1
|
Qian R, Xu Y, Zhang L, Wang L, Chen X, Wang M, Bao Q, Yao Y, Xie L. Haizao Yuhu decoction ameliorates silica-induced lung injury by inhibiting transforming growth factor-beta1/Smad pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119669. [PMID: 40122314 DOI: 10.1016/j.jep.2025.119669] [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: 01/21/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Haizao Yuhu Decoction (HYD) is a traditional Chinese herbal formula known for regulating Qi and dispersing stasis. AIM OF THE STUDY This study investigates the effects of HYD on silica-induced lung injury and the underlying mechanisms. MATERIALS AND METHODS The main constituents of HYD were identified using ultra-performance liquid chromatography Q-Exactive mass spectrometry (UPLC-QE-MS). Network pharmacology was employed to predict the targets and pathways through which HYD ameliorates silicosis, which were validated in a silica-induced lung injury mouse model and a TGF-β1-induced alveolar epithelial cell model. Pathological evaluation was conducted using hematoxylin-eosin (H&E) and Masson staining, while inflammatory cytokines and fibrosis were assessed via enzyme-linked immunosorbent assay (ELISA) and hydroxyproline quantification. Western blotting (WB) was performed to analyse protein expression levels of targeted markers. Proliferation and migration capabilities of MLE12 cells treated with HYD and its bioactive constituents (glycitein, diosmetin, and limonin) were assessed using cell counting kit-8 (CCK-8) and wound healing assays. RESULTS HYD significantly alleviated silica-induced lung injury, reducing inflammatory response and collagen deposition. A total of 176 constituents were identified in HYD, with 111 being pharmacologically active and linked to 1397 potential therapeutic targets, 107 associated with silicosis. Enrichment analyses highlighted the TGF-β1/Smad pathway and epithelial-mesenchymal transition (EMT) in HYD's anti-silicosis effects, which was validated by the restoration of TGF-β1, p-Smad2/Smad2, p-Smad3/Smad3, E-cadherin, and Vimentin following HYD treatment. Additionally, glycitein, diosmetin, and limonin inhibited the proliferation and migration of TGF-β1-induced MLE12 cells and suppressed the activation of TGF-β1/Smad pathway and EMT. CONCLUSIONS HYD effectively alleviates silica-induced lung injury by specifically inhibiting the TGF-β1/Smad pathway and EMT process.
Collapse
Affiliation(s)
- Rui Qian
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Yunyi Xu
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Luoning Zhang
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Liqun Wang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Xuxi Chen
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Mengzhu Wang
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Qixue Bao
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Yuqin Yao
- State Key Laboratory of Biotherapy, West China School of Clinical Medicine (West China Hospital) Sichuan University, Chengdu, China; Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Linshen Xie
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Liu Y, Zhang J, Hu Y, Liu Z, Yang Z, Jiao R, Liu X, Li X, Sang F. BI 1015550 Improves Silica-Induced Silicosis and LPS-Induced Acute Lung Injury in Mice. Molecules 2025; 30:1311. [PMID: 40142089 PMCID: PMC11946787 DOI: 10.3390/molecules30061311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Silicosis is an interstitial lung disease (ILD) caused by prolonged inhalation of silica particles. Acute lung injury (ALI) is a critical clinical syndrome involving bilateral lung infiltration and acute hypoxic respiratory failure. However, there is currently no effective treatment for these two diseases. Previous research has established that cyclic adenosine monophosphate (cAMP) is pivotal in the pathogenesis of silicosis and acute lung injury. Phosphodiesterase 4 (PDE4) is a hydrolase enzyme of cAMP, and BI 1015550, as an inhibitor of PDE4B, is expected to be a candidate drug for treating both. BI 1015550 has shown certain anti-inflammatory and anti-fibrotic properties in systemic sclerosis-associated interstitial lung disease (SSc-ILD) and idiopathic pulmonary fibrosis (IPF), but there is a lack of research on silicosis and acute lung injury. In this research, we successfully synthesized BI 1015550 autonomously and demonstrated that it could significantly improve lung fibrosis and inflammation in a silica-induced silicosis mouse model. Furthermore, we found that BI 1015550 could also alleviate lung inflammation in a Lipopolysaccharide (LPS)-induced acute lung injury mouse model. The mechanism of action may involve the regulation of cAMP-related signaling pathways.
Collapse
Affiliation(s)
- Yuming Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
| | - Jing Zhang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China;
| | - Yayue Hu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
| | - Zhigang Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
| | - Zhongyi Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
| | - Ran Jiao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Xueze Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China; (Y.L.); (Y.H.); (Z.L.); (Z.Y.); (R.J.); (X.L.)
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Feng Sang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China;
| |
Collapse
|
3
|
Bai L, Wang J, Wang X, Wang J, Zeng W, Pang J, Zhang T, Li S, Song M, Shi Y, Wang J, Wang C. Combined therapy with pirfenidone and nintedanib counteracts fibrotic silicosis in mice. Br J Pharmacol 2025; 182:1143-1163. [PMID: 39546810 DOI: 10.1111/bph.17390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 09/19/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND AND PURPOSE Pneumoconiosis, especially silicosis, is a prevalent occupational disease with substantial global economic implications and lacks a definitive cure. Both pneumoconiosis and idiopathic pulmonary fibrosis (IPF) are interstitial lung diseases, which share many common physiological characteristics. Because pirfenidone and nintedanib are approved to treat IPF, their potential efficacy as antifibrotic agents in advanced silicosis deserves further exploration. Thus, we aimed to evaluate the individual and combined effects of pirfenidone and nintedanib in treating advanced silicosis mice and elucidate the underlying mechanisms of their therapeutic actions via multiomics. EXPERIMENTAL APPROACH We administered monotherapy or combined therapy of pirfenidone and nintedanib, with low and high doses, in silicosis established after 6 weeks and evaluated lung function, inflammatory responses and fibrotic status. Additionally, we employed transcriptomic and metabolomic analyses to uncover the mechanisms underlying different therapeutic strategies. KEY RESULTS Both pirfenidone and nintedanib were effective in treating advanced silicosis, with superior outcomes observed in combination therapy. Transcriptomic and metabolomic analyses revealed that pirfenidone and nintedanib primarily exerted their therapeutic effects by modulating immune responses, signalling cascades and metabolic processes involving lipids, nucleotides and carbohydrates. Furthermore, we experimentally validated both monotherapy and combined therapy yielded therapeutic benefits through two common signalling pathways: steroid biosynthesis and purine metabolism. CONCLUSION AND IMPLICATIONS In conclusion, pirfenidone and nintedanib, either individually or in combination, demonstrate substantial potential in advanced silicosis. Furthermore, combined therapy outperformed monotherapy, even at low doses. These therapeutic benefits are attributed to their influence on diverse signalling pathways and metabolic processes.
Collapse
Affiliation(s)
- Lu Bai
- 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
| | - Jiaxin 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
| | - Xue Wang
- Internal Medicine, Harbin Medical University, Harbin, China
- Department of Respiratory, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jixin Wang
- School of Medicine, Tsinghua University, Beijing, China
| | - Wei Zeng
- 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
| | - 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
| | - Shengxi Li
- 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
| | - 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
| | - Yiwei Shi
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, 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
| | - Chen 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
|
4
|
Zhao M, Chen Q, Chen X, Gong S, Wang M, Zhao S, Wang S, Du W, Xu Y, Peng L, Yao Y. Tripartite motif-containing 32 regulated by miR-6236-p5 inhibited silica-induced apoptosis of alveolar macrophages. Toxicology 2025; 511:154042. [PMID: 39742911 DOI: 10.1016/j.tox.2024.154042] [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: 10/21/2024] [Revised: 12/19/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Apoptosis of alveolar macrophages (AMs) induced by silica is one of the crucial driving factors of silicosis inflammation and fibrosis. However, the mechanism of silica-induced AMs apoptosis remains unclear. In this study, transcriptome sequencing identified 11 differentially expressed (DE)-mRNAs enriched in the regulation of apoptotic signaling pathways in AMs treated with 250 μg/mL silica for 24 h, of which tripartite motif-containing 32 (Trim32) was the most significant and down-regulated. The decreased Trim32 promoted AMs apoptosis, while Trim32 overexpression inhibited the apoptosis of AMs induced by silica at 250 μg/mL for 24 h. MiR-6236-p5 was then identified by MiRNA sequencing as the most significant DE-miRNA potentially regulating Trim32 expression, and the interaction between miR-6236-p5 and Trim32 3'-UTR was confirmed by dual luciferase reporter gene assay. Treated with 100 nM miR-6236-p5 inhibitor increased the expression of Trim32 and inhibited the apoptosis of AMs induced by silica at 250 μg/mL for 24 h, while miR-6236-p5 mimic promoted the apoptosis of silica-induced AMs. In conclusion, this study identified Trim32 regulated by miR-6236-p5 played an important role in silica-induced AMs apoptosis based on RNA sequencing, which provided a novel clue for exploring the mechanism of silica-induced AMs apoptosis.
Collapse
Affiliation(s)
- Manyu Zhao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China; Laboratory of Precision Therapeutics, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qing Chen
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China
| | - Xuxi Chen
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China
| | - Shuyu Gong
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China
| | - Mengzhu Wang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China
| | - Shanshan Zhao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China
| | - Sihan Wang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China
| | - Wen Du
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center for Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yunyi Xu
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China
| | - Lijun Peng
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center for Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China.
| | - Yuqin Yao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University Chengdu, 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center for Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
5
|
Yuan H, He Y, Zhang Y, Min H, Chen J, Li C. Crystalline silica-induced endoplasmic reticulum stress promotes the pathogenesis of silicosis by augmenting proinflammatory interstitial pulmonary macrophages. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174299. [PMID: 38936737 DOI: 10.1016/j.scitotenv.2024.174299] [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: 03/20/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Crystalline silica (CS) particles are ubiquitously present in the environment, particularly in occupational settings, and exposure to respirable CS causes silicosis, imposing a significant disease burden. However, the pathogenesis of silicosis remains unclear. Exposure to external stimuli, such as CS, leads to the accumulation of unfolded proteins and triggers endoplasmic reticulum (ER) stress, disrupting tissue immune homeostasis and accelerating pathological progression. While pulmonary macrophages phagocytose CS particles to initiate the immune response, the role of ER stress in this process is unknown. Herein, we used a murine model of silicosis to simulate the pathological progression from acute inflammation to fibrosis in silicosis and conducted in vivo pharmacological inhibition of ER stress to explore the underlying mechanism. Using flow cytometry, we further classified pulmonary macrophages into monocyte-like macrophages (monocytes), interstitial macrophages (IMs), and alveolar macrophages (AMs). Our results showed that CS-induced ER stress primarily contributed to the augmentation of IMs and thereby exerted a significant impact on pulmonary macrophages. Despite coexpressing M1- and M2-like markers, IMs predominantly exhibited an M1-like polarization state and played a proinflammatory role by expressing the cytokines pro-IL-1β and TNF-α during the pathological progression of silicosis. Additionally, IMs recruited by CS-induced ER stress also exhibited high expression of MHCII and exerted active immunomodulatory effects. Overall, our study demonstrates that ER stress induced by CS particles triggers a proinflammatory immune microenvironment dominated by IMs and reveals novel insights into the pulmonary toxicological effects of CS particles.
