1
|
Ning G, Guo X, Zhu K, Xu Z, Cai P, Dang Y, Lu C, Xu F, Shen R, Kang N, Zhang R, Chen K. Human decidual mesenchymal stem cells obtained from early pregnancy attenuate bleomycin-induced lung fibrosis by inhibiting inflammation and apoptosis. Int Immunopharmacol 2024; 142:113224. [PMID: 39306886 DOI: 10.1016/j.intimp.2024.113224] [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: 04/14/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Decidual mesenchymal stem cells (DMSCs) are easily obtained and exhibit strong anti-inflammatory and anti-apoptotic effects. Compared with bone marrow mesenchymal stem cells (BMSCs), their role in cell transplantation after idiopathic pulmonary fibrosis remains unclear. We investigated whether the transplantation of BMSCs and DMSCs could alleviate pulmonary inflammation and fibrosis in a bleomycin-induced mouse model of pulmonary fibrosis. METHODS BMSCs and DMSCs were derived from healthy donors. The anti-inflammatory and anti-apoptotic effects on both cell types were evaluated in vitro. The function of DMSCs in MLE-12 cells and mouse lung fibroblasts was examined using additional transwell coculture experiments in vitro. Twenty-one days after MSC transplantation, we examined the inflammatory factors in the serum and bronchoalveolar lavage fluid, collagen content, pathology, fibrotic area, lung function, and micro-computed tomography of the lung tissue. RESULTS DMSCs exhibited better anti-inflammatory and anti-apoptotic effects than BMSCs on MLE-12 cells in vitro. In addition, DMSCs inhibited tumor growth factor β-dependent epithelial-mesenchymal transition in MLE-12 cells and attenuated mouse lung fibroblasts fibrosis. Furthermore, transplantation of DMSCs in the mouse idiopathic pulmonary fibrosis model significantly attenuated pulmonary inflammation and lung fibrosis compared with BMSCs transplantation. CONCLUSIONS DMSCs exhibited better efficacy in improving pulmonary inflammation and lung fibrosis than BMSCs. Thus, DMSCs are a potential therapeutic target for pulmonary fibrosis.
Collapse
Affiliation(s)
- Guangyao Ning
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Xiaohui Guo
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Kechao Zhu
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Ziqiang Xu
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Peian Cai
- Department of Thoracic Surgery, Henan Cancer Hospital, Zhengzhou, PR China
| | - Yan Dang
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Chen Lu
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Feng Xu
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Ruifang Shen
- Laboratory for Space Environment and Physical Sciences, Harbin Institute of Technology, Harbin, PR China
| | - Ningning Kang
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China.
| | - Renquan Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China.
| | - Kegong Chen
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Research and experiment center, the First Affiliated Hospital of Anhui Medical University, Hefei, PR China.
| |
Collapse
|
2
|
Kühl F, Brand K, Lichtinghagen R, Huber R. GSK3-Driven Modulation of Inflammation and Tissue Integrity in the Animal Model. Int J Mol Sci 2024; 25:8263. [PMID: 39125833 PMCID: PMC11312333 DOI: 10.3390/ijms25158263] [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: 06/28/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Nowadays, GSK3 is accepted as an enzyme strongly involved in the regulation of inflammation by balancing the pro- and anti-inflammatory responses of cells and organisms, thus influencing the initiation, progression, and resolution of inflammatory processes at multiple levels. Disturbances within its broad functional scope, either intrinsically or extrinsically induced, harbor the risk of profound disruptions to the regular course of the immune response, including the formation of severe inflammation-related diseases. Therefore, this review aims at summarizing and contextualizing the current knowledge derived from animal models to further shape our understanding of GSK3α and β and their roles in the inflammatory process and the occurrence of tissue/organ damage. Following a short recapitulation of structure, function, and regulation of GSK3, we will focus on the lessons learned from GSK3α/β knock-out and knock-in/overexpression models, both conventional and conditional, as well as a variety of (predominantly rodent) disease models reflecting defined pathologic conditions with a significant proportion of inflammation and inflammation-related tissue injury. In summary, the literature suggests that GSK3 acts as a crucial switch driving pro-inflammatory and destructive processes and thus contributes significantly to the pathogenesis of inflammation-associated diseases.
Collapse
Affiliation(s)
| | | | | | - René Huber
- Institute of Clinical Chemistry and Laboratory Medicine, Hannover Medical School, 30625 Hannover, Germany; (F.K.); (K.B.); (R.L.)
| |
Collapse
|
3
|
Su W, Nong Q, Wu J, Fan R, Liang Y, Hu A, Gao Z, Liang W, Deng Q, Wang H, Xia L, Huang Y, Qin Y, Zhao N. Anti-inflammatory protein TSG-6 secreted by BMSCs attenuates silica-induced acute pulmonary inflammation by inhibiting NLRP3 inflammasome signaling in macrophages. Int J Biol Macromol 2023; 253:126651. [PMID: 37709227 DOI: 10.1016/j.ijbiomac.2023.126651] [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: 03/13/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023]
Abstract
Silicosis is a severe occupational lung disease caused by inhalation of silica particles. Unfortunately, there are currently limited treatment options available for silicosis. Recent advances have indicated that bone marrow mesenchymal stem cells (BMSCs) have a therapeutic effect on silicosis, but their efficacy and underlying mechanisms remain largely unknown. In this study, we focused on the early phase of silica-induced lung injury to investigate the therapeutic effect of BMSCs. Our findings demonstrated that BMSCs attenuated silica-induced acute pulmonary inflammation by inhibiting NLRP3 inflammasome pathways in lung macrophages. To further understand the mechanisms involved, we utilized RNA sequencing to analyze the transcriptomes of BMSCs co-cultured with silica-stimulated bone marrow-derived macrophages (BMDMs). The results clued tumor necrosis factor-stimulated gene 6 (TSG-6) might be a potentially key paracrine secretion factor released from BMSCs, which exerts a protective effect. Furthermore, the anti-inflammatory and inflammasome pathway inhibition effects of BMSCs were attenuated when TSG-6 expression was silenced, both in vivo and in vitro. Additionally, treatment with exogenous recombinant mouse TSG-6 (rmTSG-6) demonstrated similar effects to BMSCs in attenuating silica-induced inflammation. Overall, our findings suggested that BMSCs can regulate the activation of inflammasome in macrophages by secreting TSG-6, thereby protecting against silica-induced acute pulmonary inflammation both in vivo and in vitro.
Collapse
Affiliation(s)
- Wenyao Su
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China; Shunde Women and Children's Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong 528300, China; School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Qiying Nong
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China
| | - Jie Wu
- Emeishan Centerfor Disease Control and Prevention, Emeishan, Sichuan 614299, China
| | - Ruihong Fan
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China
| | - Yuanting Liang
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China; School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Anyi Hu
- Health Science Center of Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Zhongxiang Gao
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China
| | - Weihui Liang
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China
| | - Qifei Deng
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China
| | - Hailan Wang
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China
| | - Lihua Xia
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China
| | - Yongshun Huang
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China
| | - Yiru Qin
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China.
| | - Na Zhao
- Guangdong Province Hospital for Occupational Diseases Prevention and Treatment, Guangzhou 510300, China; School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong 511436, China; School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
4
|
Bayati P, Taherian M, Soleimani M, Farajifard H, Mojtabavi N. Induced pluripotent stem cells modulate the Wnt pathway in the bleomycin-induced model of idiopathic pulmonary fibrosis. Stem Cell Res Ther 2023; 14:343. [PMID: 38017561 PMCID: PMC10685538 DOI: 10.1186/s13287-023-03581-4] [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: 09/23/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND The Wnt signaling pathway has been implicated in the pathogenesis of fibrotic disorders and malignancies. Hence, we aimed to assess the potential of the induced pluripotent stem cells (IPS) in modulating the expression of the cardinal genes of the Wnt pathway in a mouse model of idiopathic pulmonary fibrosis (IPF). METHODS C57Bl/6 mice were randomly divided into three groups of Control, Bleomycin (BLM), and BLM + IPS; the BLM mice received intratracheal instillation of bleomycin, BLM + IPS mice received tail vein injection of IPS cells 48 h post instillation of the BLM; The Control group received Phosphate-buffered saline instead. After 3 weeks, the mice were sacrificed and Histologic assessments including hydroxy proline assay, Hematoxylin and Eosin, and Masson-trichrome staining were performed. The expression of the genes for Wnt, β-Catenin, Lef, Dkk1, and Bmp4 was assessed utilizing specific primers and SYBR green master mix. RESULTS Histologic assessments revealed that the fibrotic lesions and inflammation were significantly alleviated in the BLM + IPS group. Besides, the gene expression analyses demonstrated the upregulation of Wnt, β-Catenin, and LEF along with the significant downregulation of the Bmp4 and DKK1 in response to bleomycin treatment; subsequently, it was found that the treatment of the IPF mice with IPS cells results in the downregulation of the Wnt, β-Catenin, and Lef, as well as upregulation of the Dkk1, but not the Bmp4 gene (P values < 0.05). CONCLUSION The current study highlights the therapeutic potential of the IPS cells on the IPF mouse model in terms of regulating the aberrant expression of the factors contributing to the Wnt signaling pathway.
