1
|
Sheng K, Ran Y, Feng X, Wang Y, Zhou S, Guan Y, Tan P, Qian S, Zhao Z, Zhang B, Ji W, Niu C, Guo X. PTN secreted by cardiac fibroblasts promotes myocardial fibrosis and inflammation of pressure overload-induced hypertrophic cardiomyopathy through the PTN-SDC4 pathway. Life Sci 2025; 363:123356. [PMID: 39765325 DOI: 10.1016/j.lfs.2024.123356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is characterized by unexplained left ventricular hypertrophy (LVH) with key pathologic processes including myocardial necrosis, fibrosis, inflammation, and hypertrophy, which are involved in heart failure (HF), stroke, and even sudden death. Our aim was to explore the communication network among various cells in the heart of transverse aortic constriction (TAC) surgery induced HCM mice. MATERIALS AND METHODS Single-cell RNA-seq data of GSE137167 was downloaded from the Gene Expression Omnibus (GEO) database. Seurat was used to perform the standard workflow. CellChat was utilized to compute the cell-cell interaction network and analyze the ligand-receptor pairs. Weighted gene co-expression network analysis (WGCNA) was conducted to identify gene co-expression modules. In vitro and in vivo studies were performed to verify bioinformatic analysis findings through real-time quantitative PCR (RT-qPCR), Edu staining, transwell assay, western blot, immunofluorescence assay, CCK-8, hematoxylin and eosin (H&E) staining, and echocardiography based on TAC mouse model. KEY FINDINGS Our results showed that after TAC surgery, the interaction between cardiac fibroblasts and macrophages was very common, and the increasing pleiotrophin (PTN) ligand secreted by cardiac fibroblasts could promote the self-proliferation or invasion for myocardial fibrosis as well as stimulate the inflammatory response of macrophages to contribute TAC surgery induced HCM through acting on Syndecan 4 (SDC4) receptor. SIGNIFICANCE Our study demonstrates that PTN derived from cardiac fibroblasts may play potential role in pressure overload-induced HCM through activating the PTN-SDC4 pathway in cardiac fibroblasts and macrophages, which may be a potential therapeutic target for pressure overload-induced HCM patients.
Collapse
Affiliation(s)
- Ke Sheng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuqing Ran
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xing Feng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yijia Wang
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Size Zhou
- Department of General Surgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuting Guan
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Pingping Tan
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Songwei Qian
- Department of General Surgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiguang Zhao
- Department of Pathology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bing Zhang
- Engineering Research Center of Techniques and Instruments for Diagnosis and Treatment of Congenital Heart Disease, , Institute of Developmental and Regenerative Medicine, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weiping Ji
- Department of General Surgery, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, China; Department of General Surgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Chao Niu
- Pediatric Research Institute, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China.
| | - Xiaoling Guo
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
2
|
Shi M, Yuan H, Li Y, Guo Z, Wei J. Targeting Macrophage Phenotype for Treating Heart Failure: A New Approach. Drug Des Devel Ther 2024; 18:4927-4942. [PMID: 39525046 PMCID: PMC11549885 DOI: 10.2147/dddt.s486816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Heart failure (HF) is a disease with high morbidity and mortality rates worldwide and significantly affects human health. Currently, the treatment options for HF are limited, and there is an urgent need to discover new therapeutic targets and strategies. Macrophages are innate immune cells involved in the development of HF. They play a crucial role in maintaining cardiac homeostasis and regulating cardiac stress. Recently, macrophages have received increasing attention as potential targets for treating HF. With the improvement of technological means, the study of macrophages in HF has made great progress. This article discusses the biological functions of macrophage phagocytosis, immune response, and tissue repair. The polarization, pyroptosis, autophagy, and apoptosis are of macrophages, deeply involved in the pathogenesis of HF. Modulation of the phenotypic changes of macrophages can improve immune-inflammation, myocardial fibrosis, energy metabolism, apoptosis, and angiogenesis in HF.