Collapse
Affiliation(s)
- Haoyang Yuan
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China
| | - Yangyang He
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China
| | - Yuting Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China
| | - Jie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China.
| | - Chao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, China Medical University, Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China.
| |
Collapse
|
6
|
Cai F, Xue S, Zhou Z, Zhang X, Kang Y, Zhang J, Zhang M. Exposure to coal dust exacerbates cognitive impairment by activating the IL6/ERK1/2/SP1 signaling pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174202. [PMID: 38925396 DOI: 10.1016/j.scitotenv.2024.174202] [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: 02/27/2024] [Revised: 06/06/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Coal dust (CD) is a common pollutant, and epidemiological surveys indicate that long-term exposure to coal dust not only leads to the occurrence of pulmonary diseases but also has certain impacts on cognitive abilities. However, there is little open-published literature on the effects and specific mechanisms of coal dust exposure on the cognition of patients with Mild Cognitive Impairment (MCI) and Alzheimer's Disease (AD). An animal model has been built in this study with clinical population samples to explore the changes in neuroinflammation and cognitive abilities with coal dust exposure. In the animal model, compared to C57BL/6 mice, APP/PS1 mice exposed to coal dust exhibited more severe cognitive impairment, accompanied by significantly elevated levels of neuroinflammatory factors Apolipoprotein E4 (AOPE4) and Interleukin-6 (IL6) in the hippocampus, and more severe neuronal damage. In clinical sample sequencing, it was found that there is significant upregulation of AOPE4, neutrophils, and IL6 expression in the peripheral blood of MCI patients compared to normal individuals. Mechanistically, cell experiments revealed that IL6 could promote the phosphorylation of ERK1/2 and enhance the expression of transcription factor SP1, thereby promoting AOPE4 expression. The results of this study suggest that coal dust can promote the upregulation of IL6 and AOPE4 in patients, exacerbating cognitive impairment.
Collapse
Affiliation(s)
- Fulin Cai
- The First Affiliated Hospital, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui University of Science and Technology, Huainan 232001, China
| | - Sheng Xue
- Anhui University of Science and Technology, Huainan 232001, China.
| | - Zan Zhou
- Department of Physiology, Shihezi University Medical College, Xinjiang, Shihezi 832000, China
| | - Xin Zhang
- Department of Blood Transfusion, The People's Hospital of Rizhao, Shandong, Rizhao 276800, China
| | - Yingjie Kang
- Department of Physiology, Shihezi University Medical College, Xinjiang, Shihezi 832000, China
| | - Jing Zhang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, Hangzhou 310000, China
| | - Mei Zhang
- The First Affiliated Hospital, Anhui University of Science and Technology, Huainan, Anhui, China
| |
Collapse
|
7
|
Chang M, Li N, Zhou Q, Yan Y, Xu W, Zhao Y, Yao S. The inhibition of MARCO by PolyG alleviates pulmonary fibrosis via regulating mitochondrial function in a silicotic rat model. ENVIRONMENTAL TOXICOLOGY 2024; 39:3808-3819. [PMID: 38523403 DOI: 10.1002/tox.24241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
Silicon dioxide (SiO2)-induced pulmonary fibrosis is potentially associated with the impairment of mitochondrial function. Previous research found that inhibition of macrophage receptor with collagenous structure (MARCO) could alleviate particle-induced lung injury by regulating phagocytosis and mitigating mitochondrial damage. The present study aims to explore the underlying anti-fibrosis mechanism of polyguanylic acid (PolyG, MARCO inhibitor) in a silicotic rat model. Hematoxylin and eosin and Masson staining were performed to visualize lung tissue pathological changes. Confocal microscopy, transmission electron microscope, western blot analysis, quantitative real-time PCR (qPCR), and adenosine triphosphate (ATP) content assay were performed to evaluate collagen content, mitochondrial function, and morphology changes in SiO2-induced rat pulmonary fibrosis. The results suggested that SiO2 exposure contributed to reactive oxygen species aggregation and the reduction of respiratory complexes and ATP synthesis. PolyG treatment could effectively reduce MARCO expression and ameliorate lung injury and fibrosis by rectifying the imbalance of mitochondrial respiration and energy synthesis. Furthermore, PolyG could maintain mitochondrial homeostasis by promoting peroxisome proliferator-activated receptor-coactivator 1 α (PGC1α)-mediated mitochondrial biogenesis and regulating fusion and fission. Together, PolyG could ameliorate SiO2-induced pulmonary fibrosis via inhibiting MARCO to protect mitochondrial function.
Collapse
Affiliation(s)
- Meiyu Chang
- School of Public Health, North China University of Science and Technology, Tangshan, China
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Ning Li
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Qiang Zhou
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Yaomin Yan
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Weiwei Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Yingzheng Zhao
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Sanqiao Yao
- School of Public Health, North China University of Science and Technology, Tangshan, China
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
8
|
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
|
9
|
Hu X, Chen C, Gao Q, Zhou L, Shao Y, Li G, Song H, Liu Q, Han L, Zhu L. Latent tuberculosis infection and infection-associated risk factors for miner workers with silicosis in eastern China. BMC Pulm Med 2024; 24:177. [PMID: 38622607 PMCID: PMC11017624 DOI: 10.1186/s12890-024-02985-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/28/2024] [Indexed: 04/17/2024] Open
Abstract
OBJECTIVES Silicosis people are at high risk of developing pulmonary tuberculosis. Whether silica exposure increases the likelihood of latent tuberculosis infection (LTBI) was not well understood, and potential factors involved in LTBI risk among silicosis people were not evaluated before. Thus, LTBI among silicosis people and potential risk factors for LTBI among silicosis people were evaluated in this study. METHODS A cross-sectional study was undertaken for 130 miner workers with silicosis. The QFT-GIT was performed for LTBI detection. RESULTS The LTBI was high to 31.6% (36/114) for silicosis participants, and 13.1% (13/99) had a history of tuberculosis. Drinking was associated with LTBI risk (OR = 6.92, 95%CI, 1.47-32.66, P = 0.015). Meanwhile, tunneling work was associated with an increased risk of LTBI compared with other mining occupations (OR = 3.91,95%CI,1.20-12.70, P = 0.024). CONCLUSIONS The LTBI rate of silicosis participants was high and more than 10% had a history of tuberculosis. Drinking alcohol and tunneling were independent risk factors for LTBI in silicosis participants.
Collapse
Affiliation(s)
- Xinsong Hu
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - Cheng Chen
- Department of Chronic Communicable Disease, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, China
| | - Qianqian Gao
- Department of Occupational Disease Prevention, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, China
| | - Lang Zhou
- Department of Occupational Disease Prevention, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, China
| | - Yan Shao
- Department of Chronic Communicable Disease, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, China
| | - Guoli Li
- Department of Chronic Communicable Disease, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, China
| | - Honghuan Song
- Department of Chronic Communicable Disease, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, China
| | - Qiao Liu
- Department of Chronic Communicable Disease, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, China
| | - Lei Han
- Department of Occupational Disease Prevention, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, China.
| | - Limei Zhu
- School of Public Health, Nanjing Medical University, Nanjing, China.
- Department of Chronic Communicable Disease, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, China.
| |
Collapse
|
10
|
Liu T, Su X, Kong X, Dong H, Wei Y, Wang Y, Wang C. Whole transcriptome sequencing identifies key lncRNAs,circRNAs, and mRNAs for exploring the pathogenesis and therapeutic target of mouse pneumoconiosis. Gene 2024; 901:148169. [PMID: 38242381 DOI: 10.1016/j.gene.2024.148169] [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: 10/22/2023] [Revised: 12/17/2023] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
BACKGROUND Pneumoconiosis is a kind of lung dysfunction caused by the inhalation of mineral dust. However, the potential molecular mechanism of pneumoconiosis have not been fully elucidated. METHODS In this study, the silica-treated pneumoconiosis mice model was constructed and the transcriptome sequencing data including lncRNA, circRNA, and mRNA were obtained. Firstly, differentially expressed lncRNA, circRNA, and mRNA (DElncRNA, DEcircRNA, DEGs) between control and pneumoconiosis/silicosis samples were screened, the target miRNAs (co-pre-miRNAs) were obtained by intersecting the miRNAs predicted by DElncRNA and DEcircRNA, respectively, and the target mRNAs (co-mRNA) were obtained by intersecting the mRNAs predicted by target miRNA and DEGs. Then, the lncRNA/circRNA-miRNA-mRNA networks were constructed by Cytoscape. Next, the key mRNAs were obtained by protein-protein interaction (PPI) analysis, and the key lncRNAs/circRNAs were selected by correlation analysis. Moreover, the expression of the key lncRNAs, circRNAs and mRNAs on chromosome were studied by the "circlize" package. Furthermore, the TFs-miRNA-mRNA network was constructed and the function of DEGs were explored by Ingenuity Pathway Analysis (IPA). To demonstrate the feasibility and value of the constructed ceRNA networks, we validated key genes and mmu-miR-682 pathway. Finally, We used the Drug-Gene Interaction database to predict potential drugs that could interfere with key genes,which may help to find promising treatment. RESULTS There were 427 DElncRNAs, 107 DEcircRNAs and 1,597 DEGs between silicosis and control groups. Totals of 77 co-pre-miRNAs and 96 co-mRNA were screened, and the lncRNA/circRNA-miRNA-mRNA networks were constructed with 27 lncRNA/25 circRNAs, 74 miRNAs and 96 mRNAs. Then, 6 key mRNAs including Igf1, Klf4, Ptgs2, Epas1, Gnao1, and Il1a were obtained by PPI, and all of these key mRNAs and 10 key lncRNAs and 8 circRNAs were significantly different between the pneumoconiosis and normal groups, in which 10 lncRNAs and 9 circRNA that have not been previously studied in pneumoconiosis/silicosis can be used as new potential therapeutic targets. Moreover, the TFs-miRNA-mRNA network were constructed with 11 TFs, 1 key miRNA (mmu-miR-682) and 3 key mRNAs (Igf1, Epas1, Ptgs2). And the validation of key genes revealing by RNA-seq through experimental approaches shows the the predictive power of this study. Finally, IPA results indicated that 41 pathways were activated and 2 pathways were suppressed in pneumoconiosis/silicosis groups, and Pathogen Induced Cytokine Storm Signaling Pathway was the most significant pathway affected by pneumoconiosis/silicosis. In addition, 93 drugs were screened out by Drug-Gene Interaction database. Among them, Hydroxychloroquine was a kind of drug which associated with Il1a and Ptgs2, may be a promising treatment. CONCLUSION This study constructed the lncRNA/circRNA-miRNA-mRNA and TFs-miRNA-mRNA networks, which could deepen the potential molecular regulatory mechanism of pneumoconiosis/silicosis.
Collapse
Affiliation(s)
- Ting Liu
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xuesen Su
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaomei Kong
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hantian Dong
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yangyang Wei
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yan Wang
- Medical School of Shanxi Datong University, Datong, Shanxi Province, China
| | - Chen Wang
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
11
|
Zhao M, Wang M, Chen X, Gao Y, Chen Q, Wang L, Bao Q, Sun D, Du W, Xu Y, Xie L, Jiang X, Zhang L, Peng L, Zhang B, Yao Y. Targeting progranulin alleviated silica particles-induced pulmonary inflammation and fibrosis via decreasing Il-6 and Tgf-β1/Smad. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133199. [PMID: 38103296 DOI: 10.1016/j.jhazmat.2023.133199] [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: 07/21/2023] [Revised: 10/12/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Long term exposure to silica particles leads to various diseases, among which silicosis is of great concern. Silicosis is an interstitial lung disease caused by inhalation of silica particles in production environments. However, the mechanisms underlying silicosis remains unclear. Our previous studies revealed that progranulin (Pgrn) promoted the expression of pro-inflammatory factors in alveolar macrophages treated with silica particles and the secretion of extracellular matrix of pulmonary fibroblasts. Nevertheless, the role of Pgrn in silica particles-induced silicosis in vivo was unknown. This study found that silica particles increased Pgrn expression in silicosis patients. Pgrn deficiency reduced lung inflammation and fibrosis in silica particles-induced silicosis mouse models. Subsequently, based on transcriptional sequencing and interleukin (Il) -6 knockout mouse models, results demonstrated that Pgrn deficiency might decrease silicosis inflammation by reducing the production of Il-6, thereby modulating pulmonary fibrosis in the early stage of silicosis mouse models. Furthermore, another mechanism through which Pgrn deficiency reduced fibrosis in silicosis mouse models was the regulation of the transforming growth factor (Tgf) -β1/Smad signaling pathway. Conclusively, Pgrn contributed to silicosis inflammation and fibrosis induced by silica particles, indicating that Pgrn could be a promising therapeutic target.