Collapse
Affiliation(s)
- Paria Bayati
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marjan Taherian
- Immunology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Farajifard
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin Mojtabavi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Chai R, Li Y, Shui L, Ni L, Zhang A. The role of pyroptosis in inflammatory diseases. Front Cell Dev Biol 2023; 11:1173235. [PMID: 37250902 PMCID: PMC10213465 DOI: 10.3389/fcell.2023.1173235] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/18/2023] [Indexed: 05/31/2023] Open
Abstract
Programmed cell death has crucial roles in the physiological maturation of an organism, the maintenance of metabolism, and disease progression. Pyroptosis, a form of programmed cell death which has recently received much attention, is closely related to inflammation and occurs via canonical, non-canonical, caspase-3-dependent, and unclassified pathways. The pore-forming gasdermin proteins mediate pyroptosis by promoting cell lysis, contributing to the outflow of large amounts of inflammatory cytokines and cellular contents. Although the inflammatory response is critical for the body's defense against pathogens, uncontrolled inflammation can cause tissue damage and is a vital factor in the occurrence and progression of various diseases. In this review, we briefly summarize the major signaling pathways of pyroptosis and discuss current research on the pathological function of pyroptosis in autoinflammatory diseases and sterile inflammatory diseases.
Collapse
Affiliation(s)
| | | | | | - Longxing Ni
- *Correspondence: Longxing Ni, ; Ansheng Zhang,
| | | |
Collapse
|
6
|
Liang TY, Lu LH, Tang SY, Zheng ZH, Shi K, Liu JQ. Current status and prospects of basic research and clinical application of mesenchymal stem cells in acute respiratory distress syndrome. World J Stem Cells 2023; 15:150-164. [PMID: 37180997 PMCID: PMC10173811 DOI: 10.4252/wjsc.v15.i4.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/20/2023] [Accepted: 03/20/2023] [Indexed: 04/26/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common and clinically devastating disease that causes respiratory failure. Morbidity and mortality of patients in intensive care units are stubbornly high, and various complications severely affect the quality of life of survivors. The pathophysiology of ARDS includes increased alveolar-capillary membrane permeability, an influx of protein-rich pulmonary edema fluid, and surfactant dysfunction leading to severe hypoxemia. At present, the main treatment for ARDS is mechanical treatment combined with diuretics to reduce pulmonary edema, which primarily improves symptoms, but the prognosis of patients with ARDS is still very poor. Mesenchymal stem cells (MSCs) are stromal cells that possess the capacity to self-renew and also exhibit multilineage differentiation. MSCs can be isolated from a variety of tissues, such as the umbilical cord, endometrial polyps, menstrual blood, bone marrow, and adipose tissues. Studies have confirmed the critical healing and immunomodulatory properties of MSCs in the treatment of a variety of diseases. Recently, the potential of stem cells in treating ARDS has been explored via basic research and clinical trials. The efficacy of MSCs has been shown in a variety of in vivo models of ARDS, reducing bacterial pneumonia and ischemia-reperfusion injury while promoting the repair of ventilator-induced lung injury. This article reviews the current basic research findings and clinical applications of MSCs in the treatment of ARDS in order to emphasize the clinical prospects of MSCs.
Collapse
Affiliation(s)
- Tian-Yu Liang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, China
| | - Li-Hai Lu
- Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Si-Yu Tang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Zi-Hao Zheng
- Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Kai Shi
- Department of Respiratory Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| | - Jing-Quan Liu
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, China.
| |
Collapse
|
7
|
Gao J, Liang Y, Chen J, Shen H, Liu H. CXCR4 enhances the inhibitory effects of bone mesenchymal stem cells on lung cell apoptosis in a rat model of smoking-induced COPD. Apoptosis 2023; 28:639-652. [PMID: 36719470 PMCID: PMC9888343 DOI: 10.1007/s10495-022-01800-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2022] [Indexed: 02/01/2023]
Abstract
Chronic obstructive pulmonary disease is the 3rd leading cause of death worldwide, and the available treatments are unsatisfactory, resulting in a major economic burden. As cellular therapy is commonly used for lung disease, we investigated a treatment with CXCR4-overexpressing BMSCs in a COPD model. We extracted and purified Bone marrow mesenchymal stem cells (BMSCs) from SD rats. COPD apoptosis model was established by cigarette smoke exposure. BMSCs (1 × 106 cells per injection)were transplanted in vivo twice a month during model establishment, and alveolar rupture in the lung was assessed. Lung cell apoptosis was assessed by terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling (TUNEL) analysis, and the concentrations of apoptotic proteins in the lungs were detected by Western blotting. We successfully isolated BMSCs and established CXCR4-overexpressing BMSCs. qRT‒PCR and Western blotting detection both reveal that CXCR4 mRNA level and protein both significantly higher expression in CXCR4-BMSCs than the pBABE-BMSCs. Continuous cigarette smoke exposure caused alveolar septal rupture: In the model group, the alveolar mean linear intercept in the first month was significantly lower than that in the third month (p < 0.05). In the third month, the alveolar mean linear intercept values of the control and CXCR4-BMSC groups were lower than those of the model group (control group p < 0.01, CXCR4-BMSC group p < 0.05), and TUNEL staining revealed that the apoptosis rates of the control and CXCR4-BMSC groups were significantly lower than those of the model group (p < 0.01). Furthermore, the levels of the apoptotic proteins cleaved caspase-8, cleaved caspase-3 and cleaved PARP-1 were higher in the model group than in the control group (p < 0.05) and significantly lower in the CXCR4-BMSC group than in the model group (p < 0.05). The transplantation of CXCR4-overexpressing BMSCs during COPD model generation significantly inhibited apoptosis via the extrinsic apoptosis pathway. CXCR4 enhances the inhibitory effects of bone mesenchymal stem cells on lung cell apoptosis in a rat model of smoking-induced COPD.
Collapse
Affiliation(s)
- Jiansheng Gao
- First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuli Liang
- First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiabao Chen
- First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Huihui Shen
- First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Hua Liu
- First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
8
|
Lopes-Pacheco M, Rocco PRM. Functional enhancement strategies to potentiate the therapeutic properties of mesenchymal stromal cells for respiratory diseases. Front Pharmacol 2023; 14:1067422. [PMID: 37007034 PMCID: PMC10062457 DOI: 10.3389/fphar.2023.1067422] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Respiratory diseases remain a major health concern worldwide because they subject patients to considerable financial and psychosocial burdens and result in a high rate of morbidity and mortality. Although significant progress has been made in understanding the underlying pathologic mechanisms of severe respiratory diseases, most therapies are supportive, aiming to mitigate symptoms and slow down their progressive course but cannot improve lung function or reverse tissue remodeling. Mesenchymal stromal cells (MSCs) are at the forefront of the regenerative medicine field due to their unique biomedical potential in promoting immunomodulation, anti-inflammatory, anti-apoptotic and antimicrobial activities, and tissue repair in various experimental models. However, despite several years of preclinical research on MSCs, therapeutic outcomes have fallen far short in early-stage clinical trials for respiratory diseases. This limited efficacy has been associated with several factors, such as reduced MSC homing, survival, and infusion in the late course of lung disease. Accordingly, genetic engineering and preconditioning methods have emerged as functional enhancement strategies to potentiate the therapeutic actions of MSCs and thus achieve better clinical outcomes. This narrative review describes various strategies that have been investigated in the experimental setting to functionally potentiate the therapeutic properties of MSCs for respiratory diseases. These include changes in culture conditions, exposure of MSCs to inflammatory environments, pharmacological agents or other substances, and genetic manipulation for enhanced and sustained expression of genes of interest. Future directions and challenges in efficiently translating MSC research into clinical practice are discussed.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- *Correspondence: Miquéias Lopes-Pacheco, ; Patricia R. M. Rocco,
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Miquéias Lopes-Pacheco, ; Patricia R. M. Rocco,
| |
Collapse
|
9
|
Chinese medicinal plant Polygonum cuspidatum ameliorates silicosis via suppressing the Wnt/β-catenin pathway. OPEN CHEM 2022. [DOI: 10.1515/chem-2022-0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Abstract
Polygonum cuspidatum (PC) extract has effect on silica-induced pulmonary fibrosis. This study aimed to explore the anti-pulmonary-fibrosis effects and mechanism of PC. Sprague–Dawley rat model was constructed by inhalation of silicon dioxide suspension through tracheal intubation method. And histopathological examination showed that PC inhibited inflammatory cell infiltration, fibrous and collagen hyperplasia, and protected the normal structure of alveoli. TUNEL assay declared that PC retarded cell apoptosis. Meanwhile, up-regulation of basic fibroblast growth factor, plated-derived growth factor, and TNF-α in silicosis rats was decreased by PC addition. In addition, human fetal lung fibroblasts (HFL-1) cells were stimulated with transforming growth factor-β1 (TGF-β1). PC administration increased the proliferation and invasion of TGF-β1-stimulated HFL-1 cells whereas decreased cell apoptosis. Moreover, western blotting exhibited that PC treatment decreased the expression of α-smooth muscle actin, collagen I, and collagen III in silicosis rats and TGF-β1-stimulated HFL-1 cells. Furthermore, the levels of Wnt/β-catenin pathway proteins were up-regulated in silicosis rats and TGF-β1-stimulated HFL-1 cells, which were weakened by PC treatment. Meanwhile, Wnt3a (an activator of Wnt/β-catenin) addition reversed the effect of PC addition. In conclusion, PC prevents silica-induced fibrosis through inhibiting the Wnt/β-catenin pathway.