Collapse
Affiliation(s)
- Min Shi
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent TCM Diagnosis and Preventive Treatment of Chronic Diseases, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Hui Yuan
- Hunan Key Laboratory of Colleges and Universities of Intelligent TCM Diagnosis and Preventive Treatment of Chronic Diseases, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 41020, People’s Republic of China
| | - Ya Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| | - Zhihua Guo
- Hunan Key Laboratory of Colleges and Universities of Intelligent TCM Diagnosis and Preventive Treatment of Chronic Diseases, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- First Clinical College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 41020, People’s Republic of China
| | - Jiaming Wei
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
- Hunan Key Laboratory of Colleges and Universities of Intelligent TCM Diagnosis and Preventive Treatment of Chronic Diseases, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China
| |
Collapse
|
3
|
Liu Z, Liang X, Zhang Y, Deng W, Wang Y, Lu Z, Liu Q, Wei L. Drug Repurposing: Research Progress of Niclosamide and Its Derivatives on Antibacterial Activity. Infect Drug Resist 2024; 17:4539-4556. [PMID: 39464831 PMCID: PMC11505561 DOI: 10.2147/idr.s490998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
The development of antibiotic resistance complicates the treatment of infectious diseases and is a global public health threat. However, drug repurposing can address this resistance issue and reduce research and development costs. Niclosamide is a salicylanilide compound approved by the Food and Drug Administration (FDA), and it has been used clinically for treating parasitic infections for many years. Recent studies have shown that niclosamide can inhibit bacterial and fungus activity by affecting the quorum sensing system, biofilm formation, cell membrane potential, and other mechanisms. Here, we discuss recent advances in the antimicrobial applications of niclosamide and its derivatives to provide new perspectives in treating infectious diseases.
Collapse
Affiliation(s)
- Zhihong Liu
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| | - Xiaofang Liang
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
| | - Yu Zhang
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
| | - Wenbo Deng
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, People’s Republic of China
| | - Yulin Wang
- Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Zhangping Lu
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| | - Qianqian Liu
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| | - Lianhua Wei
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, People’s Republic of China
| |
Collapse
|
4
|
Lu Y, Liang X, Song J, Guan Y, Yang L, Shen R, Niu Y, Guo Z, Zhu N. Niclosamide modulates phenotypic switch and inflammatory responses in human pulmonary arterial smooth muscle cells. Mol Cell Biochem 2024:10.1007/s11010-024-05061-6. [PMID: 38980591 DOI: 10.1007/s11010-024-05061-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 06/29/2024] [Indexed: 07/10/2024]
Abstract
Excessive proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) represent key steps of pulmonary vascular remodeling, leading to the development of pulmonary arterial hypertension (PAH) and right ventricular failure. Niclosamide (NCL), an FDA-approved anthelmintic, has been shown to regulate cell proliferation, migration, invasion, and apoptosis through a variety of signaling pathways. However, its role on modulating the phenotypic switch and inflammatory responses in PASMCs remains unclear. In this study, cell proliferation assay showed that NCL inhibited PDGF-BB induced proliferation of human PASMCs in a dose-dependent manner. Western blot analysis further confirmed a notable reduction in the expression of cyclin D1 and PCNA proteins. Subsequently, flow cytometry analysis demonstrated that NCL induced an increased percentage of cells in the G1 phase while promoting apoptosis in PASMCs. Moreover, both scratch wound assay and transwell assay confirmed that NCL decreased PDGF-BB-induced migration of PASMCs. Mechanistically, western blot revealed that pretreatment of PASMCs with NCL markedly restored the protein levels of SMA, SM22, and calponin, while reducing phosphorylation of P38/STAT3 signaling in the presence of PDGF-BB. Interestingly, macrophages adhesion assay showed that NCL markedly reduced recruitment of Calcein-AM labeled RAW264.7 by TNFα-stimulated PASMCs. Western blot revealed that NCL suppressed TNFα-induced expression of both of VCAM-1 and ICAM-1 proteins. Furthermore, pretreatment of PASMCs with NCL significantly inhibited NLRP3 inflammasome activity through reducing NLRP3, AIM2, mature interleukin-1β (IL-β), and cleaved Caspase-1 proteins expression. Together, these results suggested versatile effects of NCL on controlling of proliferation, migration, and inflammatory responses in PASMCs through modulating different pathways, indicating that repurposing of NCL may emerge as a highly effective drug for PAH treatment.
Collapse
Affiliation(s)
- Yuwen Lu
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiaogan Liang
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Jingwen Song
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yugen Guan
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Liang Yang
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Rongrong Shen
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yunpu Niu
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Zhifu Guo
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Ni Zhu
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
5
|
Zhang H, Muhetarijiang M, Chen RJ, Hu X, Han J, Zheng L, Chen T. Mitochondrial Dysfunction: A Roadmap for Understanding and Tackling Cardiovascular Aging. Aging Dis 2024:AD.2024.0058. [PMID: 38739929 DOI: 10.14336/ad.2024.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
Cardiovascular aging is a progressive remodeling process constituting a variety of cellular and molecular alterations that are closely linked to mitochondrial dysfunction. Therefore, gaining a deeper understanding of the changes in mitochondrial function during cardiovascular aging is crucial for preventing cardiovascular diseases. Cardiac aging is accompanied by fibrosis, cardiomyocyte hypertrophy, metabolic changes, and infiltration of immune cells, collectively contributing to the overall remodeling of the heart. Similarly, during vascular aging, there is a profound remodeling of blood vessel structure. These remodeling present damage to endothelial cells, increased vascular stiffness, impaired formation of new blood vessels (angiogenesis), the development of arteriosclerosis, and chronic vascular inflammation. This review underscores the role of mitochondrial dysfunction in cardiac aging, exploring its impact on fibrosis and myocardial alterations, metabolic remodeling, immune response remodeling, as well as in vascular aging in the heart. Additionally, we emphasize the significance of mitochondria-targeted therapies in preventing cardiovascular diseases in the elderly.