Collapse
Affiliation(s)
- Manyu Zhao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Mengzhu Wang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xuxi Chen
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Gao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Qing Chen
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Liqun Wang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Qixue Bao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Donglei Sun
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Wen Du
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yunyi Xu
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Linshen Xie
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xia Jiang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Zhang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Lijun Peng
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Ben Zhang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Departments of Cardiology, Neurology, and Oncology, Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou 570311, China.
| | - Yuqin Yao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
12
|
Li J, Deng B, Zhang J, Zhang X, Cheng L, Li G, Su P, Miao X, Yang W, Xie J, Wang R. The Peptide DH α-(4-pentenyl)-ANPQIR-NH 2 Exhibits Antifibrotic Activity in Multiple Pulmonary Fibrosis Models Induced by Particulate and Soluble Chemical Fibrogenic Agents. J Pharmacol Exp Ther 2024; 388:701-714. [PMID: 38129127 DOI: 10.1124/jpet.123.001849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 12/23/2023] Open
Abstract
Interstitial lung diseases (ILDs) are a group of restrictive lung diseases characterized by interstitial inflammation and pulmonary fibrosis. The incidence of ILDs associated with exposure to multiple hazards such as inhaled particles, fibers, and ingested soluble chemicals is increasing yearly, and there are no ideal drugs currently available. Our previous research showed that the novel and low-toxicity peptide DHα-(4-pentenyl)-ANPQIR-NH2 (DR3penA) had a strong antifibrotic effect on a bleomycin-induced murine model. Based on the druggability of DR3penA, we sought to investigate its effects on respirable particulate silicon dioxide (SiO2)- and soluble chemical paraquat (PQ)-induced pulmonary fibrosis in this study by using western blot, quantitative reverse-transcription polymerase chain reaction (RT-qPCR), immunofluorescence, H&E and Masson staining, immunohistochemistry, and serum biochemical assays. The results showed that DR3penA alleviated the extent of fibrosis by inhibiting the expression of fibronectin and collagen I and suppressed oxidative stress and epithelial-mesenchymal transition (EMT) in vitro and in vivo. Further study revealed that DR3penA may mitigate pulmonary fibrosis by negatively regulating the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway and mitogen-activated protein kinase (MAPK) pathway. Unexpectedly, through the conversion of drug bioavailability under different routes of administration, DR3penA exerted antifibrotic effects equivalent to those of the positive control drug pirfenidone (PFD) at lower doses. In summary, DR3penA may be a promising lead compound for various fibrotic ILDs. SIGNIFICANCE STATEMENT: Our study verified that DHα-(4-pentenyl)-ANPQIR-NH2 (DR3penA) exhibited positive antifibrotic activity in pulmonary fibrosis induced by silicon dioxide (SiO2) particles and soluble chemical paraquat (PQ) and demonstrated a low-dose advantage compared to the small-molecule drug pirfenidone (PFD). The peptide DR3penA can be further developed for the treatment of multiple fibrotic lung diseases.
Collapse
Affiliation(s)
- Jieru Li
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Bochuan Deng
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Jiao Zhang
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Xiang Zhang
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Lu Cheng
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Guofeng Li
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Ping Su
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Xiaokang Miao
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Wenle Yang
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Junqiu Xie
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Rui Wang
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (J.L., R.W.); Department of General Surgery, The Second Hospital and Clinical Medical School (J.L.) and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066 (B.D., J.Z., X.Z., P.S., X.M., W.Y., J.X., R.W.), Lanzhou University, Lanzhou, China; and School of Biomedical Engineering (L.C.) and School of Pharmaceutical Sciences (G.L.), Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| |
Collapse
|
13
|
Lu Y, Mu M, RenChen X, Wang W, Zhu Y, Zhong M, Jiang Y, Tao X. 2-Deoxy-D-glucose ameliorates inflammation and fibrosis in a silicosis mouse model by inhibiting hypoxia-inducible factor-1α in alveolar macrophages. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115767. [PMID: 38039851 DOI: 10.1016/j.ecoenv.2023.115767] [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: 06/17/2023] [Revised: 11/12/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023]
Abstract
Inhaling silica causes the occupational illness silicosis, which mostly results in the gradual fibrosis of lung tissue. Previous research has demonstrated that hypoxia-inducible factor-1α (HIF-1α) and glycolysis-related genes are up-regulated in silicosis. The role of 2-deoxy-D-glucose (2-DG) as an inhibitor of glycolysis in silicosis mouse models and its molecular mechanisms remain unclear. Therefore, we used 2-DG to observe its effect on pulmonary inflammation and fibrosis in a silicosis mouse model. Furthermore, in vitro cell experiments were conducted to explore the specific mechanisms of HIF-1α. Our study found that 2-DG down-regulated HIF-1α levels in alveolar macrophages induced by silica exposure and reduced the interleukin-1β (IL-1β) level in pulmonary inflammation. Additionally, 2-DG reduced silica-induced pulmonary fibrosis. From these findings, we hypothesize that 2-DG reduced glucose transporter 1 (GLUT1) expression by inhibiting glycolysis, which inhibits the expression of HIF-1α and ultimately reduces transcription of the inflammatory cytokine, IL-1β, thus alleviating lung damage. Therefore, we elucidated the important regulatory role of HIF-1α in an experimental silicosis model and the potential defense mechanisms of 2-DG. These results provide a possible effective strategy for 2-DG in the treatment of silicosis.
Collapse
Affiliation(s)
- Yuting Lu
- School of Public Health, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, China
| | - Min Mu
- School of Public Health, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, China; Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, China; Joint Research Center of Occupational Medicine and Health, Institute of Grand Health, Hefei Comprehensive National Science Center, Anhui University of Science and Technology, China.
| | - Xiaotian RenChen
- School of Public Health, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, China
| | - Wenyang Wang
- School of Public Health, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, China; Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, China
| | - Yingrui Zhu
- School of Public Health, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, China
| | - Meiping Zhong
- School of Public Health, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, China
| | - Yuerong Jiang
- School of Public Health, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, China
| | - Xinrong Tao
- School of Public Health, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, China; Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, China; Joint Research Center of Occupational Medicine and Health, Institute of Grand Health, Hefei Comprehensive National Science Center, Anhui University of Science and Technology, China
| |
Collapse
|
14
|
Wang D, Deng B, Cheng L, Li J, Guo X, Zhang J, Zhang X, Su P, Li G, Miao X, Yang W, Xie J, Wang R. The novel peptide DR4penA attenuates the bleomycin- and paraquat-induced pulmonary fibrosis by suppressing the TGF-β/Smad signaling pathway. FASEB J 2023; 37:e23225. [PMID: 37855708 DOI: 10.1096/fj.202301363r] [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: 07/07/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 10/20/2023]
Abstract
Pulmonary fibrosis (PF), which is caused by continuous alveolar epithelial cell injury and abnormal repair, is referred to as a difficult disease of the lung system by the World Health Organization due to its rapid progression, poor prognosis, and high mortality rate. However, there is still a lack of ideal therapeutic strategies. The peptide DR8 (DHNNPQIR-NH2 ), which is derived from rapeseed, exerted antifibrotic activity in the lung, liver, and kidney in our previous studies. By studying the structure-activity relationship and rational design, we introduced an unnatural hydrophobic amino acid (α-(4-pentenyl)-Ala) into DR8 and screened the novel peptide DR4penA (DHNα-(4-pentenyl)-APQIR-NH2 ), which had higher anti-PF activity, higher antioxidant activity and a longer half-life than DR8. Notably, DR4penA attenuated bleomycin- and paraquat-induced PF, and the anti-PF activity of DR4penA was equivalent to that of pirfenidone. Additionally, DR4penA suppressed the TGF-β/Smad pathway in TGF-β1-induced A549 cells and paraquat-induced rats. This study demonstrates that the novel peptide DR4penA is a potential candidate compound for PF therapy, and its antifibrotic activity in different preclinical models of PF provides a theoretical basis for further study.
Collapse
Affiliation(s)
- Dan Wang
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College, Nanchong, China
| | - Bochuan Deng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Lu Cheng
- School of Biomedical Engineering, Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Jieru Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaomin Guo
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jiao Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiang Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Ping Su
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Guofeng Li
- School of Pharmaceutical Sciences, Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China
| | - Xiaokang Miao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Wenle Yang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Junqiu Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Rui Wang
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
15
|
Pu S, Yang Z, Zhang X, Li M, Han N, Yang X, He J, Yu G, Meng X, Jia Q, Shao H. Fermented cordyceps powder alleviates silica-induced pulmonary inflammation and fibrosis in rats by regulating the Th immune response. Chin Med 2023; 18:131. [PMID: 37828528 PMCID: PMC10571334 DOI: 10.1186/s13020-023-00823-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/20/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Silicosis is an important occupational disease caused by inhalation of free silica and is characterized by persistent pulmonary inflammation, subsequent fibrosis and lung dysfunction. Until now, there has been no effective treatment for the disease due to the complexity of pathogenesis. Fermented cordyceps powder (FCP) has a similar effect to natural cordyceps in tonifying the lung and kidney. It has started to be used in the adjuvant treatment of silicosis. This work aimed to verify the protective effects of FCP against silicosis, and to explore the related mechanism. METHODS Wistar rats were randomly divided into four groups including the saline-instilled group, the silica-exposed group, the silica + FCP (300 mg/kg) group and the silica + FCP (600 mg/kg) group. Silicosis rat models were constructed by intratracheal instillation of silica (50 mg). Rats in the FCP intervention groups received the corresponding dose of FCP daily by intragastric gavage. Rats were sacrificed on days 7, 28 and 56 after treatment, then samples were collected for further analysis. RESULTS FCP intervention reduced the infiltration of inflammatory cells and the concentration of interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and transforming growth factor-β1 (TGF-β1) at days 7, 28, 56, and decreased the expression of collagen, α-smooth muscle actin (α-SMA) and fibronectin (FN) at days 28 and 56 in the lung of silicosis rats. FCP also decreased the immune response of Th1 and Th17 at days 7, 28, 56 and inhibited the enhancement of the Th2 response at day 56. CONCLUSIONS FCP intervention could alleviate silica-induced pulmonary inflammation and fibrosis, the protective effect may be achieved by reducing Th1 and Th17 immune responses and inhibiting the enhancement of the Th2 response.
Collapse
Affiliation(s)
- Shuangshuang Pu
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250355, Shandong, China
- Hospital Affiliated to Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China
| | - Zhifeng Yang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Xiaofeng Zhang
- Linyi County Center for Disease Control and Prevention, Linyi County, 91 Yongxing Street, Dezhou, 251500, Shandong, China
| | - Ming Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Na Han
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250355, Shandong, China
| | - Xiaohan Yang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Jin He
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Gongchang Yu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Xiangjing Meng
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China.
| | - Hua Shao
- Shandong University of Traditional Chinese Medicine, 4655 University Road, Changqing District, Jinan, 250355, Shandong, China.