Collapse
|
10
|
Nassef NAA, Abd-El Hamid MS, Abusikkien SA, Ahmed AI. Quercetin ameliorates acute lung injury in a rat model of hepatopulmonary syndrome. BMC Complement Med Ther 2022; 22:320. [PMID: 36463144 PMCID: PMC9719635 DOI: 10.1186/s12906-022-03785-w] [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] [Received: 05/23/2022] [Accepted: 11/09/2022] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND Common bile duct ligation (BDL) is a rat experimental model to induce biliary cirrhosis. Lung fibrosis and pulmonary vascular angiogenesis and congestion are the most common complications of biliary cirrhosis that is known as hepatopulmonary syndrome. The aim of the present work is to investigate the acute lung injury in a BDL model and to investigate the possible protective effect of quercetin on this injury. METHODS Twenty-four adult male albino rats of the Wister strain (weighing 150-250 g). Animals were divided into 3 groups, with 8 rats each: Group I: Sham-operated group (control). Group II: Bile duct ligation group (BDL) sacrificed after 28 days from the surgery. Group III: Quercetin-treated bile duct ligation group (Q-BDL) was given orally by gastric gavage in a dose of 50 mg/kg/day, starting from the 4th day of the operation until the 28th day. At the end of the experiment, at day 28, all rats were sacrificed. Lung specimens were processed to measure Endothelin B receptor gene expression by PCR, lung surfactant by ELISA, "eNO" s by immunohistochemistry. Histological assessment was done using; H&E, Masson's trichrome, PAS, toluidine blue-stained semi-thin sections, transmission electron microscope. Histomorphometric and statistical studies were done. RESULTS BDL group showed significant increase in lung index together with mononuclear cellular infiltration denoting lung inflammatory state. Also, the significant increase in pulmonary endothelial nitric oxide synthase ("eNO" s) area percent and endothelin B receptor (ETB) gene expression indicates enhanced angiogenesis. Pulmonary surfactant concentration was significantly decreased together with thickening of interalveolar septa denoting lung injury and fibrosis. Quercetin led to significant decrease in lung index, pulmonary "eNO" s area percent, ETB gene expression and significant increase in pulmonary surfactant concentration. Quercetin treatment improved histological changes and morphometric measurements, limited mononuclear cellular infiltration and decreased perivascular and perialveolar collagen deposition. CONCLUSION Quercetin ameliorates the hepatopulmonary syndrome-induced lung injury through its anti-inflammatory, antioxidative and antifibrotic effects.
Collapse
Affiliation(s)
- Noha Abdel-Aziz Nassef
- grid.7269.a0000 0004 0621 1570Assistant Professor of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Manal S. Abd-El Hamid
- grid.7269.a0000 0004 0621 1570Assistant Professor of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Samy A. Abusikkien
- grid.7269.a0000 0004 0621 1570Lecturer of Anatomy, Rabigh Faculty of Medicine, King Abdulaziz University, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Asmaa Ibrahim Ahmed
- grid.7269.a0000 0004 0621 1570Assistant Professor of Anatomy, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
11
|
Fikry H, Saleh LA, Gawad SA. Therapeutic effect of adipose-derived mesenchymal stem cells (AD-MSCs) compared to pirfenidone on corticosteroid resistance in a mouse model of acute exacerbation of idiopathic pulmonary fibrosis. Histol Histopathol 2022; 37:1065-1083. [PMID: 35816024 DOI: 10.14670/hh-18-493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Acute exacerbation-idiopathic pulmonary fibrosis (AE-IPF) is a life-threatening condition. In the treatment of AE-IPF, corticosteroid medication is commonly utilized. However, there is insufficient evidence to justify its usage. Pirfenidone (PFD) has recently been discovered to be effective in the treatment of AE-IPF patients. However, regenerative therapy, such as stem cell therapy or tissue engineering, is necessary due to ineffective and limited therapies. Combining MSC transplantation with pharmacological therapy may also give additional benefits; nevertheless, its use must be proven experimentally. As a result, the goal of this study was to assess the therapeutic effects of adipose-derived mesenchymal stem cells (AD-MSCs) on corticosteroid resistance in an animal model of AE-IPF caused by bleomycin compared to PFD. MATERIALS AND METHODS Seventy C57BL/6J male mice were randomly divided into seven groups, control, BLM, methylprednisolone (MP), PFD, AD-MSCs, PFD +MP, and AD-MSCs +MP. RESULTS In terms of survival, collagen deposition, the acute lung injury score (ALI), and the Ashcroft score, AD-MSCs exceeded PFD. AD-MSCs + MP provided protection and preserved the lung's architecture in BLM-induced AE. In addition, AD-MSCs successfully decreased chemokine (CC motif) ligand-2 (CCL2) positive cells and lower pro-fibrotic and pro-inflammatory cytokines. CONCLUSIONS AD-MSCs enhanced histological structure, Ashcroft and ALI scores, lung collagen deposition, survival, and cytokines in an animal model of AE-IPF. As a result, we believe that AD-MSCs may be more therapeutically helpful for AE-IPF than presently available therapies, either alone or in conjunction with MP.
Collapse
Affiliation(s)
- Heba Fikry
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Lobna A Saleh
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sara Abdel Gawad
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
12
|
Abstract
Pulmonary fibrosis (PF) is a chronic and relentlessly progressive interstitial lung disease in which the accumulation of fibroblasts and extracellular matrix (ECM) induces the destruction of normal alveolar structures, ultimately leading to respiratory failure. Patients with advanced PF are unable to perform physical labor and often have concomitant cough and dyspnea, which markedly impair their quality of life. However, there is a paucity of available pharmacological therapies, and to date, lung transplantation remains the only possible treatment for patients suffering from end-stage PF; moreover, the complexity of transplantation surgery and the paucity of donors greatly restrict the application of this treatment. Therefore, there is a pressing need for alternative therapeutic strategies for this complex disease. Due to their capacity for pluripotency and paracrine actions, stem cells are promising therapeutic agents for the treatment of interstitial lung disease, and an extensive body of literature supports the therapeutic efficacy of stem cells in lung fibrosis. Although stem cell transplantation may play an important role in the treatment of PF, some key issues, such as safety and therapeutic efficacy, remain to be resolved. In this review, we summarize recent preclinical and clinical studies on the stem cell-mediated regeneration of fibrotic lungs and present an analysis of concerning issues related to stem cell therapy to guide therapeutic development for this complex disease.
Collapse
|
13
|
Li C, Wang B. Mesenchymal Stem/Stromal Cells in Progressive Fibrogenic Involvement and Anti-Fibrosis Therapeutic Properties. Front Cell Dev Biol 2022; 10:902677. [PMID: 35721482 PMCID: PMC9198494 DOI: 10.3389/fcell.2022.902677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/13/2022] [Indexed: 11/22/2022] Open
Abstract
Fibrosis refers to the connective tissue deposition and stiffness usually as a result of injury. Fibrosis tissue-resident mesenchymal cells, including fibroblasts, myofibroblast, smooth muscle cells, and mesenchymal stem/stromal cells (MSCs), are major players in fibrogenic processes under certain contexts. Acknowledging differentiation potential of MSCs to the aforementioned other types of mesenchymal cell lineages is essential for better understanding of MSCs’ substantial contributions to progressive fibrogenesis. MSCs may represent a potential therapeutic option for fibrosis resolution owing to their unique pleiotropic functions and therapeutic properties. Currently, clinical trial efforts using MSCs and MSC-based products are underway but clinical data collected by the early phase trials are insufficient to offer better support for the MSC-based anti-fibrotic therapies. Given that MSCs are involved in the coagulation through releasing tissue factor, MSCs can retain procoagulant activity to be associated with fibrogenic disease development. Therefore, MSCs’ functional benefits in translational applications need to be carefully balanced with their potential risks.
Collapse
Affiliation(s)
- Chenghai Li
- Stem Cell Program of Clinical Research Center, People’s Hospital of Zhengzhou University and Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan University, Zhengzhou, China
- *Correspondence: Chenghai Li, ; Bin Wang,
| | - Bin Wang
- Department of Neurosurgery, People’s Hospital of Zhengzhou University and Henan Provincial People’s Hospital, Zhengzhou, China
- *Correspondence: Chenghai Li, ; Bin Wang,
| |
Collapse
|
14
|
Liu Z, Zhou S, Zhang Y, Zhao M. Rat bone marrow mesenchymal stem cells (BMSCs) inhibit liver fibrosis by activating GSK3β and inhibiting the Wnt3a/β-catenin pathway. Infect Agent Cancer 2022; 17:17. [PMID: 35440002 PMCID: PMC9017036 DOI: 10.1186/s13027-022-00432-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/11/2022] [Indexed: 11/20/2022] Open
Abstract
Background Bone marrow mesenchymal stem cells (BMSCs) can effectively alleviate liver fibrosis, which is a pathological injury caused by various chronic liver diseases. This study aimed to investigate the antifibrotic effects of BMSCs and elucidate the underlying mechanism by which BMSCs affect liver fibrosis in vitro and in vivo. Methods After the rat liver fibrosis model was induced by continuous injection of carbon tetrachloride (CCl4), BMSCs were administered for 4 weeks, and histopathological analysis and liver function tests were performed. T6 hepatic stellate cells (HSC-T6 cells) were stimulated by TGF-β1, and the activation and proliferation of cells were analyzed by CCK-8 assays, flow cytometry, real-time PCR, western blotting and enzyme-linked immunosorbent assay (ELISA). Results Our data demonstrated that BMSCs effectively reduced the accumulation of collagen, enhanced liver functionality and ameliorated liver fibrosis in vivo. BMSCs increased the sub-G1 population in HSC-T6 cells. In addition, coculture with BMSCs reduced the expression of α-SMA, collagen I, cyclin-D1, and c-Myc in HSC-T6 cells and activated the phosphorylation of GSK3β. The GSK3β inhibitor SB216763 reversed the effect of BMSCs. The Wnt/β-catenin signalling pathway was involved in BMSC-mediated inhibition of HSC-T6 cell activation. Conclusions Our data suggested that BMSCs exerted antifibrotic effects by activating the expression of GSK3β and inhibiting the Wnt3a/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Zhaoguo Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.,Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, China
| | - Song Zhou
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ya Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ming Zhao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China. .,Department of Organ Transplantation, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, Guangdong Province, China.
