astragaloside IV on silica-induced pulmonary fibrosis via inactivating TGF-β1/Smad3 signaling. Biomed Pharmacother 2019; 119:109387. [PMID: 31487583 DOI: 10.1016/j.biopha.2019.109387] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/16/2019] [Accepted: 08/22/2019] [Indexed: 11/22/2022] Open
Abstract
PURPOSE To observe the effect of astragaloside ASV (ASV) on silicosis fibroblasts, and further investigate its regulatory mechanism on TGF-β1/Smad3 signaling pathway. METHODS Silica-induced rats model was established in this study. RT-qPCR was performed to detect α-SMA, Collagen I, Collagen III, Smad2, Smad3 and Smad7 expression. Immunofluorescence was conducted to detect α-SMA, Collagen I, Collagen III and p-Smad3 protein and the nucleoplasmic distribution of p-Smad3.Western-blotting was performed to detect the protein of Smad2, p-Smad2, Smad3, p-Smad3 and Smad7. RESULTS 20 μg/mL ASV could effectively reduce the expression of α-SMA, Collagen I, Collagen III. TGF-β1 stimulated the proliferation of fibroblasts, promoted phosphorylation of Smad2 and Smad3, and down-regulated Smad7 expression. Among them, continuous phosphorylation of Smad3 is a major factor in causing fibrosis. Besides, ASV can inhibit silica-induced lung fibroblast fibrosis through TGF-β1/Smad3 signaling pathway, thereby inhibiting the formation of silicosis. CONCLUSION ASV could inhibit the expression of collagen in fibroblasts and the transformation to myofibroblasts, and has an anti-silicosis fibrosis effect, which may be related to the continuous phosphorylation of Smad3 in the TGF-β1/Smad signaling pathway.
Collapse
|