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, 18877 Jingshi Road, Lixia District, Jinan, 250062, Shandong, China.
| |
Collapse
|
16
|
Yuan H, You Y, He Y, Wei Y, Zhang Y, Min H, Li C, Chen J. Crystalline Silica-Induced Proinflammatory Interstitial Macrophage Recruitment through Notch3 Signaling Promotes the Pathogenesis of Silicosis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:14502-14514. [PMID: 37721423 DOI: 10.1021/acs.est.3c03980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Crystalline silica (CS) particles are ubiquitous in the environment, especially in occupational conditions, and exposure to respirable CS causes silicosis. The initial response to CS is mediated by innate immunity, where pulmonary macrophages act as central orchestrators. However, the repercussions of CS on functionally distinct macrophage subsets remain to be inconclusive. Herein, to study the effects of inhaled CS, we divided macrophages into three subsets: circulating monocytes, interstitial macrophages (IMs), and alveolar macrophages (AMs). CS-induced massive IMs increase in the lung, the phenotype and function of which differed from those of tissue-resident AMs and circulating monocytes. The augmented IMs were driven by recruitment of circulating macrophages rather than cell proliferation in situ. Moreover, the IMs predominantly exerted a classic activated (M1) phenotype and expressed proinflammatory cytokines, contributing to CS-induced lung injury. Notably, we demonstrated that IMs augmented Notch3 expression. Mechanistically, using myeloid-specific Notch3-knockout mice, we demonstrated that Notch3 signaling not only promoted IMs recruitment by regulating CCR2 expression but also manipulated the proinflammatory phenotype. Mice with conditional Notch3-knockout exhibited alleviation of CS-induced inflammation and fibrosis in lung. Overall, our study identifies IMs as critical mediators in response to CS and highlights the role of Notch3 in IMs recruitment and activation, providing new insights into CS toxicological effects in the lung.
Collapse
Affiliation(s)
- Haoyang Yuan
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yichuan You
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yangyang He
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yungeng Wei
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yuting Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Chao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Jie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| |
Collapse
|
17
|
Yuan J, Mo Y, Zhang Y, Zhang Y, Zhang Q. Nickel nanoparticles induce autophagy and apoptosis via HIF-1α/mTOR signaling in human bronchial epithelial cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 329:121670. [PMID: 37080518 PMCID: PMC10231338 DOI: 10.1016/j.envpol.2023.121670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/31/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
With the rapid development of nanotechnology, the potential adverse health effects of nanoparticles have been caught more attention and become global concerns. However, the underlying mechanisms in metal nanoparticle-induced toxic effects are still largely obscure. In this study, we investigated whether exposure to nickel nanoparticles (Nano-Ni) and titanium dioxide nanoparticles (Nano-TiO2) would alter autophagy and apoptosis levels in normal human bronchial epithelial BEAS-2B cells and the underlying mechanisms involved in this process. Our results showed that the expressions of autophagy- and apoptosis-associated proteins were dysregulated in cells exposed to Nano-Ni. However, exposure to the same doses of Nano-TiO2 had no significant effects on these proteins. In addition, exposure to Nano-Ni, but not Nano-TiO2, led to nuclear accumulation of HIF-1α and decreased phosphorylation of mTOR in BEAS-2B cells. Inhibition of HIF-1α by CAY10585 abolished Nano-Ni-induced decreased phosphorylation of mTOR, while activation of mTOR by MHY1485 did not affect Nano-Ni-induced nuclear accumulation of HIF-1α. Furthermore, both HIF-1α inhibition and mTOR activation abolished Nano-Ni-induced autophagy but enhanced Nano-Ni-induced apoptosis. Blockage of autophagic flux by Bafilomycin A1 exacerbated Nano-Ni-induced apoptosis, while activation of autophagy by Rapamycin effectively rescued Nano-Ni-induced apoptosis. In conclusion, our results demonstrated that Nano-Ni exposure caused increased levels of autophagy and apoptosis via the HIF-1α/mTOR signaling axis. Nano-Ni-induced autophagy has a protective role against Nano-Ni-induced apoptosis. These findings provide us with further insight into Nano-Ni-induced toxicity.
Collapse
Affiliation(s)
- Jiali Yuan
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Yiqun Mo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Yue Zhang
- Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yuanbao Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Qunwei Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
18
|
Li K, Liu X, Hou R, Zhao H, Zhao P, Tian Y, Li J. Uncovering mechanisms of Baojin Chenfei formula treatment for silicosis by inhibiting inflammation and fibrosis based on serum pharmacochemistry and network analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 260:115082. [PMID: 37257350 DOI: 10.1016/j.ecoenv.2023.115082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/19/2023] [Accepted: 05/26/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Baojin Chenfei formula (BCF), a Chinese herbal formula, has significant effects on improving the clinical symptoms of patients with silicosis. However, its active compounds and the underlying mechanisms have not yet fully been elucidated. PURPOSE This study aimed to explore the underlying mechanisms of BCF in treating silicosis. METHODS The rat model of silicosis was developed via a single intratracheal instillation of SiO2 suspension to examine the therapeutic impacts of BCF on silicosis. Subsequently, the active compounds, targets, and mechanisms of BCF were analyzed based on serum pharmacochemistry and network analysis. Finally, the underlying mechanisms of representative compounds of BCF were validated in vitro experiments. RESULTS BCF significantly alleviated SiO2-induced silicosis in rats, evidenced by improved lung function, decreased pathological injury, and reduced inflammatory response and fibrosis. 19 active compounds were identified from the rat serum samples after BCF gavage. Subsequently, 299 targets for these 19 compounds in BCF and 257 genes related to silicosis were collected. 26 overlapping targets, including AKT1, TNF, IL6, MAPK3, EGFR, and others, were obtained from the intersection of the 299 BCF-related targets and 257 silicosis-associated genes. These overlapping targets mainly corresponded to glycyrrhetic acid and paeoniflorin and were mainly associated with positive regulation of smooth muscle cell proliferation, positive regulation of MAP kinase activity, and inflammatory response. In vitro experiments also demonstrated that the representative compounds of BCF (glycyrrhetic acid and paeoniflorin) could suppress inflammatory response by the MAPK pathway, and also inhibited fibroblast activation by the EGFR-PI3K-AKT pathway. CONCLUSION Active compounds of BCF, such as glycyrrhetic acid and paeoniflorin, could suppress inflammatory response by the MAPK pathway and suppress fibroblast activation by the EGFR-PI3K-AKT pathway. These might be the mechanisms of BCF in treating silicosis.
Collapse
Affiliation(s)
- Kangchen Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China; Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Xinguang Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Runsu Hou
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Hulei Zhao
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China; Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Peng Zhao
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Yange Tian
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China; Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China; Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China.
| |
Collapse
|
19
|
Andrade da Silva LH, Vieira JB, Cabral MR, Antunes MA, Lee D, Cruz FF, Hanes J, Rocco PRM, Morales MM, Suk JS. Development of nintedanib nanosuspension for inhaled treatment of experimental silicosis. Bioeng Transl Med 2023; 8:e10401. [PMID: 36925690 PMCID: PMC10013831 DOI: 10.1002/btm2.10401] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 11/11/2022] Open
Abstract
Silicosis is an irreversible and progressive fibrotic lung disease caused by massive inhalation of crystalline silica dust at workplaces, affecting millions of industrial workers worldwide. A tyrosine kinase inhibitor, nintedanib (NTB), has emerged as a potential silicosis treatment due to its inhibitory effects on key signaling pathways that promote silica-induced pulmonary fibrosis. However, chronic and frequent use of the oral NTB formulation clinically approved for treating other fibrotic lung diseases often results in significant side effects. To this end, we engineered a nanocrystal-based suspension formulation of NTB (NTB-NS) possessing specific physicochemical properties to enhance drug retention in the lung for localized treatment of silicosis via inhalation. Our NTB-NS formulation was prepared using a wet-milling procedure in presence of Pluronic F127 to endow the formulation with nonadhesive surface coatings to minimize interactions with therapy-inactivating delivery barriers in the lung. We found that NTB-NS, following intratracheal administration, provided robust anti-fibrotic effects and mechanical lung function recovery in a mouse model of silicosis, whereas a 100-fold greater oral NTB dose given with a triple dosing frequency failed to do so. Importantly, several key pathological phenotypes were fully normalized by NTB-NS without displaying notable local or systemic adverse effects. Overall, NTB-NS may open a new avenue for localized treatment of silicosis and potentially other fibrotic lung diseases.
Collapse
Affiliation(s)
- Luisa Helena Andrade da Silva
- Laboratory of Pulmonary InvestigationCarlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de JaneiroBrazil
- Rio de Janeiro Innovation Network in Nanosystems for Health – NanoSAÚDE/FAPERJRio de JaneiroBrazil
| | - Juliana Borges Vieira
- Laboratory of Pulmonary InvestigationCarlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de JaneiroBrazil
| | - Marianna Ribeiro Cabral
- Laboratory of Pulmonary InvestigationCarlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de JaneiroBrazil
| | - Mariana Alves Antunes
- Laboratory of Pulmonary InvestigationCarlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de JaneiroBrazil
| | - Daiheon Lee
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of OphthalmologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary InvestigationCarlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de JaneiroBrazil
| | - Justin Hanes
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of OphthalmologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary InvestigationCarlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de JaneiroBrazil
- Rio de Janeiro Innovation Network in Nanosystems for Health – NanoSAÚDE/FAPERJRio de JaneiroBrazil
| | - Marcelo Marcos Morales
- Laboratory of Cellular and Molecular PhysiologyCarlos Chagas Filho Biophysics Institute, Federal University of Rio de JaneiroRio de JaneiroBrazil
| | - Jung Soo Suk
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of OphthalmologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
20
|
Liu H, He H, Tian Y, Cui J, Wang S, Wang H. Cyclophilin A accelerates SiO 2-induced macrophage foaming. Cell Signal 2023; 103:110562. [PMID: 36535629 DOI: 10.1016/j.cellsig.2022.110562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Silicosis is a common occupational disease characterized by lung inflammation, fibrosis and pulmonary dysfunction caused by long-term inhalation of free SiO2. Cell foaming and the change of CyPA have been observed in SiO2-induced macrophages, but the specific mechanism of CyPA in SiO2-induced foam cells remains poorly understood. The purpose of this study is to explore the mechanism of CyPA in SiO2-induced macrophage foaming and its effect on silicosis. We found that overexpression of CyPA promoted the macrophage foaming and the expression of COL I and α-SMA, while silencing CyPA inhibites the macrophage foaming and the expression of COL I and α-SMA. After blocking the expression of CD36 on the basis of overexpression CyPA, we found it inhibites the macrophage foaming. In conclusion, CyPA can affect the foaming of macrophages and may participate in silicosis fibrosis.
Collapse
Affiliation(s)
- Heliang Liu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China; Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Hailan He
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Ying Tian
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Jie Cui
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Shuang Wang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Hongli Wang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China.
| |
Collapse
|
21
|
Ma R, Fan Y, Huang X, Wang J, Li S, Wang Y, Ye Q. Lipid dysregulation associated with progression of silica-induced pulmonary fibrosis. Toxicol Sci 2023; 191:296-307. [PMID: 36477571 DOI: 10.1093/toxsci/kfac124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Silicosis is an irreversible, progressive, fibrotic lung disease caused by long-term exposure to dust-containing silica particles at the workplace. Despite the precautions enforced, the rising incidence of silicosis continues to occur globally, particularly in developing countries. A better understanding of the disease progression and potential metabolic reprogramming of silicosis is warranted. The low- or high-dose silica-induced pulmonary fibrosis in mice was constructed to mimic chronic or accelerated silicosis. Silica-induced mice lung fibrosis was analyzed by histology, lung function, and computed tomography scans. Non-targeted metabolomics of the lung tissues was conducted by ultra-high-performance liquid chromatography-mass spectrometry to show the temporal metabolic trajectory. The low-dose silica-induced silicosis characterized inflammation for up to 42 days, with the onset of cellular silicon nodules. Conversely, the high-dose silica-induced silicosis characterized inflammation for up to 14 days, after which the disease developed rapidly, with a large volume of collagen deposition, presenting progressive massive fibrosis. Both low- and high silica-induced fibrosis had aberrant lipid metabolism. Combined with the RNA-Seq data, this multiomics study demonstrated alterations in the enzymes involved in sphingolipid metabolism. Time-dependent metabolic reprogramming revealing abnormal glycerophospholipid metabolism was intimately associated with the process of inflammation, whereas sphingolipid metabolism was crucial during lung fibrosis. These findings suggest that lipid dysregulation, especially sphingolipid metabolism, was involved in the process of silicosis.