| |
Collapse
|
15
|
Li Y, Shen Z, Jiang X, Wang Y, Yang Z, Mao Y, Wu Z, Li G, Chen H. Mouse mesenchymal stem cell-derived exosomal miR-466f-3p reverses EMT process through inhibiting AKT/GSK3β pathway via c-MET in radiation-induced lung injury. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:128. [PMID: 35392967 PMCID: PMC8988379 DOI: 10.1186/s13046-022-02351-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/29/2022] [Indexed: 01/06/2023]
Abstract
Background Radiation-induced lung fibrosis (RILF) is a common complication of thoracic radiotherapy. Alveolar epithelial cells play a crucial role in lung fibrosis via epithelial-mesenchymal transition (EMT). Exosomes derived from mesenchymal stem cells own the beneficial properties to repair and regeneration of damaged tissues, however the underlying mechanisms remain poorly understood. Methods Mouse mesenchymal stem cells-derived exosomes (mMSCs-Exo) were isolated by differential centrifugation, and their protective effects were assessed in vivo and in vitro, respectively. EMT-associated proteins were measured via western blot assay and/or immunofluorescence staining. The miRNA expression was measured by microarray assay and qPCR. Furthermore, bioinformatics prediction with KEGG analysis, luciferase assay, and rescue experiments were performed to explore the molecular mechanism underlying miR-466f-3p. Results mMSCs-Exos were efficiently isolated ranging from 90-150 nm with high expression of exosomal markers (CD63, TSG101, and CD9). mMSCs-Exos administration efficiently relieved radiation-induced lung injury with less collagen deposition and lower levels of IL-1β and IL-6. Meanwhile, in vitro results showed mMSCs-Exos treatment obviously reversed EMT process induced by radiation. Among enriched miRNA cargo in exosomes, miR-466f-3p was primarily responsible for the protective effects via inhibition of AKT/GSK3β pathway. Our mechanistic study further demonstrated that c-MET was the direct target of miR-466f-3p, whose restoration partially abrogated mMSCs-Exo-mediated inhibition in both EMT process and AKT/GSK3β signaling activity induced by radiation. Conclusions Our findings indicated that exosomal miR-466f-3p derived from mMSCs may possess anti-fibrotic properties and prevent radiation-induced EMT through inhibition of AKT/GSK3β via c-MET, providing a promising therapeutic modality for radiation-induced lung fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02351-z.
Collapse
Affiliation(s)
- Yi Li
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, 212 Daguan Road, Kunming, 650032, China.
| | - Zhufu Shen
- Department of Geriatrics, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xiao Jiang
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, 212 Daguan Road, Kunming, 650032, China
| | - Yuanyuan Wang
- Department of Pathology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, 650032, China
| | - Zuozhang Yang
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Yuchi Mao
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, 212 Daguan Road, Kunming, 650032, China
| | - Zhixian Wu
- Department of Hepatobiliary Disease, 900th Hospital of Joint Logistics Support Force, Fuzhou, 354200, China
| | - Gaofeng Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China.
| | - Hong Chen
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, 212 Daguan Road, Kunming, 650032, China.
| |
Collapse
|
16
|
Chen S, Han B, Geng X, Li P, Lavin MF, Yeo AJ, Li C, Sun J, Peng C, Shao H, Du Z. Microcrystalline silica particles induce inflammatory response via pyroptosis in primary human respiratory epithelial cells. ENVIRONMENTAL TOXICOLOGY 2022; 37:385-400. [PMID: 34766707 DOI: 10.1002/tox.23405] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 06/13/2023]
Abstract
The mechanism of the sterile inflammatory response in the respiratory tract induced by exposure to sterile particles has not been fully elucidated. The aim of our study is to explore the earlier events in initiating inflammatory response at molecular and cellular level in primary cultured human airway epithelial cells (AEC) exposed to silica particles in order to provide information for earlier diagnosis and prevention of silica particle-induced toxicity as well as possible information on the genesis of silicosis. We isolated primary AEC from three healthy adults and treated them with silica particles at different concentrations for 48 h. We found evidence for silica-induced inflammasome activation by the co-localization of Caspase-1 and NLRP3, as well as increased levels of IL-1β and IL-18. Lactate dehydrogenase and NucGreen analysis proved the occurrence of pyroptosis. High throughput mRNA sequencing showed that the inflammatory response and NF-κB signaling pathways were significantly enriched in gene ontology and Kyoto encyclopedia of genes and genomes analysis, and pyroptosis-related genes were up-regulated. The miR-455-3p and five lncRNAs (LOC105375913, NEAT1, LOC105375181, LOC100506098, and LOC105369370) were verified as key factors related to the mechanism by ceRNA network analysis. LOC105375913 was first discovered to be associated with inflammation in AEC. These data suggest that microcrystalline silica can induce significant inflammation and pyroptosis in human primary AEC through NLRP3 inflammasome pathway and NF-κB signaling pathway at both the gene and protein levels, and the possible mechanism could be miR-455-3p mediated ceRNA hypothesis. Our data provide a method for the studies of the respiratory toxicity of fine particulate matter and the pathogenesis of early silicosis. The miR-455-3p and five lncRNAs related ceRNA network might be the toxicity mechanism of microcrystalline silica particles to AEC.
Collapse
Affiliation(s)
- Shangya Chen
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Basic Research & International Cooperation, Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
| | - Bing Han
- Department of Head and Neck Surgery, Affiliated Hospital of Shandong Academy of Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiao Geng
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Peng Li
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Martin F Lavin
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- The University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Australia
| | - Abrey J Yeo
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- The University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Australia
| | - Chao Li
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jiayin Sun
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Cheng Peng
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, Australia
| | - Hua Shao
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhongjun Du
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
17
|
Zhang X, Ye L, Tang W, Ji Y, Zheng L, Chen Y, Ge Q, Huang C. Wnt/β-Catenin Participates in the Repair of Acute Respiratory Distress Syndrome-Associated Early Pulmonary Fibrosis via Mesenchymal Stem Cell Microvesicles. Drug Des Devel Ther 2022; 16:237-247. [PMID: 35082486 PMCID: PMC8784273 DOI: 10.2147/dddt.s344309] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/12/2022] [Indexed: 12/11/2022] Open
Abstract
Purpose The main aim of the present study was to establish whether mesenchymal stem cell microvesicles (MSC MVs) exert anti-fibrotic effects and investigate the mechanisms underlying these effects in a mouse model of acute respiratory distress syndrome (ARDS)-associated early pulmonary fibrosis. Methods An ARDS-associated pulmonary fibrosis model was established in mice by an intratracheal injection of lipopolysaccharide (LPS). At 1, 3, and 7 days after LPS-mediated injury, the lungs of mice treated with MSC MVs and untreated controls were carefully excised and fibrosis was assessed based on the extent of collagen deposition. In addition, the development of epithelial–mesenchymal transition (EMT) was evaluated based on loss of E-cadherin and zona occludens-1 (ZO-1) along with the acquisition of α-smooth muscle actin (α-SMA) and N-cadherin. Nuclear translocation and β-catenin expression analyses were also used to evaluate activation of the Wnt/β-catenin signaling pathway. Results Blue-stained collagen fibers were evident as early as 7 days after LPS injection. Treatment with MSC MVs suppressed pathological progression to a significant extent. MSC MVs markedly reversed the upregulation of N-cadherin and α-SMA and attenuated the downregulation of E-cadherin and ZO-1. The expression and nuclear translocation of β-catenin were clearly decreased on day 7 after MSC MV treatment. Conclusion Analyses indicated that MSC MVs could ameliorate ARDS-associated early pulmonary fibrosis via the suppression of EMT and might be related to Wnt/β-catenin transition signaling.
Collapse
Affiliation(s)
- Xingcai Zhang
- Department of Anesthesiology, Ningbo City First Hospital, Ningbo, Zhejiang, People’s Republic of China
| | - Lifang Ye
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Wan Tang
- Department of Anesthesiology, Ningbo City First Hospital, Ningbo, Zhejiang, People’s Republic of China
| | - Yiqin Ji
- Department of Anesthesiology, Ningbo City First Hospital, Ningbo, Zhejiang, People’s Republic of China
| | - Li Zheng
- Department of Anesthesiology, Ningbo City First Hospital, Ningbo, Zhejiang, People’s Republic of China
| | - Yijun Chen
- Department of Anesthesiology, Ningbo City First Hospital, Ningbo, Zhejiang, People’s Republic of China
| | - Qidong Ge
- Department of Breast Surgery, HuaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, People’s Republic of China
| | - Changshun Huang
- Department of Anesthesiology, Ningbo City First Hospital, Ningbo, Zhejiang, People’s Republic of China
- Correspondence: Changshun Huang; Qidong Ge, Tel +86-574-87085521, Fax +86-574-87085588, Email ;
| |
Collapse
|
18
|
Liu Y, Tong Z, Wang C, Xia R, Li H, Yu H, Jing J, Cheng W. TiO2 nanotubes regulate histone acetylation through F-actin to induce the osteogenic differentiation of BMSCs. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2021; 49:398-406. [PMID: 33914666 DOI: 10.1080/21691401.2021.1910282] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/21/2021] [Indexed: 12/30/2022]
Abstract
Bone integration on the surface of titanium prosthesis is critical to the success of implant surgery. Good Bone integration at the contact interface is the basis of long-term stability. TiO2 nanotubes have become one of the most commonly used modification techniques for artificial joint prostheses and bone defect implants due to their good biocompatibility, mechanical properties and chemical stability. TiO2 nanotubes can promote F-actin polymerization in bone mesenchymal stem cells (BMSCs) and osteogenic differentiation. The possibility of F-actin as an upstream part to regulate GCN5 initiation of osteogenesis was discussed. The results of gene loss and functional acquisition assay, immunoblotting assay and fluorescence staining assay showed that TiO2 nanotubes could promote the differentiation of BMSCs into osteoblasts. The intervention of TiO2 nanotubes can make BMSCs form stronger F-actin fibre bundles, which can drive the differentiation process of osteogenesis. Our results showed that F-actin mediated nanotube-induced cell differentiation through promoting the expression of GCN5 and enhancing the function of GCN5 and GCN5 was a key regulator of the osteogenic differentiation of BMSCs induced by TiO2 nanotubes as a downstream mediated osteogenesis of F-actin, providing a novel insight into the study of osteogenic differentiation on surface of TiO2 nanotubes.