Collapse
Affiliation(s)
- Han Zhang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Mairedan Muhetarijiang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ryan J Chen
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaosheng Hu
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Han
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liangrong Zheng
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
6
|
Hu N, Sun M, Lv N, Gao Y, Fu X, Xing D, Guo X, Zhai S, Zhang R. ROS-Suppression Nanoplatform Combined Activation of STAT3/Bcl-2 Pathway for Preventing Myocardial Infarction in Mice. ACS APPLIED MATERIALS & INTERFACES 2024; 16:12188-12201. [PMID: 38288981 DOI: 10.1021/acsami.3c16735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2024]
Abstract
Myocardial infarction (MI) is the leading cause of death worldwide. The most effective way to treat myocardial infarction is to rescue ischemic cardiomyocytes. After an ischemic event, the overproduction of reactive oxygen species (ROS) is a key driver of myocardial injury. The produced ROS affects mitochondrial function and induces apoptosis in cardiomyocytes. This was accomplished by constructing platelet-membrane-encapsulated ROS-responsive drug-releasing nanoparticles (PMN@NIC-MalNPs) to deliver malonate and niclosamide (NIC). The results revealed that PMN@NIC-MalNPs degraded and released malonate and niclosamide in a high-level ROS microenvironment, effectively reducing the oxidative stress and apoptosis rate. By enhancing basal mitochondrial oxygen consumption rate (OCR), adenosine triphosphate (ATP) production, and spare respiratory capacity (SRC) in vitro, reduced the oxidative stress levels and restored mitochondrial function. In vivo studies revealed that the PMN@NIC-MalNPs improved cardiac dysfunction, inhibited succinate dehydrogenase (SDH) activity, increased ATP production, and reduced the myocardial infarct size in myocardial infarction model mice. Further, transcriptome analysis and Western blot revealed that PMN@NIC-MalNPs prevented apoptosis by activating the expressions of the signal transducer and activator of transcription 3 (STAT3) and Bcl-2, and inhibiting the expression of Bax. Thus, this study provides a novel therapeutic solution for treating myocardial infarction and predicting the viability of an antioxidant and antiapoptotic therapeutic solution in the treatment of myocardial injury.
Collapse
Affiliation(s)
- Nan Hu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430030, China
| | - Meng Sun
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430030, China
- Cardiology Department, First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030032, China
| | - Nan Lv
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, 56 Xinjian South Road, Taiyuan 030032, China
| | - Yangyang Gao
- The First Clinical Medical College, Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, China
| | - Xiaohong Fu
- The First Clinical Medical College, Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, China
| | - Dayi Xing
- The First Clinical Medical College, Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, China
| | - Xiang Guo
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430030, China
| | - Shaodong Zhai
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430030, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
7
|
Bin S, Xinyi F, Huan P, Xiaoqin Z, Jiming W, Yi H, Ziyue L, Xiaochun Z, Zhouqi L, Bangwei Z, Jing J, Shihui L, Jinlai G. SOX4 as a potential therapeutic target for pathological cardiac hypertrophy. Eur J Pharmacol 2023; 958:176071. [PMID: 37741429 DOI: 10.1016/j.ejphar.2023.176071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/20/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
Pathological cardiac hypertrophy can lead to heart failure, making its prevention crucial. SOX4, a SOX transcription factor, regulates tissue growth and development, although its role in pathological cardiac hypertrophy is unclear. We found that the SOX4 expression was elevated in hypertrophic hearts and angiotensin II (Ang II)-treated neonatal rat cardiomyocytes (NRCMs), and knocking down the SOX4 expression in NRCMs and mouse hearts significantly reduced the hypertrophic response. Mechanistically, SOX4 can bind to the SIRT3 promoter, inhibit SIRT3 transcription and expression, and thus affect downstream MnSOD acetylation levels, leading to abnormal increases in ROS and oxidative stress levels and promoting the occurrence of cardiac hypertrophy. In conclusion, this study identified a new role for SOX4 in regulating cardiac hypertrophy, and decreasing SOX4 expression may be a potential treatment for pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Shen Bin
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, 314000, China
| | - Feng Xinyi
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, 314000, China
| | - Pan Huan
- Department of Central Laboratory, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| | - Zhang Xiaoqin
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, 314000, China
| | - Wu Jiming
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, 314000, China
| | - He Yi
- Department of Central Laboratory, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China; Department of Urology, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| | - Li Ziyue
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, 314000, China
| | - Zou Xiaochun
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, 314000, China
| | - Lu Zhouqi
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, 314000, China
| | - Zhou Bangwei
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, 314000, China
| | - Jin Jing
- Department of Urology, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China.