Collapse
Affiliation(s)
- Ruimin Ma
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yali Fan
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xiaoxi Huang
- Medical Research Center, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jingwei Wang
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Shuang Li
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yuanying Wang
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Qiao Ye
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| |
Collapse
|
22
|
Zhang Z, Cao Z, Hou L, Song M, Zhou Y, Chen Y, Hu H, Hou Y, Liu Y, Li B, Song X, Ge W, Li B, Jiang X, Yang J, Song D, Zhang X, Pang J, Zhang T, Zhang H, Yang P, Wang J, Wang C. Adenovirus-mediated Overexpression of FcγRIIB Attenuates Pulmonary Inflammation and Fibrosis. Am J Respir Cell Mol Biol 2023; 68:213-227. [PMID: 36227848 DOI: 10.1165/rcmb.2022-0056oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Progressive fibrosing interstitial lung diseases (PF-ILDs) result in high mortality and lack effective therapies. The pathogenesis of PF-ILDs involves macrophages driving inflammation and irreversible fibrosis. Fc-γ receptors (FcγRs) regulate macrophages and inflammation, but their roles in PF-ILDs remain unclear. We characterized the expression of FcγRs and found upregulated FcγRIIB in human and mouse lungs after exposure to silica. FcγRIIB deficiency aggravated lung dysfunction, inflammation, and fibrosis in silica-exposed mice. Using single-cell transcriptomics and in vitro experiments, FcγRIIB was found in alveolar macrophages, where it regulated the expression of fibrosis-related genes Spp1 and Ctss. In mice with macrophage-specific overexpression of FcγRIIB and in mice treated with adenovirus by intratracheal instillation to upregulate FcγRIIB, silica-induced functional and histological changes were ameliorated. Our data from three genetic models and a therapeutic model suggest that FcγRIIB plays a protective role that can be enhanced by adenoviral overexpression, representing a potential therapeutic strategy for PF-ILDs.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China.,Department of Physiology and.,Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,National Health Commission Key Laboratory of Pneumoconiosis, Taiyuan, China
| | | | - Lin Hou
- Department of Physiology and
| | - Meiyue Song
- Department of Physiology and.,Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yitian Zhou
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yiling Chen
- Department of Physiology and.,Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao tong University, Xi'an, China; and
| | - Huiyuan Hu
- Department of Physiology and.,Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiao tong University, Xi'an, China; and
| | - Yangfeng Hou
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | | | - Bolun Li
- Department of Physiology and.,Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaomin Song
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Weipeng Ge
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Baicun Li
- Department of Physiology and.,Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | | | | | - Dingyun Song
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xinri Zhang
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China.,National Health Commission Key Laboratory of Pneumoconiosis, Taiyuan, China
| | - Junling Pang
- Department of Physiology and.,Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Tiantian Zhang
- Department of Physiology and.,Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | | | | | - Jing Wang
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China.,Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China.,Department of Physiology and.,National Health Commission Key Laboratory of Pneumoconiosis, Taiyuan, China.,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
23
|
Song J, Xu M, Wang T, Hao J, Li W, Lu X, Wang L, Zhang H, Kong X, Zhang X. Exosomal miRNAs contribute to coal dust particle-induced pulmonary fibrosis in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114454. [PMID: 38321673 DOI: 10.1016/j.ecoenv.2022.114454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/19/2022] [Accepted: 12/18/2022] [Indexed: 02/08/2024]
Abstract
Coal workers' pneumoconiosis (CWP) is a fatal occupational disease caused by inhalation of coal dust particles, which leads to progressive pulmonary fibrosis. Recently, as new signal carriers for intercellular communication, exosomal miRNAs have been validated in the pathogenesis of multiple diseases. However, the research on exosomal miRNAs in CWP is still in the preliminary stage. Here, using miRNA sequencing, exosomal miRNA profiles in bronchoalveolar lavage fluid (BALF) from rats with pulmonary fibrosis induced by coal dust particles were analyzed, and the underlying biological function of putative target genes was explored by GO term analysis and KEGG pathway enrichment analysis. According to the results, intratracheal instillation of coal dust particles can alter the exosomal miRNAs expression in the BALF of rats. Further bioinformatics analysis provided some clues to reveal their function in pathological process of pneumoconiosis. More importantly, we identified 4 differentially expressed exosomal miRNAs (miRNA-21-5p, miRNA-29a-3p, miRNA-26a-5p, and miRNA-34a-5p) by qRT‑PCR and further verified the temporal changes in the expression of these exosomal miRNAs in animal models from 2 weeks to 16 weeks postexposure. In addition, we conducted a preliminary study on Smad7 as a potential target of miRNA-21-5p and found that exosomal miRNA 21-5p/Smad7 may contribute to the pulmonary fibrosis induced by coal dust particles. Our study confirmed the contribution of exosomal miRNAs to coal dust particle-induced pulmonary fibrosis and provided new insights into the pathogenesis of CWP.
Collapse
Affiliation(s)
- Jing Song
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, China; National Health Commission Key Laboratory of Pneumoconiosis, Taiyuan, China.
| | - Mengtong Xu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Tiantian Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Jiarui Hao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Wenjing Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Xiaoting Lu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, China; National Health Commission Key Laboratory of Pneumoconiosis, Taiyuan, China
| | - Linping Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, China; National Health Commission Key Laboratory of Pneumoconiosis, Taiyuan, China
| | - Huifang Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, China; National Health Commission Key Laboratory of Pneumoconiosis, Taiyuan, China
| | - Xiaomei Kong
- National Health Commission Key Laboratory of Pneumoconiosis, Taiyuan, China
| | - Xinri Zhang
- National Health Commission Key Laboratory of Pneumoconiosis, Taiyuan, China
| |
Collapse
|
24
|
Wang Z, Shen L, Wang J, Huang J, Tao H, Zhou X. Prognostic analysis of m6A-related genes as potential biomarkers in idiopathic pulmonary fibrosis. Front Genet 2022; 13:1059325. [PMID: 36523766 PMCID: PMC9744785 DOI: 10.3389/fgene.2022.1059325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/07/2022] [Indexed: 10/28/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, fatal lung disease with limited treatment options. N6-methyladenosine (m6A) is a reversible RNA modification and has been implicated in various biological processes. However, there are few studies on m6A in IPF. This project mainly explores the prognostic value of m6A-related genes as potential biomarkers in IPF, in order to establish a set of accurate prognostic prediction model. In this study, we used GSE28042 dataset in GEO database to screen out 218 m6A-related candidate genes with high IPF correlation and high differential expression through differentially expressed gene analysis, WGCNA and m6A correlation analysis. The genes associated with the prognosis of IPF were screened out by univariate Cox regression analysis, LASSO analysis, and multivariate Cox regression analysis, and the multivariate Cox model of prognostic risk of related genes was constructed. We found that RBM11, RBM47, RIC3, TRAF5 and ZNF14 were key genes in our model. Finally, the prognostic prediction ability and independent prognostic characteristics of the risk model were evaluated by survival analysis and independent prognostic analysis, and verified by the GSE93606 dataset, which proved that the prognostic risk model we constructed has a strong and stable prediction efficiency.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Biochemistry and Molecular Biology, Medical College, Soochow University, Suzhou, China
| | - Lanyu Shen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Junjie Wang
- Department of Biochemistry and Molecular Biology, Medical College, Soochow University, Suzhou, China
| | - Jiaqian Huang
- Department of Biochemistry and Molecular Biology, Medical College, Soochow University, Suzhou, China
| | - Huimin Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiumin Zhou
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
25
|
Ganesan N, Ronsmans S, Hoet P. Differential immunological effects of silica nanoparticles on peripheral blood mononuclear cells of silicosis patients and controls. Front Immunol 2022; 13:1025028. [PMID: 36311760 PMCID: PMC9606771 DOI: 10.3389/fimmu.2022.1025028] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
Silicosis is a fibrotic disease caused by the inhalation of respirable silica particles, which are typically engulfed by alveolar macrophages and subsequently induce the release of inflammatory cytokines. Various animal experimental and human studies have focused on modeling silicosis, to assess the interactions of macrophages and other cell types with silica particles. There is still, however, limited knowledge on the differential response upon silica-exposure between silicosis patients and controls. We focused on studying the responsiveness of peripheral blood mononuclear cells (PBMCs) to silica nanoparticles (SiNPs) - Ludox and NM-200 - of silicosis patients and controls. The proliferative capacity of T- CD3+ and B- CD19+ cells, were evaluated via Carboxyfluorescein succinimidyl ester (CFSE) assay. The activation status of lymphocyte subsets and response to silica were also evaluated by comparing the extent of micro-granuloma or aggregate formation with the cytokine secretion profiles between both groups of individuals. The proliferative capacity of CD19+ cells was elevated in silicotic patients as opposed to controls. Subsets of regulatory T cells (CD4+ CD25+ and CD8+ CD25+) and immunoglobulins IgM and IgG were also significantly increased in patients. The number and the size of aggregates formed were higher with SiNPs stimulation in patients compared to controls. Multivariable analysis also elucidated the role of key cytokines like interleukin-1β (IL-1β), IL-6 and interferon-gamma (IFN-γ), which were upregulated in SiNP-stimulated PBMCs of patients compared to controls. Our ex vivo model thus has potential to provide insights into the immunological effects of silica particles in lymphocytes of silicosis patients and controls.
Collapse
Affiliation(s)
- Nirosha Ganesan
- Laboratory of Toxicology, Unit of Environment & Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | - Steven Ronsmans
- Laboratory of Toxicology, Unit of Environment & Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
- Clinic for Occupational and Environmental Medicine, Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Peter Hoet
- Laboratory of Toxicology, Unit of Environment & Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
- *Correspondence: Peter Hoet,
| |
Collapse
|
26
|
Chen Y, Song M, Li Z, Hou L, Zhang H, Zhang Z, Hu H, Jiang X, Yang J, Zou X, Pang J, Zhang T, Yang P, Wang J, Wang C. FcεRI deficiency alleviates silica-induced pulmonary inflammation and fibrosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114043. [PMID: 36087468 DOI: 10.1016/j.ecoenv.2022.114043] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/14/2022] [Accepted: 08/29/2022] [Indexed: 06/15/2023]
Abstract
Silicosis is one of the most important occupational diseases worldwide, caused by inhalation of silica particles or free crystalline silicon dioxide. As a disease with high mortality, it has no effective treatment and new therapeutic targets are urgently needed. Recent studies have identified FCER1A, encoding α-subunit of the immunoglobulin E (IgE) receptor FcεRI, as a candidate gene involved in the biological pathways leading to respiratory symptoms. FcεRI is known to be important in allergic asthma, but its role in silicosis remains unclear. In this study, serum IgE concentrations and FcεRI expression were assessed in pneumoconiosis patients and silica-exposed mice. The role of FcεRI was explored in a silica-induced mouse model using wild-type and FcεRI-deficient mice. The results showed that serum IgE concentrations were significantly elevated in both pneumoconiosis patients and mice exposed to silica compared with controls. The mRNA and protein expression of FcεRI were also significantly increased in the lung tissue of patients and silica-exposed mice. FcεRI deficiency significantly attenuated the changes in lung function caused by silica exposure. Silica-induced elevations of IL-1β, IL-6, and TNF-α were significantly attenuated in the lung tissue and bronchoalveolar lavage fluid (BALF) of FcεRI-deficient mice compared with wild-type controls. Additionally, FcεRI-deficient mice showed a significantly lower score of pulmonary fibrosis than wild-type mice following exposure to silica, with significantly lower hydroxyproline content and expression of fibrotic genes Col1a1 and Fn1. Immunofluorescent staining suggested FcεRI mainly on mast cells. Mast cell degranulation took place after silica exposure, as shown by increased serum histamine levels and β-hexosaminidase activity, which were significantly reduced in FcεRI-deficient mice compared with wild-type controls. Together, these data showed that FcεRI deficiency had a significant protective effect against silica-induced pulmonary inflammation and fibrosis. Our findings provide new insights into the pathophysiological mechanisms of silica-induced pulmonary fibrosis and a potential target for the treatment of silicosis.