Collapse
Affiliation(s)
- Yanchang Liu
- Department of Orthopaedic Surgery, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhicheng Tong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chen Wang
- Department of Orthopaedic Surgery, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Runzhi Xia
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Huiwu Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Haoran Yu
- Department of Orthopaedic Surgery, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Juehua Jing
- Department of Orthopaedic Surgery, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wendan Cheng
- Department of Orthopaedic Surgery, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
19
|
Li B, Mu M, Sun Q, Cao H, Liu Q, Liu J, Zhang J, Xu K, Hu D, Tao X, Wang J. A suitable silicosis mouse model was constructed by repeated inhalation of silica dust via nose. Toxicol Lett 2021; 353:1-12. [PMID: 34626813 DOI: 10.1016/j.toxlet.2021.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/14/2021] [Accepted: 09/30/2021] [Indexed: 11/18/2022]
Abstract
Silicosis as the serious occupational disease is highly necessary to construct a suitable mouse model for disclosing mechanism of occurrence and development in this disease. Here, the volume-effect relationship and volume-based survival curves in mice who inhaled silica suspension intranasally were analyzed. Notable, the optimal volume 80 μl repeated-inhalation by nose to silica suspension in the inbred mouse C57BL/6 J with the highest susceptibility to silicosis led to a great entrance into the lung and a high survival rate after instillation. After repeated-exposure to 20 mg/mL, 80 μl silica for 16 days and then fed without silica exposure until 31 days, weight of mice showed a trend of first decrease and then recover. Moreover, the degree of pulmonary inflammation and fibrosis in mice were analyzed by pathological and immunohistochemistry staining. Transforming growth factor-beta (TGF-β), smooth muscle alpha-actin (α-SMA) and collagen type-I (collagen I, Col-I) were significantly increased in the silica-exposed mouse lung at post-exposure day 16 compared with the controls. Sirius red stain and Micro-CT analysis showed that lung fibrosis formed at post-exposure day 31. This study highlights the critical importance of perfusion volume and repeated nasal drops in inducing inflammatory response and pulmonary fibrosis in silicosis.
Collapse
Affiliation(s)
- Bing Li
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China.
| | - Min Mu
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety, Anhui University of Science and Technology of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China.
| | - Qixian Sun
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Hangbing Cao
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Qiang Liu
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Jiaxin Liu
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China
| | - Jinfeng Zhang
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety, Anhui University of Science and Technology of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China
| | - Keyi Xu
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety, Anhui University of Science and Technology of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China
| | - Dong Hu
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety, Anhui University of Science and Technology of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China
| | - Xinrong Tao
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety, Anhui University of Science and Technology of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China.
| | - Jianhua Wang
- Key Laboratory of Industrial Dust Control and Occupational Health of the Ministry of Education, Anhui University of Science and Technology, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety, Anhui University of Science and Technology of Anhui Higher Education Institutes, Anhui University of Science and Technology, China; School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, Anhui University of Science and Technology, China; Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.
| |
Collapse
|
20
|
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: 0.7] [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
|
21
|
Li DY, Li RF, Sun DX, Pu DD, Zhang YH. Mesenchymal stem cell therapy in pulmonary fibrosis: a meta-analysis of preclinical studies. Stem Cell Res Ther 2021; 12:461. [PMID: 34407861 PMCID: PMC8371890 DOI: 10.1186/s13287-021-02496-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a devastating disease characterized by remodeling of lung architecture and abnormal deposition of fibroblasts in parenchymal tissue and ultimately results in respiratory failure and death. Preclinical studies suggest that mesenchymal stem cell (MSC) administration may be a safe and promising option in treating PF. The objective of our meta-analysis is to assess the efficacy of MSC therapy in preclinical models of PF. METHODS We performed a comprehensive literature search in PubMed, EMBASE, Web of Science, and Cochrane Library databases from inception to March 17, 2021. Studies that assessed the efficacy of MSC therapy to animals with PF were included. The SYRCLE bias risk tool was employed to evaluate the bias of included studies. The primary outcomes included survival rate and pulmonary fibrosis scores. Meta-analysis was conducted via Cochrane Collaboration Review Manager (version 5.4) and Stata 14.0 statistical software. RESULTS A total of 1120 articles were reviewed, of which 24 articles met inclusion criteria. Of these, 12 studies evaluated the survival rate and 20 studies evaluated pulmonary fibrosis scores. Compared to the control group, MSC therapy was associated with an improvement in survival rate (odds ratios (OR) 3.10, 95% confidence interval (CI) 2.06 to 4.67, P < 0.001, I2 = 0%) and a significant reduction in pulmonary fibrosis scores (weighted mean difference (WMD) 2.05, 95% CI -2.58 to -1.51, P < 0.001, I2 = 90%). CONCLUSIONS MSC therapy is a safe and effective method that can significantly improve the survival and pulmonary fibrosis of PF animals. These results provide an important basis for future translational clinical studies.
Collapse
Affiliation(s)
- Deng-Yuan Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People's Republic of China.,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People's Republic of China.,Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, Kunming, 650022, People's Republic of China
| | - Ru-Fang Li
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People's Republic of China.,Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, Kunming, 650022, People's Republic of China
| | - Dan-Xiong Sun
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People's Republic of China.,Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, Kunming, 650022, People's Republic of China
| | - Dan-Dan Pu
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People's Republic of China.,Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, Kunming, 650022, People's Republic of China
| | - Yun-Hui Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People's Republic of China. .,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650500, People's Republic of China. .,Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, Kunming, 650022, People's Republic of China.
| |
Collapse
|
22
|
Zhang E, Geng X, Shan S, Li P, Li S, Li W, Yu M, Peng C, Wang S, Shao H, Du Z. Exosomes derived from bone marrow mesenchymal stem cells reverse epithelial-mesenchymal transition potentially via attenuating Wnt/β-catenin signaling to alleviate silica-induced pulmonary fibrosis. Toxicol Mech Methods 2021; 31:655-666. [PMID: 34225584 DOI: 10.1080/15376516.2021.1950250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pulmonary fibrosis induced by silica dust is an irreversible, chronic, and fibroproliferative lung disease with no effective treatment at present. BMSCs-derived exosomes (BMSCs-Exo) possess similar functions to their parent cells. In this study, we investigated the therapeutic potential and underlying molecular mechanism for BMSCs-Exo in the treatment of silica-induced pulmonary fibrosis. The rat model of experimental silicosis pulmonary fibrosis was induced with 1.0 mL of one-off infusing silica suspension using the non-exposed intratracheal instillation (50 mg/mL/rat). In vivo transplantation of BMSCs-Exo effectively alleviated silica-induced pulmonary fibrosis, including a reduction in collagen accumulation, inhibition of TGF-β1, and decreased HYP content. Treatment of BMSCs-Exo increased the expression of epithelial marker proteins including E-cadherin (E-cad) and cytokeratin19 (CK19) and reduced the expression of fibrosis marker proteins including α-Smooth muscle actin (α-SMA) after exposure to silica suspension. Furthermore, we found that BMSCs-Exo inhibited the expression of Wnt/β-catenin pathway components (P-GSK3β, β-catenin, Cyclin D1) in pulmonary fibrosis tissue. BMSCs-Exo is involved in the alleviation of silica-induced pulmonary fibrosis by reducing the level of profibrotic factor TGF-β1 and inhibiting the progression of epithelial-mesenchymal transition (EMT). Additionally, attenuation of the Wnt/β-catenin signaling pathway closely related to EMT may be one of the mechanisms involved in anti-fibrotic effects of exosomes.
Collapse
Affiliation(s)
- Enguo Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China.,Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Xiao Geng
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Shan Shan
- School of Public Health, Shandong University, Jinan, People's Republic of China
| | - Peng Li
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Shumin Li
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Wentao Li
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Meili Yu
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Cheng Peng
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China.,Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, Australia
| | - Shijun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Hua Shao
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Zhongjun Du
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| |
Collapse
|
23
|
The Mechanism and Effect of Autophagy, Apoptosis, and Pyroptosis on the Progression of Silicosis. Int J Mol Sci 2021; 22:ijms22158110. [PMID: 34360876 PMCID: PMC8348676 DOI: 10.3390/ijms22158110] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Silicosis remains one of the most severe pulmonary fibrotic diseases worldwide, caused by chronic exposure to silica dust. In this review, we have proposed that programmed cell death (PCD), including autophagy, apoptosis, and pyroptosis, is closely associated with silicosis progression. Furthermore, some autophagy, apoptosis, or pyroptosis-related signaling pathways or regulatory proteins have also been summarized to contribute greatly to the formation and development of silicosis. In addition, silicosis pathogenesis depends on the crosstalk among these three ways of PCD to a certain extent. In summary, more profound research on these mechanisms and effects may be expected to become promising targets for intervention or therapeutic methods of silicosis in the future.