| | - Liu Shihui
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, 314000, China.
| | - Gao Jinlai
- Department of Pharmacology, College of Medical, Jiaxing University, Jiaxing, 314000, China.
| |
Collapse
|
8
|
Zhang L, Li W, Liu X, Guo J, Wu X, Wang J. Niclosamide inhibits TGF-β1-induced fibrosis of human Tenon's fibroblasts by regulating the MAPK-ERK1/2 pathway. Exp Eye Res 2023; 235:109628. [PMID: 37619828 DOI: 10.1016/j.exer.2023.109628] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 07/24/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Preventing postoperative bleb scar formation is an effective way of improving glaucoma filtration surgery (GFS) outcome. Use of more effective antifibrotic drugs with fewer adverse effects may be a good way to address the problem. In the present study, we use a primary cell model, consisting of Tenon's fibroblasts obtained from patients with glaucoma, which were stimulated with TGF-β1 to induce the fibrotic phenotype. We explored the effects of niclosamide on TGF-β1-induced fibrosis in these cells and examined its underlying mechanism of action. A transcriptome sequencing assay was used to explore possible signaling pathways involved. Niclosamide inhibited cell proliferation and migration, and decreased the levels of alpha-smooth muscle actin, type I and type III collagen in human Tenon's fibroblasts induced by TGF-β1. Niclosamide also induced apoptosis and counteracted TGF-β1-induced cytoskeletal changes and extracellular matrix accumulation. Moreover, niclosamide decreased TGF-β1-induced phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2) protein expression in human Tenon's fibroblasts. The results indicate that niclosamide inhibits TGF-β1-induced fibrosis in human Tenon's fibroblasts by blocking the MAPK-ERK1/2 signaling pathway. Thus, niclosamide is a potentially promising antifibrotic drug that could improve glaucoma filtration surgery success rate.
Collapse
Affiliation(s)
- Liyun Zhang
- Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Wei Li
- Department of Pediatric Respiratory Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Xin Liu
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, 518000, Guangdong, China
| | - Junhong Guo
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, 518000, Guangdong, China
| | - Xueping Wu
- Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Jiantao Wang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
9
|
Ishikane S, Arioka M, Takahashi-Yanaga F. Promising small molecule anti-fibrotic agents: Newly developed or repositioned drugs targeting myofibroblast transdifferentiation. Biochem Pharmacol 2023; 214:115663. [PMID: 37336252 DOI: 10.1016/j.bcp.2023.115663] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023]
Abstract
Fibrosis occurs in all organs and tissues except the brain, and its progression leads to dysfunction of affected organs. Fibrosis-induced organ dysfunction results from the loss of elasticity, strength, and functionality of tissues due to the extracellular matrix secreted by myofibroblasts that express smooth muscle-type actin as a marker. Myofibroblasts, which play a major role in fibrosis, were once thought to originate exclusively from activated fibroblasts; however, it is now clear that myofibroblasts are diverse in origin, from epithelial cells, endothelial cells, adipocytes, macrophages, and other cells. Fibrosis of vital organs, such as the heart, lungs, kidneys, and liver, is a serious chronic disease that ultimately leads to death. Currently, anti-cancer drugs have made remarkable progress, as evidenced by the development of many molecular-targeted drugs, and are making a significant contribution to improving the prognosis of cancer treatment. However, the development of anti-fibrotic agents, which also play an important role in prognosis, has lagged. In this review, the current knowledge regarding myofibroblasts is summarized, with particular attention given to their origin and transdifferentiation signaling pathways (e.g., TGF-β, Wnt/β-catenin, YAP/TAZ and AMPK signaling pathways). The development of new small molecule anti-fibrotic agents and the repositioning of existing drugs targeting myofibroblast transdifferentiation are discussed.
Collapse
Affiliation(s)
- Shin Ishikane
- Department of Pharmacology, Faculty of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Masaki Arioka
- Department of Pharmacology, Faculty of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Fumi Takahashi-Yanaga
- Department of Pharmacology, Faculty of Medicine, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan.
| |
Collapse
|
10
|
Garvin AM, Hale TM. State of Change: Epigenetic and Mitochondrial Regulation of Cardiac Fibroblast Activation. CURRENT OPINION IN PHYSIOLOGY 2022. [DOI: 10.1016/j.cophys.2022.100557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|