Collapse
Affiliation(s)
- Yiling Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Meiyue Song
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Zhaoguo Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Lin Hou
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Hong Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Zhe Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China; NHC Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| | - Huiyuan Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Xuehan Jiang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Jie Yang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Xuan Zou
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Junling Pang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Tiantian Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Peiran Yang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| | - Chen Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
27
|
Tang Q, Xing C, Li M, Jia Q, Bo C, Zhang Z. Pirfenidone ameliorates pulmonary inflammation and fibrosis in a rat silicosis model by inhibiting macrophage polarization and JAK2/STAT3 signaling pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114066. [PMID: 36108436 DOI: 10.1016/j.ecoenv.2022.114066] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 06/15/2023]
Abstract
Macrophages play an important role in causing silicosis eventually becoming an irreversible fibrotic disease, and there are no specific drugs for silicosis in the clinic so far. Pirfenidone has consistently been shown to have anti-inflammatory and anti-fibrotic effects, but the specific mechanism by which it ameliorates fibrosis in silicosis is unclear. A rat silicosis model was established in this study, and lung tissues and serum were collected by batch execution at 14, 28, and 56 days. Also, the effects of Pirfenidone on macrophage polarization and pulmonary fibrosis were evaluated in silicosis with early intervention and late treatment by histological examination, Enzyme-linked immunosorbent assay, Hydroxyproline assay, Western blot and Quantitative reverse transcription polymerase chain reaction. The results showed that Pirfenidone significantly reduced pulmonary fibrosis in rats with silicosis, and both early intervention and late treatment effectively inhibited the expression of α-SMA, Col-I, Vimentin, Hydroxyproline, IL-1β, IL-18, and the M2 macrophage marker CD206 and Arg-1, while only early intervention effectively inhibited E-cad, TGF-β1, TNF-α, and the M1 macrophage marker iNOS, CD86. Furthermore, Pirfenidone dramatically reduced the mRNA expression of the JAK2/STAT3. These findings imply that Pirfenidone may reduce pulmonary fibrosis in silicosis rats by inhibiting macrophage polarization via the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Qiong Tang
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China; Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong 250000, China
| | - Chen Xing
- Jinan Center For Disease Control And Prevention, Jinan, Shandong 250000, China
| | - Ming Li
- Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong 250000, China
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong 250000, China
| | - Cunxiang Bo
- Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong 250000, China.
| | - Zhenling Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong 250000, China.
| |
Collapse
|
28
|
Peng Z, Duan M, Zhao K, Tang Y, Liang F. RAB20 deficiency promotes the development of silicosis via NLRP3 inflammasome. Front Immunol 2022; 13:967299. [PMID: 36131930 PMCID: PMC9484360 DOI: 10.3389/fimmu.2022.967299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Silicosis is a worldwide serious occupational disease that is caused by inhalation of silica crystals. However, little is known about the pathogenesis mechanism of silicosis. We performed single-cell sequencing in bronchoalveolar lavage fluid (BALF) from mine workers with silicosis and their co-workers who did not develop silicosis, and found that the RAB20 deficiency in monocytes/macrophages was strongly linked to the development of silicosis. In the silicosis murine model, RAB20 knockout markedly enhanced the silica crystal-induced pulmonary interstitial fibrosis and respiratory dysfunction. Moreover, this process is strongly accompanied by IL-1β release and NLRP3 activation. In vitro, RAB20 knockout macrophages aggravated the crystalline silica-induced IL-1β release and NLRP3 inflammasome activation partly by increased ratio of crystalline silica/phagosomal areas/volumes to induce lysosomal injury. Thus, these findings provide novel molecular insights into the intricate mechanisms underlying lysosomal protein RAB20 that are necessary for environmental irritant-mediated innate immunity, and shed light on the future development of novel therapy target for the prevention of silicosis.
Collapse
Affiliation(s)
- Zhouyangfan Peng
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Mingwu Duan
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Kai Zhao
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Yiting Tang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Fang Liang
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Fang Liang,
| |
Collapse
|
29
|
Lv J, Xiao J, Jia Q, Meng X, Yang Z, Pu S, Li M, Yu T, Zhang Y, Wang H, Liu L, Li Z, Chen X, Yang H, Li Y, Qiao M, Duan A, Shao H, Li B. Identification of key pathways and genes in the progression of silicosis based on WGCNA. Inhal Toxicol 2022; 34:304-318. [PMID: 35913820 DOI: 10.1080/08958378.2022.2102700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Silicosis, induced by inhaling silica particles in workplaces, is one of the most common occupational diseases. The prognosis of silicosis and its consequent fibrosis is extremely poor due to limited treatment modalities and lack of understanding of the disease mechanisms. In this study, a Wistar rat model for silicosis fibrosis was established by intratracheal instillation of silica (0, 50, 100 and 200 mg/mL, 1 mL) with the evidence of Hematoxylin and Eosin (HE) and Masson staining and the expressions of inflammatory and fibrotic proteins of rats' lung tissues. RNA of lung tissues of rats exposed to 200 mg/mL silica particles and normal saline for 14 d and 28 d was extracted and sequenced to detect differentially expressed genes (DEGs) and to identify silicosis fibrosis-associated modules and hub genes by Weighted gene co-expression network analysis (WGCNA). Predictions of gene functions and signaling pathways were conducted using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. In this study, it has been demonstrated the promising role of the Hippo signaling pathway in silicosis fibrosis, which will be conducive to elucidating the specific mechanism of pulmonary fibrosis induced by silica and to determining molecular initiating event (MIE) and adverse outcome pathway (AOP) of silicosis fibrosis.
Collapse
Affiliation(s)
- Jiaqi Lv
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jingwei Xiao
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qiang Jia
- Department of Toxicology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, China
| | - Xiangjing Meng
- Department of Toxicology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, China
| | - Zhifeng Yang
- Department of Toxicology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, China
| | - Shuangshuang Pu
- Department of Toxicology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, China
| | - Ming Li
- Department of Toxicology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, China
| | - Tao Yu
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yi Zhang
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haihua Wang
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li Liu
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhongsheng Li
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiao Chen
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haitao Yang
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yulu Li
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Mengyun Qiao
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Airu Duan
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hua Shao
- Department of Toxicology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, China
| | - Bin Li
- Department of Toxicology, Key Lab of Chemical Safety and health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
30
|
Peng Z, Duan M, Tang Y, Wu J, Zhao K, Zhong Y, He Z, Meng J, Chen F, Xiao X, Wang H, Billiar TR, Lu B, Liang F. Impaired interferon-γ signaling promotes the development of silicosis. iScience 2022; 25:104647. [PMID: 35800765 PMCID: PMC9254453 DOI: 10.1016/j.isci.2022.104647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/18/2022] [Accepted: 06/15/2022] [Indexed: 12/27/2022] Open
Abstract
Silicosis is caused by inhalation of crystalline silica dust particles and known as one of the most serious occupational diseases worldwide. However, little is known about intrinsic factors leading to disease susceptibility. Single-cell sequencing of bronchoalveolar lavage fluid cells of mine workers with silicosis and their co-workers who did not develop silicosis revealed that the impaired interferon (IFN)-γ signaling in myeloid cells was strongly associated with the occurrence of silicosis. Global or myeloid cell-specific deletion of interferon γ receptor (IFN-γR) markedly enhanced the crystalline silica-induced pulmonary injury in wild-type but not in NLRP3 deficient mice. In vitro, IFN-γ priming of macrophages suppressed the crystalline silica-induced NLRP3 inflammasome activation partly by inducing the formation of spacious phagosomes with relatively reduced ratio of crystalline silica/phagosomal areas volumes to resistant crystalline silica-induced lysosomal membrane damage. Thus, these findings provide molecular insights into the intricate mechanisms underlying innate immunity-mediated host responses to environmental irritants.
Collapse
Affiliation(s)
- Zhouyangfan Peng
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Mingwu Duan
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Yiting Tang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410000, P.R. China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Kai Zhao
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Yanjun Zhong
- ICU Center, The Second Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Zhihui He
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Jie Meng
- Department of Respiratory Diseases and Critical Care Illness, The 3Road Xiangya Hospital, Central South University, Changsha 410000, P.R. China
- Hunan Key Laboratory of Organ Fibrosis, Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Fangping Chen
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| | - Xianzhong Xiao
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan Province 410000, P.R. China
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410000, P.R. China
| | - Haichao Wang
- The Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Ben Lu
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan Province 410000, P.R. China
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410000, P.R. China
| | - Fang Liang
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha 410000, P.R. China
| |
Collapse
|
31
|
Li Y, Jin F, Li T, Yang X, Cai W, Li S, Gao X, Mao N, Liu H, Xu H, Yang F. Minute Cellular Nodules as Early Lesions in Rats with Silica Exposure via Inhalation. Vet Sci 2022; 9:vetsci9060251. [PMID: 35737303 PMCID: PMC9227695 DOI: 10.3390/vetsci9060251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 12/01/2022] Open
Abstract
Mechanisms of silicosis have yet to be clarified, and pathological conditions are inaccurately described in some experimental studies on silicosis. This study was aimed at describing initial lesions in silicosis, as observed in rats with silica exposure via inhalation, and major histopathologic alterations. Male Wistar rats were exposed to silica for 24 weeks. Hematoxylin and eosin staining indicated the presence of “cellular nodule+ macrophage alveolitis” in rats exposed to silica from the 2–16 weeks time points and “fibrotic cellular + cellular nodule” in rats exposed to silica via inhalation for 24 weeks. By immunohistochemistry, the following were noted: a continual increase in the positive expression of CD68 in macrophages in the lungs of rats exposed to silica; hyperplasia in alveolar type II cells (AT2); loss of original phenotypes in fibrotic cellular nodules, macrophages, and AT2 cells; loss of endothelial cells in silicotic nodules; and positive expression of α-smooth muscle actin in macrophages. Typical pathological changes in silicosis were also summarized. Among these changes were macrophage alveolitis, cellular nodules, and fibrotic cellular nodules, including an increase in minute cellular nodules in the early stages and the formation of fibrotic cellular nodules in the late stages.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Hong Xu
- Correspondence: (H.X.); (F.Y.); Tel.: +86-151-33967479 (H.X.); +86-188-32571018 (F.Y.)
| | - Fang Yang
- Correspondence: (H.X.); (F.Y.); Tel.: +86-151-33967479 (H.X.); +86-188-32571018 (F.Y.)
| |
Collapse
|
32
|
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
|
33
|
Song MY, Wang JX, Sun YL, Han ZF, Zhou YT, Liu Y, Fan TH, Li ZG, Qi XM, Luo Y, Yang PR, Li BC, Zhang XR, Wang J, Wang C. Tetrandrine alleviates silicosis by inhibiting canonical and non-canonical NLRP3 inflammasome activation in lung macrophages. Acta Pharmacol Sin 2022; 43:1274-1284. [PMID: 34417574 PMCID: PMC9061833 DOI: 10.1038/s41401-021-00693-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/07/2021] [Indexed: 12/02/2022]
Abstract
Silicosis caused by inhalation of silica particles leads to more than ten thousand new occupational exposure-related deaths yearly. Exacerbating this issue, there are currently few drugs reported to effectively treat silicosis. Tetrandrine is the only drug approved for silicosis treatment in China, and despite more than decades of use, its efficacy and mechanisms of action remain largely unknown. Here, in this study, we established silicosis mouse models to investigate the effectiveness of tetrandrine of early and late therapeutic administration. To this end, we used multiple cardiopulmonary function test, as well as markers for inflammation and fibrosis. Moreover, using single cell RNA sequencing and transcriptomics of lung tissue and quantitative microarray analysis of serum from silicosis and control mice, our results provide a novel description of the target pathways for tetrandrine. Specifically, we found that tetrandrine attenuated silicosis by inhibiting both the canonical and non-canonical NLRP3 inflammasome pathways in lung macrophages. Taken together, our work showed that tetrandrine yielded promising results against silicosis-associated inflammation and fibrosis and further lied the groundwork for understanding its molecular targets. Our results also facilitated the wider adoption and development of tetrandirne, potentially accelerating a globally accepted therapeutic strategy for silicosis.