Collapse
|
24
|
Pang J, Luo Y, Wei D, Cao Z, Qi X, Song M, Liu Y, Li Z, Zhang J, Li B, Chen J, Wang J, Wang C. Comparative Transcriptome Analyses Reveal a Transcriptional Landscape of Human Silicosis Lungs and Provide Potential Strategies for Silicosis Treatment. Front Genet 2021; 12:652901. [PMID: 34149803 PMCID: PMC8210851 DOI: 10.3389/fgene.2021.652901] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
Silicosis is a fatal occupational lung disease which currently has no effective clinical cure. Recent studies examining the underlying mechanism of silicosis have primarily examined experimental models, which may not perfectly reflect the nature of human silicosis progression. A comprehensive profiling of the molecular changes in human silicosis lungs is urgently needed. Here, we conducted RNA sequencing (RNA-seq) on the lung tissues of 10 silicosis patients and 7 non-diseased donors. A total of 2,605 differentially expressed genes (DEGs) and critical pathway changes were identified in human silicosis lungs. Further, the DEGs in silicosis were compared with those in idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary diseases (COPD), to extend current knowledge about the disease mechanisms and develop potential drugs. This analysis revealed both common and specific regulations in silicosis, along with several critical genes (e.g., MUC5AC and FGF10), which are potential drug targets for silicosis treatment. Drugs including Plerixafor and Retinoic acid were predicted as potential candidates in treating silicosis. Overall, this study provides the first transcriptomic fingerprint of human silicosis lungs. The comparative transcriptome analyses comprehensively characterize pathological regulations resulting from silicosis, and provide valuable cues for silicosis treatment.
Collapse
Affiliation(s)
- Junling Pang
- State Key Laboratory of Medical Molecular Biology, Department of Pathophysiology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Ya Luo
- State Key Laboratory of Medical Molecular Biology, Department of Pathophysiology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Dong Wei
- Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Zhujie Cao
- State Key Laboratory of Medical Molecular Biology, Department of Pathophysiology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Xianmei Qi
- State Key Laboratory of Medical Molecular Biology, Department of Pathophysiology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Meiyue Song
- Department of Pulmonary and Critical Care Medicine/Others, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Ying Liu
- State Key Laboratory of Medical Molecular Biology, Department of Pathophysiology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhaoguo Li
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jin Zhang
- Department of Thoracic Surgery and Lung Transplantation, China-Japan Friendship Hospital, Beijing, China
| | - Baicun Li
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyu Chen
- Transplant Center, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China.,Department of Thoracic Surgery and Lung Transplantation, China-Japan Friendship Hospital, Beijing, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Department of Pathophysiology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Chen Wang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Qin X, Lin X, Liu L, Li Y, Li X, Deng Z, Chen H, Chen H, Niu Z, Li Z, Hu Y. Macrophage-derived exosomes mediate silica-induced pulmonary fibrosis by activating fibroblast in an endoplasmic reticulum stress-dependent manner. J Cell Mol Med 2021; 25:4466-4477. [PMID: 33834616 PMCID: PMC8093963 DOI: 10.1111/jcmm.16524] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/22/2021] [Accepted: 03/19/2021] [Indexed: 12/11/2022] Open
Abstract
Macrophages play a key role in silicosis, and exosomes are potent mediators of intercellular communication. This suggests that macrophage-derived exosomes have a potential contribution to the pathogenesis of silicosis. To investigate whether macrophage-derived exosomes promote or inhibit lung fibrosis, in vitro, silica-exposed macrophage-derived exosomes (SiO2 -Exos) were collected and cocultured with fibroblasts. The expression of collagen I and α-SMA was evaluated. Furthermore, the endoplasmic reticulum (ER) stress markers BIP, XBP1s and P-eIF2α were assessed after treatment with or without the ER stress inhibitor 4-PBA. In vivo, mice were pre-treated with the exosome secretion inhibitor GW4869 prior to silica exposure. After sacrifice, lung tissues were histologically examined, and the expression of proinflammatory cytokines (TNF-α, IL-1β and IL-6) in bronchoalveolar lavage fluid (BALF) was measured. The results showed that the expression of collagen I and α-SMA was up-regulated after treatment with SiO2 -Exos, accompanied by increased expression of BIP, XBP1s and P-eIF2α. Pre-treatment with 4-PBA reversed this effect. More importantly, an in vivo study demonstrated that pre-treatment with GW4869 decreased lung fibrosis and the expression of TNF-α, IL-1β and IL-6 in BALF. These results suggested that SiO2 -Exos are profibrogenic and that the facilitating effect is dependent on ER stress.
Collapse
Affiliation(s)
- Xiaofeng Qin
- Department of PathologySchool of Basic Medical ScienceCentral South UniversityChangshaChina
| | - Xiaofang Lin
- Department of PathologySchool of Basic Medical ScienceCentral South UniversityChangshaChina
| | - Lang Liu
- Department of Occupational DiseasesHunan Prevention and Treatment Institute for Occupational DiseasesChangshaChina
| | - Ying Li
- Department of Occupational DiseasesHunan Prevention and Treatment Institute for Occupational DiseasesChangshaChina
| | - Xiang Li
- Department of PathologySchool of Basic Medical ScienceCentral South UniversityChangshaChina
- Department of PathologyXiangya HospitalCentral South UniversityChangshaChina
| | - Zhenghao Deng
- Department of PathologySchool of Basic Medical ScienceCentral South UniversityChangshaChina
- Department of PathologyXiangya HospitalCentral South UniversityChangshaChina
| | - Huiping Chen
- Department of PathologyXiangya HospitalCentral South UniversityChangshaChina
| | - Hui Chen
- Department of PathologySchool of Basic Medical ScienceCentral South UniversityChangshaChina
| | - Zhiyuan Niu
- Department of PathologySchool of Basic Medical ScienceCentral South UniversityChangshaChina
| | - Zisheng Li
- Department of PathologyXiangya HospitalCentral South UniversityChangshaChina
| | - Yongbin Hu
- Department of PathologySchool of Basic Medical ScienceCentral South UniversityChangshaChina
- Department of PathologyXiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
26
|
Adamcakova J, Mokra D. New Insights into Pathomechanisms and Treatment Possibilities for Lung Silicosis. Int J Mol Sci 2021; 22:ijms22084162. [PMID: 33920534 PMCID: PMC8072896 DOI: 10.3390/ijms22084162] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Inhalation of silica particles is an environmental and occupational cause of silicosis, a type of pneumoconiosis. Development of the lung silicosis is a unique process in which the vicious cycle of ingestion of inhaled silica particles by alveolar macrophages and their release triggers inflammation, generation of nodular lesions, and irreversible fibrosis. The pathophysiology of silicosis is complex, and interactions between the pathomechanisms have not been completely understood. However, elucidation of silica-induced inflammation cascades and inflammation-fibrosis relations has uncovered several novel possibilities of therapeutic targeting. This article reviews new information on the pathophysiology of silicosis and points out several promising treatment approaches targeting silicosis-related pathways.
Collapse
|
27
|
Zhao Q, Hao C, Wei J, Huang R, Li C, Yao W. Bone marrow-derived mesenchymal stem cells attenuate silica-induced pulmonary fibrosis by inhibiting apoptosis and pyroptosis but not autophagy in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 216:112181. [PMID: 33848736 DOI: 10.1016/j.ecoenv.2021.112181] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 06/12/2023]
Abstract
This study aimed to investigate the effect of bone marrow-derived mesenchymal stem cells (BMSCs) on silica-induced lung fibrosis in a rat model. Thirty SD rats were randomly divided into three groups: control group, silica group, and BMSC group (n = 10 rats per group). BMSCs were injected successively into rats on the 14th, 28th, and 42nd days after silica exposure. All rats were sacrificed 56 days after silica exposure. We detected the pathological and fibrotic changes, apoptosis, autophagy, and pyroptosis in their lung tissue by histopathological examination, hydroxyproline content assays, real-time quantitative polymerase chain reactions, western blot assays, immunohistochemistry staining, immunofluorescence staining, and enzyme-linked immunosorbent assays. We found that BMSCs significantly relieved lung inflammatory infiltrates, collagen deposition, hydroxyproline content, and the mRNA and protein levels of collagen 1 and fibronectin. Compared to the silica group, in the BMSC group, apoptosis-associated proteins, including cleaved caspase 3 and Bax, were significantly downregulated, and Bcl-2/Bax was significantly upregulated; pyroptosis-related proteins, including Nlrp3, cleaved caspase 1, IL-1β, and IL-18, were significantly reduced. However, the BMSCs had no significant impact on autophagy-related proteins, including Beclin 1, P62, and LC3. In summary, BMSCs protected lung tissue against severe fibrosis by inhibiting apoptosis and pyroptosis but not autophagy.
Collapse
Affiliation(s)
- Qiuyan Zhao
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Changfu Hao
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Jingjing Wei
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Ruoxuan Huang
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Chao Li
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Wu Yao
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China.
| |
Collapse
|
28
|
Yang S, Liu P, Jiang Y, Wang Z, Dai H, Wang C. Therapeutic Applications of Mesenchymal Stem Cells in Idiopathic Pulmonary Fibrosis. Front Cell Dev Biol 2021; 9:639657. [PMID: 33768094 PMCID: PMC7985078 DOI: 10.3389/fcell.2021.639657] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/15/2021] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial disease of unknown etiology characterized by progressive pulmonary fibrosis. Pirfenidone and nintedanib are the only drugs that can prolong the time to disease progression, slow down the decline in lung function, and prolong survival. However, they do not offer a cure and are associated with tolerability issues. The pluripotency of mesenchymal stem cells (MSCs) and their ability to regulate immunity, inhibit inflammation, and promote epithelial tissue repair highlight the promise of MSC therapy for treating interstitial lung disease. However, optimal protocols are lacking for multi-parameter selection in MSC therapy. This review summarizes preclinical studies on MSC transplantation for the treatment of interstitial lung disease and clinical studies with known results. An analysis of relevant factors for the optimization of treatment plans is presented, including MSCs with different sources, administration routes and timing, dosages, frequencies, and pretreatments with MSCs. This review proposes an optimized plan for guiding the design of future clinical research to identify therapeutic options for this complex disease.