Collapse
Affiliation(s)
- Mei-yue Song
- grid.24695.3c0000 0001 1431 9176Beijing University of Chinese Medicine, Beijing, 100029 China ,grid.415954.80000 0004 1771 3349Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029 China ,grid.415954.80000 0004 1771 3349National Clinical Research Center for Respiratory Diseases, Beijing, 100029 China
| | - Jia-xin Wang
- grid.12527.330000 0001 0662 3178Tsinghua-Peking Center for Life Sciences, Department of Biology, College of Medicine, Tsinghua University, Beijing, 100084 China
| | - You-liang Sun
- grid.12527.330000 0001 0662 3178School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084 China
| | - Zhi-fa Han
- grid.12527.330000 0001 0662 3178Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084 China
| | - Yi-tian Zhou
- grid.506261.60000 0001 0706 7839Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100730 China ,grid.506261.60000 0001 0706 7839Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730 China ,grid.506261.60000 0001 0706 7839Peking Union Medical College, MD Program, Beijing, 100730 China
| | - Ying Liu
- grid.506261.60000 0001 0706 7839Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100730 China
| | - Tian-hui Fan
- grid.506261.60000 0001 0706 7839Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100730 China
| | - Zhao-guo Li
- grid.412463.60000 0004 1762 6325Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 China
| | - Xian-mei Qi
- grid.506261.60000 0001 0706 7839Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100730 China
| | - Ya Luo
- grid.506261.60000 0001 0706 7839Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100730 China
| | - Pei-ran Yang
- grid.506261.60000 0001 0706 7839Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100730 China
| | - Bai-cun Li
- grid.506261.60000 0001 0706 7839Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100730 China
| | - Xin-ri Zhang
- grid.452461.00000 0004 1762 8478Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, 030001 China
| | - Jing Wang
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100730, China.
| | - Chen Wang
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
34
|
Yang S, Liu P, Gao T, Song D, Zhao X, Li Y, Wu J, Wang L, Wang Z, Hao J, Wang C, Dai H. Every road leads to Rome: therapeutic effect and mechanism of the extracellular vesicles of human embryonic stem cell-derived immune and matrix regulatory cells administered to mouse models of pulmonary fibrosis through different routes. Stem Cell Res Ther 2022; 13:163. [PMID: 35413874 PMCID: PMC9006546 DOI: 10.1186/s13287-022-02839-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/02/2021] [Indexed: 12/27/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal interstitial lung disease. Whether extracellular vesicles are effective in treating IPF and what is the optimal administrative route is not clear. Our previous studies have shown that immunity and matrix regulatory cells (IMRCs) derived from human embryonic stem cells can safely treat lung injury and fibrosis in mouse models, and its mechanism of action is related to the paracrine effect. In this study, we investigated the therapeutic effects of IMRC-derived extracellular vesicles (IMRC-EVs) on a bleomycin-induced pulmonary fibrosis mouse model and explored the optimal route of administration. Methods To study the biodistribution of IMRC-EVs after administration via different routes, NIR labeled-IMRC-EVs were delivered by intratracheal (IT) or intravenous (IV) route, and in vivo imaging was acquired at different time points. The therapeutic effects of IMRC-EVs delivered by different routes were analyzed by assessing histology, lung function, cytokines levels, and transcriptome profiling. RNA-seq of lung tissues was performed to investigate the mechanisms of EV treatment through IT or IV administrations. Results IMRC-EVs mainly reserved in the liver and spleen when administrated via IV route; and mainly retained in the lungs via the IT route. IMRC-EVs administrated via both routes demonstrated a therapeutic effect as attenuated pulmonary fibrosis, improved lung function, and histological parameters. Based on our RNA-seq results, different pathways may be affected by IMRC-EVs administrated via IT or IV routes. In addition, in vitro experiments showed that IMRC-EVs inhibited epithelial-to-mesenchymal transition induced by TGF-β. Conclusion IMRC-EVs administrated via IT or IV routes generate different biodistributions, but are both effective for the treatment of bleomycin-induced pulmonary fibrosis. The therapeutic mechanisms of IMRC-EVs administrated via different routes may be different. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02839-7.
Collapse
Affiliation(s)
- Shengnan Yang
- Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.,National Center for Respiratory Medicine, Beijing, 100029, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
| | - Peipei Liu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.,National Center for Respiratory Medicine, Beijing, 100029, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China.,Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Tingting Gao
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, 100190, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Dingyun Song
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.,National Center for Respiratory Medicine, Beijing, 100029, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
| | - Xinyu Zhao
- Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.,National Center for Respiratory Medicine, Beijing, 100029, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
| | - Yupeng Li
- Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.,National Center for Respiratory Medicine, Beijing, 100029, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
| | - Jun Wu
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, 100190, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Liu Wang
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, 100190, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Jie Hao
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, 100190, China. .,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China. .,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Chen Wang
- Harbin Medical University, Harbin, 150081, Heilongjiang Province, China. .,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China. .,National Center for Respiratory Medicine, Beijing, 100029, China. .,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China. .,Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China.
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China. .,National Center for Respiratory Medicine, Beijing, 100029, China. .,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China.
| |
Collapse
|
35
|
Zhang J, Yuan C, Li E, Guo Y, Cui J, Liu H, Hao X, Guo L. The significance of serum S100 calcium-binding protein A4 in silicosis. BMC Pulm Med 2022; 22:127. [PMID: 35379204 PMCID: PMC8981710 DOI: 10.1186/s12890-022-01918-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/22/2022] [Indexed: 11/19/2022] Open
Abstract
Background Silicosis is a chronic occupational pulmonary disease characterized by persistent inflammation and irreversible fibrosis. Considerable evidences now indicate that S100 calcium-binding protein A4 (S100A4) has been associated with fibrotic diseases. However, the role of S100A4 in silicosis is still unclear. Methods In this study, serum levels of S100A4, transforming growth factor-β1 (TGF-β1), connective tissue growth factor (CTGF), interleukin-6 (IL-6) and tumour necrosis factor-α (TNF-α) in patients with silicosis (n = 42) and control group (CG, n = 12) were measured by ELISA. S100A4 expression in lung tissues and primary alveolar macrophages (AMs) of mice with and without silicosis was detected by immunohistochemistry (IHC)/real-time PCR. The correlations between S100A4 and cytokines or lung function were assessed by Spearman's rank correlation analyses. Results Compared with CG, the levels of S100A4 were significantly increased in silicosis patients (70.84 (46.22, 102.46) ng/ml vs (49.84 (42.86, 60.02) ng/ml). The secretions of TGF-β1, CTGF, IL-6 and TNF-α in silicosis group were significantly higher than that in control group (p < 0.05). Serum S100A4 levels were positively correlated with TGF-β1 and IL-6, while were negatively correlated with lung function parameters including percentage of predicted forced vital capacity (FVC%pre), maximum vital capacity (Vcmax), deep inspiratory capacity (IC) and peak expiratory flow at 75% of vital capacity (PEF75). In receiver operating characteristic (ROC) analyses, S100A4 > 61.7 ng/ml had 63.4% sensitivity and 83.3% specificity for silicosis, and the area under the curve (AUC) was 0.707. Furthermore, immunostaining of lung tissues showed the accumulation of S100A4-positive cells in the areas of nodules of silicotic mice. The mRNA expression of S100A4 in the lung tissues and AMs of silicotic mice were significantly higher than controls. Conclusion These data suggested that increased S100A4 might contribute to the pathogenesis of silicosis.
Collapse
Affiliation(s)
- Jing Zhang
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Cuifang Yuan
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Enhong Li
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Yiming Guo
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Jie Cui
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Heliang Liu
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Xiaohui Hao
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, Hebei, China
| | - Lingli Guo
- School of Public Health, Hebei Key Laboratory for Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, Hebei, China.
| |
Collapse
|
36
|
Cao ZJ, Liu Y, Zhang Z, Yang PR, Li ZG, Song MY, Qi XM, Han ZF, Pang JL, Li BC, Zhang XR, Dai HP, Wang J, Wang C. Pirfenidone ameliorates silica-induced lung inflammation and fibrosis in mice by inhibiting the secretion of interleukin-17A. Acta Pharmacol Sin 2022; 43:908-918. [PMID: 34316030 DOI: 10.1038/s41401-021-00706-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/24/2021] [Indexed: 12/21/2022]
Abstract
Silicosis is a global occupational disease characterized by lung dysfunction, pulmonary inflammation, and fibrosis, for which there is a lack of effective drugs. Pirfenidone has been shown to exert anti-inflammatory and anti-fibrotic properties in the lung. However, whether and how pirfenidone is effective against silicosis remains unknown. Here, we evaluated the efficacy of pirfenidone in the treatment of early and advanced silicosis in an experimental mouse model and explored its potential pharmacological mechanisms. We found that pirfenidone alleviated silica-induced lung dysfunction, secretion of inflammatory cytokines (TNF-α, IL-1β, IL-6) and deposition of fibrotic proteins (collagen I and fibronectin) in both early and advanced silicosis models. Moreover, we observed that both 100 and 200 mg/kg pirfenidone can effectively treat early-stage silicosis, while 400 mg/kg was recommended for advanced silicosis. Mechanistically, antibody array and bioinformatic analysis showed that the pathways related to IL-17 secretion, including JAK-STAT pathway, Th17 differentiation, and IL-17 pathway, might be involved in the treatment of silicosis by pirfenidone. Further in vivo experiments confirmed that pirfenidone reduced the production of IL-17A induced by silica exposure via inhibiting STAT3 phosphorylation. Neutralizing IL-17A by anti-IL-17A antibody improved lung function and reduced pulmonary inflammation and fibrosis in silicosis animals. Collectively, our study has demonstrated that pirfenidone effectively ameliorated silica-induced lung dysfunction, pulmonary inflammation and fibrosis in mouse models by inhibiting the secretion of IL-17A.