Collapse
Affiliation(s)
- Shengnan Yang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China.,WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, Beijing, China.,Harbin Medical University, Harbin, China
| | - Peipei Liu
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yale Jiang
- School of Medicine, Tsinghua University, Beijing, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China.,WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, Beijing, China
| | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China.,WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, Beijing, China.,Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
29
|
Huang J, Huang J, Ning X, Luo W, Chen M, Wang Z, Zhang W, Zhang Z, Chao J. CT/NIRF dual-modal imaging tracking and therapeutic efficacy of transplanted mesenchymal stem cells labeled with Au nanoparticles in silica-induced pulmonary fibrosis. J Mater Chem B 2021; 8:1713-1727. [PMID: 32022096 DOI: 10.1039/c9tb02652e] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mesenchymal stem cells (MSCs) have shown promising therapeutic effects in cell-based therapies and regenerative medicine. Efficient tracking of MSCs is an urgent clinical need that will help us to understand their behavior after transplantation and allow adjustment of therapeutic strategies. However, no clinically approved tracers are currently available, which limits the clinical translation of stem cell therapy. In this study, a nanoparticle (NP) for computed tomography (CT)/fluorescence dual-modal imaging, Au@Albumin@ICG@PLL (AA@ICG@PLL), was developed to track bone marrow-derived mesenchymal stem cells (BMSCs) that were administered intratracheally into mice with silica-induced pulmonary fibrosis, which facilitated understanding of the therapeutic effect and the possible molecular mechanism of stem cell therapy. The AuNPs were first formed in bovine serum albumin (BSA) solution and modified with indocyanine green (ICG), and subsequently coated with a poly-l-lysine (PLL) layer to enhance intracellular uptake and biocompatibility. BMSCs were labeled with AA@ICG@PLL NPs with high efficiency without an effect on biological function or therapeutic capacity. The injected AA@ICG@PLL-labeled BMSCs could be tracked via CT and near-infrared fluorescence (NIRF) imaging for up to 21 days after transplantation. Using these NPs, the molecular anti-inflammatory mechanism of transplanted BMSCs was revealed, which included the downregulation of proinflammatory cytokines, suppression of macrophage activation, and delay of the fibrosis process. This study suggests a promising role for imaging-guided MSC-based therapy for pulmonary fibrosis, such as idiopathic pulmonary fibrosis (IPF) and pneumoconiosis.
Collapse
Affiliation(s)
- Jie Huang
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Jie Huang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, China.
| | - Xinyu Ning
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, China.
| | - Wei Luo
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China. and Department of Respiration, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China and Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Mengling Chen
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China. and Department of Respiration, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China and Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhangyan Wang
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China. and Department of Respiration, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China and Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Wei Zhang
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China. and Department of Respiration, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China and Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhijun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, China.
| | - Jie Chao
- Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China. and Department of Respiration, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China and Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China and School of Medicine, Xizang Minzu University, Xianyang, Shanxi 712082, China
| |
Collapse
|
30
|
Huang B, Feng Z, Zhu L, Zhang S, Duan J, Zhao C, Zhang X. Silencing of MicroRNA-503 in Rat Mesenchymal Stem Cells Exerts Potent Antitumorigenic Effects in Lung Cancer Cells. Onco Targets Ther 2021; 14:67-81. [PMID: 33442267 PMCID: PMC7797339 DOI: 10.2147/ott.s282322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/16/2020] [Indexed: 12/20/2022] Open
Abstract
Purpose Mesenchymal stem cells (MSCs) are largely studied for their potential clinical use. Recently, there has been gained further interest in the relationship between MSCs and tumorigenesis. MSCs are reported to both promote and abrogate tumor growth. The present study was designed to investigate whether miRNAs are involved in the interactions between MSCs and tumor cells in the tumor microenvironment. Materials and Methods Rat bone marrow-derived MSCs (rMSCs) were cultured with or without tumor-conditioned medium (TCM) to observe the effect upon MSCs by TCM. Microarrays and real-time PCR were performed between the two groups. A series of experiments were used to reveal the functional significance of microRNA-503 (miR-503) in rMSCs. Furthermore, the antitumorigenic effect of silencing of miR-503 in rMSCs (miR-503-i-rMSCs) in vivo was measured. Results We found that rMSCs in vitro exhibited tumor-promoting properties in TCM, and the microRNA profiles of rMSCs were significantly altered in TCM. However, miR-503-i-rMSCs can decrease the angiogenesis and growth of A549 cells. We also demonstrated in an in vivo tumor model that miR-503-i-rMSCs inhibited A549 tumor angiogenesis and significantly abrogated tumor initiation and growth. CD133 assays in peripheral blood and A549 xenografts further validated that miR-503-i-rMSCs, rather than rMSCs, exerted an antitumorigenic action in the A549 tumor model. Conclusion Our results suggest that miR-503-i-rMSCs are capable of tumor suppression. Further studies are required to develop clinical therapies based on the inhibition of the tumor-promoting properties and potentiation of the anti-tumor properties of MSCs.
Collapse
Affiliation(s)
- Bo Huang
- Public Health, Guilin Medical University, Guilin 541100, People's Republic of China
| | - Zhichun Feng
- Affiliated BaYi Children's Hospital, Seventh Medical Center of PLA General Hospital, Beijing 100700, People's Republic of China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing 100700, People's Republic of China
| | - Lina Zhu
- Affiliated BaYi Children's Hospital, Seventh Medical Center of PLA General Hospital, Beijing 100700, People's Republic of China
| | - Sheng Zhang
- Affiliated BaYi Children's Hospital, Seventh Medical Center of PLA General Hospital, Beijing 100700, People's Republic of China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing 100700, People's Republic of China
| | - Jun Duan
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, People's Republic of China
| | - Chaochao Zhao
- Public Health, Guilin Medical University, Guilin 541100, People's Republic of China
| | - Xiaoying Zhang
- Public Health, Guilin Medical University, Guilin 541100, People's Republic of China.,Affiliated BaYi Children's Hospital, Seventh Medical Center of PLA General Hospital, Beijing 100700, People's Republic of China
| |
Collapse
|
31
|
Ellison-Hughes GM, Colley L, O'Brien KA, Roberts KA, Agbaedeng TA, Ross MD. The Role of MSC Therapy in Attenuating the Damaging Effects of the Cytokine Storm Induced by COVID-19 on the Heart and Cardiovascular System. Front Cardiovasc Med 2020; 7:602183. [PMID: 33363221 PMCID: PMC7756089 DOI: 10.3389/fcvm.2020.602183] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/17/2020] [Indexed: 01/08/2023] Open
Abstract
The global pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes coronavirus disease 2019 (COVID-19) has led to 47 m infected cases and 1. 2 m (2.6%) deaths. A hallmark of more severe cases of SARS-CoV-2 in patients with acute respiratory distress syndrome (ARDS) appears to be a virally-induced over-activation or unregulated response of the immune system, termed a "cytokine storm," featuring elevated levels of pro-inflammatory cytokines such as IL-2, IL-6, IL-7, IL-22, CXCL10, and TNFα. Whilst the lungs are the primary site of infection for SARS-CoV-2, in more severe cases its effects can be detected in multiple organ systems. Indeed, many COVID-19 positive patients develop cardiovascular complications, such as myocardial injury, myocarditis, cardiac arrhythmia, and thromboembolism, which are associated with higher mortality. Drug and cell therapies targeting immunosuppression have been suggested to help combat the cytokine storm. In particular, mesenchymal stromal cells (MSCs), owing to their powerful immunomodulatory ability, have shown promise in early clinical studies to avoid, prevent or attenuate the cytokine storm. In this review, we will discuss the mechanistic underpinnings of the cytokine storm on the cardiovascular system, and how MSCs potentially attenuate the damage caused by the cytokine storm induced by COVID-19. We will also address how MSC transplantation could alleviate the long-term complications seen in some COVID-19 patients, such as improving tissue repair and regeneration.
Collapse
Affiliation(s)
- Georgina M. Ellison-Hughes
- Faculty of Life Sciences & Medicine, Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, King's College London Guy's Campus, London, United Kingdom
| | - Liam Colley
- School of Sport, Health, and Exercise Sciences, Bangor University, Bangor, United Kingdom
| | - Katie A. O'Brien
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Kirsty A. Roberts
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Thomas A. Agbaedeng
- Faculty of Health & Medical Sciences, Centre for Heart Rhythm Disorders, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Mark D. Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| |
Collapse
|
32
|
A Positive Feed Forward Loop between Wnt/ β-Catenin and NOX4 Promotes Silicon Dioxide-Induced Epithelial-Mesenchymal Transition of Lung Epithelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3404168. [PMID: 33376577 PMCID: PMC7744200 DOI: 10.1155/2020/3404168] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/08/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022]
Abstract
Silicosis is a chronic fibrotic lung disease caused by the accumulation of silica dust in the distal lung. Canonical Wnt signaling and NADPH oxidase 4 (NOX4) have been demonstrated to play a crucial role in the pathogenesis of pulmonary fibrosis including silicosis. However, the underlying mechanisms of crosstalk between these two signalings are not fully understood. In the present study, we aimed to explore the interaction of Wnt/β-catenin and NOX4 of human epithelial cells in response to an exposure of silica dust. Results demonstrated an elevated expression of key components of Wnt/β-catenin signaling and NOX4 in the lungs of silicon dioxide- (SiO2-) induced silicosis mice. Furthermore, the activated Wnt/β-catenin and NOX4 signaling are accompanied by an inhibition of cell proliferation, an increase of ROS production and cell apoptosis, and an upregulation of profibrogenic factors in BEAS-2B human lung epithelial cells exposed to SiO2. A mechanistic study further demonstrated that the Wnt3a-mediated activation of canonical Wnt signaling could augment the SiO2-induced NOX4 expression and reactive oxygen species (ROS) production but reduced glutathione (GSH), while Wnt inhibitor DKK1 exhibited an opposite effect to Wnt3a. Vice versa, an overexpression of NOX4 further activated SiO2-induced Wnt/β-catenin signaling and NFE2-related factor 2 (Nrf2) antioxidant response along with a reduction of GSH, whereas the shRNA-mediated knockdown of NOX4 showed an opposite effect to NOX4 overexpression. These results imply a positive feed forward loop between Wnt/β-catenin and NOX4 signaling that may promote epithelial-mesenchymal transition (EMT) of lung epithelial cells in response to an exposure of silica dust, which may thus provide an insight into the profibrogenic role of Wnt/β-catenin and NOX4 crosstalk in lung epithelial cell injury and pathogenesis of silicosis.