Collapse
|
37
|
Zhao M, Wang L, Wang M, Bao Q, Qian R, Peng L, Fang A, Du W, Xie L, Zhang Z, Yao Y, Zhang B. Alveolar macrophage-derived progranulin mediated pro-inflammatory Il-6 expression via regulating Creb1 in silicosis model. Int Immunopharmacol 2022; 107:108705. [PMID: 35338960 DOI: 10.1016/j.intimp.2022.108705] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/03/2022] [Accepted: 03/12/2022] [Indexed: 11/05/2022]
Abstract
Progranulin (PGRN) is a secreted factor involved in inflammatory diseases. However, the function of PGRN in silica-induced lung inflammation has not been elucidated. In this study, we demonstrated that PGRN in serum and lung tissues was markedly increased in silicosis mouse model. And immunohistochemistry results showed that PGRN was mainly expressed in alveolar macrophages, which was further confirmed in silica-treated alvelar macrophages cell line (MH-S) in vitro. PGRN promoted pro-inflammatory cytokines transcription such as interleukin (Il)-6, tumor necrosis factor-α (Tnf-α) and Il-1β in MH-S cells, and the increasing of Il-6 was most obvious. Knockdown of PGRN blocked the silica-induced elevation of intracellular Il-6 in MH-S cells. Furthermore, we also found that PGRN could increase the phosphorylation of Cyclic AMP-responsive element-binding protein 1 (Creb1), a transcriptional regulator of Il-6. Inhibition of p-Creb1 by the phosphorylation inhibitor of Creb1 (666-15) decreased PGRN-induced intracellular Il-6 production in MH-S cells. In conclusion, PGRN was highly increased in silicosis mouse model and upregulated inflammatory cytokines expression. These findings suggested that PGRN might be a key mediator in silica-induced inflammation and provided a new clue for the diagnosis and drug therapy of silicosis.
Collapse
Affiliation(s)
- Manyu Zhao
- Department of Environment and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liqun Wang
- Department of Hygienic Toxicology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mengzhu Wang
- Department of Environment and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qixue Bao
- Department of Environment and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rui Qian
- Department of Hygienic Toxicology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lijun Peng
- Department of Occupational Disease, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Aiping Fang
- Department of Occupational Disease, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Wen Du
- Department of Occupational Disease, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Linshen Xie
- Department of Occupational Disease, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Zunzhen Zhang
- Department of Environment and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuqin Yao
- Department of Environment and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Department of Hygienic Toxicology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Department of Occupational Disease, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Ben Zhang
- Department of Environment and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Department of Occupational Disease, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
38
|
Fan M, Xiao H, Song D, Zhu L, Zhang J, Zhang X, Wang J, Dai H, Wang C. A Novel N-Arylpyridone Compound Alleviates the Inflammatory and Fibrotic Reaction of Silicosis by Inhibiting the ASK1-p38 Pathway and Regulating Macrophage Polarization. Front Pharmacol 2022; 13:848435. [PMID: 35401236 PMCID: PMC8983992 DOI: 10.3389/fphar.2022.848435] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/03/2022] [Indexed: 11/21/2022] Open
Abstract
Silicosis is one of the potentially fatal occupational diseases characterized by respiratory dysfunction, chronic interstitial inflammation, and fibrosis, for which treatment options are limited. Previous studies showed that a novel N-arylpyridone compound named AKEX0011 exhibited anti-inflammatory and anti-fibrotic effects in bleomycin-induced pulmonary fibrosis; however, it is unknown whether it could also be effective against silicosis. Therefore, we sought to investigate the preventive and therapeutic roles of AKEX0011 in a silicosis rodent model and in a silica-stimulated macrophage cell line. In vivo, our results showed that AKEX0011 ameliorated silica-induced imaging lung damages, respiratory dysfunction, reduced the secretion of inflammatory and fibrotic factors (TNF-α, IL-1β, IL-6, TGF-β, IL-4, and IL-10), and the deposition of fibrosis-related proteins (collagen I, fibronectin, and α-SMA), regardless of early or advanced therapy. Specifically, we found that AKEX0011 attenuated silicosis by inhibiting apoptosis, blocking the ASK1-p38 MAPK signaling pathway, and regulating polarization of macrophages. In vitro, AKEX0011 inhibited macrophages from secreting inflammatory cytokines and inhibited apoptosis of macrophages in pre-treated and post-treated models, concurrent with blocking the ASK1-p38 pathway and inhibiting M1 polarization. Collectively, AKEX0011, as a novel N-arylpyridone compound, exerted protective effects for silica-induced pulmonary inflammation and fibrosis both in vivo and in vitro, and hence, it could be a strong drug candidate for the treatment of silicosis.
Collapse
Affiliation(s)
- Mingming Fan
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Jilin, China,Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Huijuan Xiao
- Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,Department of Pulmonary and Critical Care Medicine, China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, China
| | - Dingyun Song
- Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lili Zhu
- Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jie Zhang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Jilin, China
| | - Xinran Zhang
- Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,*Correspondence: Huaping Dai, ; Jing Wang, ; Chen Wang,
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,*Correspondence: Huaping Dai, ; Jing Wang, ; Chen Wang,
| | - Chen Wang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Jilin, China,Department of Pulmonary and Critical Care Medicine Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China,*Correspondence: Huaping Dai, ; Jing Wang, ; Chen Wang,
| |
Collapse
|
39
|
Song M, Wang J, Sun Y, Pang J, Li X, Liu Y, Zhou Y, Yang P, Fan T, Liu Y, Li Z, Qi X, Li B, Zhang X, Wang J, Wang C. Inhibition of gasdermin D-dependent pyroptosis attenuates the progression of silica-induced pulmonary inflammation and fibrosis. Acta Pharm Sin B 2022; 12:1213-1224. [PMID: 35530143 PMCID: PMC9069405 DOI: 10.1016/j.apsb.2021.10.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/12/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022] Open
Abstract
Silicosis is a leading cause of occupational disease-related morbidity and mortality worldwide, but the molecular basis underlying its development remains unclear. An accumulating body of evidence supports gasdermin D (GSDMD)-mediated pyroptosis as a key component in the development of various pulmonary diseases. However, there is little experimental evidence connecting silicosis and GSDMD-driven pyroptosis. In this work, we investigated the role of GSDMD-mediated pyroptosis in silicosis. Single-cell RNA sequencing of healthy and silicosis human and murine lung tissues indicated that GSDMD-induced pyroptosis in macrophages was relevant to silicosis progression. Through microscopy we then observed morphological alterations of pyroptosis in macrophages treated with silica. Measurement of interleukin-1β release, lactic dehydrogenase activity, and real-time propidium iodide staining further revealed that silica induced pyroptosis of macrophages. Additionally, we verified that both canonical (caspase-1-mediated) and non-canonical (caspase-4/5/11-mediated) signaling pathways mediated silica-induced pyroptosis activation, in vivo and in vitro. Notably, Gsdmd knockout mice exhibited dramatically alleviated silicosis phenotypes, which highlighted the pivotal role of pyroptosis in this disease. Taken together, our results demonstrated that macrophages underwent GSDMD-dependent pyroptosis in silicosis and inhibition of this process could serve as a viable clinical strategy for mitigating silicosis.
Collapse
|
40
|
Lv J, Zhang Y, Yu T, Yang H, Li Y, Xiao J, Li B. A promising transwell co-culture cell model for silicosis. Toxicol In Vitro 2022; 81:105318. [DOI: 10.1016/j.tiv.2022.105318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/03/2022] [Accepted: 01/17/2022] [Indexed: 10/19/2022]
|
41
|
Cai Q, Ma J, Wang J, Wang J, Cui J, Wu S, Wang Z, Wang N, Wang J, Yang D, Yang J, Xue J, Li F, Chen J, Liu X. Adenoviral Transduction of Dickkopf-1 Alleviates Silica-Induced Silicosis Development in Lungs of Mice. Hum Gene Ther 2021; 33:155-174. [PMID: 34405699 DOI: 10.1089/hum.2021.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Silicosis is an occupational disease caused by inhalation of silica dust, which is hallmarked by progressive pulmonary fibrosis associated with poor prognosis. Wnt/β-catenin signaling is implicated in the development of fibrosis and is a therapeutic target for fibrotic diseases. Previous clinical studies of patients with pneumoconiosis, including silicosis, revealed an increased concentration of circulating WNT3A and DKK1 proteins and inflammatory cells in bronchoalveolar lavage compared with healthy subjects. The present study evaluated the effects of adenovirus-mediated transduction of Dickkopf-1 (Dkk1), a Wnt/β-catenin signaling inhibitor, on the development of pulmonary silicosis in mice. Consistent with previous human clinical studies, our experimental studies in mice demonstrated an aberrant Wnt/β-catenin signaling activity coinciding with increased Wnt3a and Dkk1 proteins and inflammation in lungs of silica-induced silicosis mice compared with controls. Intratracheal delivery of adenovirus expressing murine Dkk1 (AdDkk1) inhibited Wnt/β-catenin activity in mouse lungs. The adenovirus-mediated Dkk1 gene transduction demonstrated the potential to prevent silicosis development and ameliorate silica-induced lung fibrogenesis in mice, accompanied by the reduced expression of epithelia--mesenchymal transition markers and deposition of extracellular matrix proteins compared with mice treated with "null" adenoviral vector. Mechanistically, AdDkk1 is able to attenuate the lung silicosis by inhibiting a silica-induced spike in TGF-β/Smad signaling. In addition, the forced expression of Dkk1 suppressed silica-induced epithelial cell proliferation in polarized human bronchial epithelial cells. This study provides insight into the underlying role of Wnt/β-catenin signaling in promoting the pathogenesis of silicosis and is proof-of-concept that targeting Wnt/β-catenin signaling by Dkk1 gene transduction may be an alternative approach in the prevention and treatment of silicosis lung disease.
Collapse
Affiliation(s)
- Qian Cai
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources of Western China, College of Life Science, Ningxia University, Yinchuan, China.,Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa, USA.,Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Jia Ma
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources of Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Jing Wang
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Juying Wang
- Department of Occupational Disease, The Fifth People's Hospital of Ningxia, Shizuishan, China
| | - Jieda Cui
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Shuang Wu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources of Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Zhaojun Wang
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Na Wang
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jiaqi Wang
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Dandan Yang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources of Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Jiali Yang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources of Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Jing Xue
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources of Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Feng Li
- Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Juan Chen
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiaoming Liu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources of Western China, College of Life Science, Ningxia University, Yinchuan, China.,Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
42
|
Cheng D, Xu Q, Wang Y, Li G, Sun W, Ma D, Zhou S, Liu Y, Han L, Ni C. Metformin attenuates silica-induced pulmonary fibrosis via AMPK signaling. J Transl Med 2021; 19:349. [PMID: 34399790 PMCID: PMC8365894 DOI: 10.1186/s12967-021-03036-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/09/2021] [Indexed: 02/08/2023] Open
Abstract
Background Silicosis is one of the most common occupational pulmonary fibrosis caused by respirable silica-based particle exposure, with no ideal drugs at present. Metformin, a commonly used biguanide antidiabetic agent, could activate AMP-activated protein kinase (AMPK) to exert its pharmacological action. Therefore, we sought to investigate the role of metformin in silica-induced lung fibrosis. Methods The anti-fibrotic role of metformin was assessed in 50 mg/kg silica-induced lung fibrosis model. Silicon dioxide (SiO2)-stimulated lung epithelial cells/macrophages and transforming growth factor-beta 1 (TGF-β1)-induced differentiated lung fibroblasts were used for in vitro models. Results At the concentration of 300 mg/kg in the mouse model, metformin significantly reduced lung inflammation and fibrosis in SiO2-instilled mice at the early and late fibrotic stages. Besides, metformin (range 2–10 mM) reversed SiO2-induced cell toxicity, oxidative stress, and epithelial-mesenchymal transition process in epithelial cells (A549 and HBE), inhibited inflammation response in macrophages (THP-1), and alleviated TGF-β1-stimulated fibroblast activation in lung fibroblasts (MRC-5) via an AMPK-dependent pathway. Conclusions In this study, we identified that metformin might be a potential drug for silicosis treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03036-5.
Collapse
Affiliation(s)
- Demin Cheng
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Qi Xu
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yue Wang
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Guanru Li
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wenqing Sun
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Dongyu Ma
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Siyun Zhou
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yi Liu
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Lei Han
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210028, China.
| | - Chunhui Ni
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|