Collapse
|
33
|
Cao Z, Song M, Liu Y, Pang J, Li Z, Qi X, Shu T, Li B, Wei D, Chen J, Li B, Wang J, Wang C. A novel pathophysiological classification of silicosis models provides some new insights into the progression of the disease. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 202:110834. [PMID: 32622305 DOI: 10.1016/j.ecoenv.2020.110834] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/27/2020] [Accepted: 05/30/2020] [Indexed: 06/11/2023]
Abstract
Silicosis is caused by massive inhalation of silica-based particles, which leads to pulmonary inflammation, pulmonary fibrosis and lung dysfunction. Currently, the pathophysiological process of silicosis has not been well studied. Here, we defined the progression of silicosis as four stages by unsupervised clustering analysis: normal stage, inflammatory stage, progressive stage and fibrotic stage. Specifically, in normal stage, the lung function was normal, and no inflammation or fibrosis was detected in the lung tissue. Inflammatory stage showed a remarkable pulmonary inflammation but mild fibrosis and lung dysfunction. In progressive stage, significant lung dysfunction was observed, while pulmonary inflammation and fibrosis continued to deteriorate. Fibrotic stage revealed the most severe pulmonary fibrosis and lung dysfunction but no significant deterioration in inflammation. Since the common features were founded in both silicosis patients and rodents, we speculated that the pathophysiological processes of silicosis in patients might be similar to the rodents. Collectively, our new classification identified the process of silicosis, clarified the pathophysiological features of each stage, and provided some new insights for the progression of the disease.
Collapse
Affiliation(s)
- Zhujie Cao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Meiyue Song
- Beijing University of Chinese Medicine, Beijing, China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Ying Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Junling Pang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Zhaoguo Li
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xianmei Qi
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Ting Shu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Baicun Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Dong Wei
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Jingyu Chen
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Bolun Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China.
| | - Chen Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China.
| |
Collapse
|
34
|
Wan X, Chen S, Fang Y, Zuo W, Cui J, Xie S. Mesenchymal stem cell-derived extracellular vesicles suppress the fibroblast proliferation by downregulating FZD6 expression in fibroblasts via micrRNA-29b-3p in idiopathic pulmonary fibrosis. J Cell Physiol 2020; 235:8613-8625. [PMID: 32557673 DOI: 10.1002/jcp.29706] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/04/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF), a progressive and fatal lung disease, usually leads to an irreversible distortion of the pulmonary structure. The functional roles of bone marrow-derived mesenchymal stem cells (BMSC)-secreted extracellular vesicles (EVs) in fibroblasts have been implicated, yet their actions in the treatment of IPF are not fully understood. This study investigated the roles of BMSC-derived EVs expressing miR-29b-3p in fibroblasts in IPF treatment. EVs derived from BMSCs were successfully isolated and could be internalized by pulmonary fibroblasts, and Cell Counting Kit-8 (CCK-8) and Transwell assay results identified that EVs inhibited the activation of fibroblast in IPF. miR-29b-3p, frizzled 6 (FZD6), α-skeletal muscle actin (α-SMA), and Collagen I expressions were examined, which revealed that miR-29b-3p was poorly expressed and FZD6, α-SMA, and Collagen I were overexpressed in pulmonary tissues. Dual-luciferase reporter assay results demonstrated that miR-29b-3p could inversely target FZD6 expression. The gain- and loss-of-function assays were conducted to determine regulatory effects of FZD6 and miR-29b-3p on IPF. CCK-8 and Transwell assays results displayed that BMSCs-derived EVs overexpressing miR-29b-3p contributed to inhibited pulmonary interstitial fibroblast proliferation, migration, invasion, and differentiation. Furthermore, the effects of BMSCs-derived EVs overexpressing miR-29b-3p on IPF progression were assessed in vivo, which confirmed the repressive effects of BMSCs-derived EVs overexpressing miR-29b-3p on IPF progression. Collectively, BMSCs-derived EVs overexpressing miR-29b-3p relieve IPF through FZD6.
Collapse
Affiliation(s)
- Xuan Wan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shuyun Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan Fang
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Wei Zuo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Cui
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shiguang Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
35
|
Chen G, Wang Q, Li Z, Yang Q, Liu Y, Du Z, Zhang G, Song Y. Circular RNA CDR1as promotes adipogenic and suppresses osteogenic differentiation of BMSCs in steroid-induced osteonecrosis of the femoral head. Bone 2020; 133:115258. [PMID: 32018039 DOI: 10.1016/j.bone.2020.115258] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 12/13/2022]
Abstract
Steroid-induced osteonecrosis of the femoral head (SONFH) is a common debilitating orthopedic disease. The bone marrow mesenchymal stem cells (BMSCs) are a type of mesenchymal stem cells which play crucial roles in bone repair. The adipogenic/osteogenic differentiation disorder of BMSCs has been widely perceived contributing to SONFH. However, the regulatory mechanism of BMSCs differentiation disorder still remains unclear. Circular RNA (circRNA), a kind of stable ncRNA, plays important roles in regulating gene expression via various ways. To date, there are no studies to uncover the circRNA expression profile and screen out the key circRNAs playing crucial roles in adipogenic/osteogenic differentiation disorder of SONFH-BMSCs. In present study, we detected the circRNA expression profiles in SONFH-BMSCs for the first time. A total of 820 circRNAs were differentially expressed in SONFH-BMSCs, including 460 up- and 360 down-regulated circRNAs. Bioinformatics analysis indicates circRNA CDR1as, one up-regulated circRNA, may play crucial role in adipogenic/osteogenic differentiation disorder of SONFH-BMSCs via CDR1as-miR-7-5p-WNT5B axis. Knocking-down CDR1as resulted in increasing of osteogenic differentiation and decreasing of adipogenic differentiation of BMSCs, while over-expressing CDR1as resulted in decreasing of osteogenic differentiation and increasing of adipogenic differentiation of BMSCs. The miR-7-5p binding sites of CDR1as and WNT5B were verified by luciferase reporter gene assay. Our study may provide new insights into the molecular mechanisms of osteogenic/adipogenic differentiation disorder of SONFH-BMSCs and new biomarkers for the diagnosis and treatment of SONFH.
Collapse
Affiliation(s)
- Gaoyang Chen
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China.
| | - Qingyu Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China
| | - Zhaoyan Li
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China
| | - Qiwei Yang
- Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China.
| | - Yuzhe Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China.
| | - Zhenwu Du
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China
| | - Guizhen Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China
| | - Yang Song
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China.
| |
Collapse
|
36
|
Feng YL, Chen DQ, Vaziri ND, Guo Y, Zhao YY. Small molecule inhibitors of epithelial-mesenchymal transition for the treatment of cancer and fibrosis. Med Res Rev 2019; 40:54-78. [PMID: 31131921 DOI: 10.1002/med.21596] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/20/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Abstract
Tissue fibrosis and cancer both lead to high morbidity and mortality worldwide; thus, effective therapeutic strategies are urgently needed. Because drug resistance has been widely reported in fibrotic tissue and cancer, developing a strategy to discover novel targets for targeted drug intervention is necessary for the effective treatment of fibrosis and cancer. Although many factors lead to fibrosis and cancer, pathophysiological analysis has demonstrated that tissue fibrosis and cancer share a common process of epithelial-mesenchymal transition (EMT). EMT is associated with many mediators, including transcription factors (Snail, zinc-finger E-box-binding protein and signal transducer and activator of transcription 3), signaling pathways (transforming growth factor-β1, RAC-α serine/threonine-protein kinase, Wnt, nuclear factor-kappa B, peroxisome proliferator-activated receptor, Notch, and RAS), RNA-binding proteins (ESRP1 and ESRP2) and microRNAs. Therefore, drugs targeting EMT may be a promising therapy against both fibrosis and tumors. A large number of compounds that are synthesized or derived from natural products and their derivatives suppress the EMT by targeting these mediators in fibrosis and cancer. By targeting EMT, these compounds exhibited anticancer effects in multiple cancer types, and some of them also showed antifibrotic effects. Therefore, drugs targeting EMT not only have both antifibrotic and anticancer effects but also exert effective therapeutic effects on multiorgan fibrosis and cancer, which provides effective therapy against fibrosis and cancer. Taken together, the results highlighted in this review provide new concepts for discovering new antifibrotic and antitumor drugs.
Collapse
Affiliation(s)
- Ya-Long Feng
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Dan-Qian Chen
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Nosratola D Vaziri
- Department of Medicine, University of California Irvine, Irvine, California
| | - Yan Guo
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China.,Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Ying-Yong Zhao
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|