1
|
Liang Q, Liu X, Peng X, Luo T, Su Y, Xu X, Xie H, Gao H, Chen Z, Xie C. Salvianolic acid B in fibrosis treatment: a comprehensive review. Front Pharmacol 2024; 15:1442181. [PMID: 39139645 PMCID: PMC11319160 DOI: 10.3389/fphar.2024.1442181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Fibrosis is a public health issue of great concern characterized by the excessive deposition of extracellular matrix, leading to the destruction of parenchymal tissue and organ dysfunction that places a heavy burden on the global healthcare system due to its high incidence, disability, and mortality. Salvianolic acid B (SalB) has positively affected various human diseases, including fibrosis. In this review, we concentrate on the anti-fibrotic effects of SalB from a molecular perspective while providing information on the safety, adverse effects, and drug interactions of SalB. Additionally, we discuss the innovative SalB formulations, which give some references for further investigation and therapeutic use of SalB's anti-fibrotic qualities. Even with the encouraging preclinical data, additional research is required before relevant clinical trials can be conducted. Therefore, we conclude with recommendations for future studies. It is hoped that this review will provide comprehensive new perspectives on future research and product development related to SalB treatment of fibrosis and promote the efficient development of this field.
Collapse
Affiliation(s)
- Qingzhi Liang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaoqin Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xi Peng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ting Luo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yi Su
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xin Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Hongyan Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Hong Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhengtao Chen
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Yu J, Feng D, Bao L, Zhang B. TRIM32 Inhibits NEK7 Ubiquitylation-Dependent Microglia Pyroptosis After Spinal Cord Injury. Mol Biotechnol 2023:10.1007/s12033-023-00989-4. [PMID: 38030945 DOI: 10.1007/s12033-023-00989-4] [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: 06/29/2023] [Accepted: 11/10/2023] [Indexed: 12/01/2023]
Abstract
Spinal cord injury (SCI) is a disabling disease associated with microglial activation. Tripartite motif containing 32 (TRIM32) is an E3 ubiquitin ligase that plays a role in SCI. This study aimed to explore the role of TRIM32 in SCI and its potential mechanisms. We established an SCI mouse model to assess the function of TRIM32 using quantitative real-time polymerase chain reaction (qPCR), and hematoxylin and eosin staining. Additionally, a lipopolysaccharides (LPS)-induced cell injury model was generated to explore the impact of TRIM32 on pyroptosis using qPCR, propidium iodide staining, and western blotting. The ubiquitylation of NEK7 was analyzed using western blotting, co-immunoprecipitation, and immunofluorescence staining. The results showed that TRIM32 expression was increased in SCI mice and LPS-induced BV-2 cells. Overexpression of TRIM32 ameliorated SCI in mice and suppressed pyroptosis in LPS-treated BV-2 cells. Additionally, the E3 ligase TRIM32 promoted the ubiquitylation of NEK7 at the K64 site, leading to the downregulation of NEK7 levels. Inhibiting NEK7 ubiquitylation reversed the suppression of pyroptosis by TRIM32. In conclusion, TRIM32 inhibits microglia pyroptosis by facilitating the ubiquitylation of NEK7 at the K64 site, thereby alleviating the progression of SCI. The findings suggest that TRIM32 has the potential to be a therapeutic target of SCI.
Collapse
Affiliation(s)
- Jiasheng Yu
- Department of Orthopedics, Shuyang Hospital of Traditional Chinese Medicine (Shuyang Hospital of Traditional Chinese Medicine affiliated to Yangzhou University), No. 28, Shanghai Middle Road, Shuyang County, Suqian City, 223600, Jiangsu Province, China
| | - Dongqian Feng
- Department of Orthopedics, Shuyang Hospital of Traditional Chinese Medicine (Shuyang Hospital of Traditional Chinese Medicine affiliated to Yangzhou University), No. 28, Shanghai Middle Road, Shuyang County, Suqian City, 223600, Jiangsu Province, China
| | - Lei Bao
- Department of Orthopedics, Shuyang Hospital of Traditional Chinese Medicine (Shuyang Hospital of Traditional Chinese Medicine affiliated to Yangzhou University), No. 28, Shanghai Middle Road, Shuyang County, Suqian City, 223600, Jiangsu Province, China
| | - Bin Zhang
- Department of Orthopedics, Shuyang Hospital of Traditional Chinese Medicine (Shuyang Hospital of Traditional Chinese Medicine affiliated to Yangzhou University), No. 28, Shanghai Middle Road, Shuyang County, Suqian City, 223600, Jiangsu Province, China.
| |
Collapse
|
3
|
Chen C, Hu X, Chen X. Saikosaponin A protects against uremic toxin indole‑3 acetic acid‑induced damage to the myocardium. Mol Med Rep 2023; 28:159. [PMID: 37417356 PMCID: PMC10407609 DOI: 10.3892/mmr.2023.13046] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/17/2023] [Indexed: 07/08/2023] Open
Abstract
Chronic kidney disease (CKD)‑associated cardiac injury is a common complication in patients with CKD. Indole‑3 acetic acid (IAA) is a uremic toxin that injures the cardiovascular system. Saikosaponin A (SSA) protects against pressure overload‑induced cardiac fibrosis. However, the role and molecular mechanisms of IAA and SSA in CKD‑associated cardiac injury remain unclear. The present study investigated the effects of IAA and SSA on CKD‑associated cardiac injury in neonatal mouse cardiomyocytes and a mouse model of CKD. The expression of tripartite motif‑containing protein 16 (Trim16), receptor interacting protein kinase 2 (RIP2) and phosphorylated‑p38 were assessed using western blotting. The ubiquitination of RIP2 was measured by coimmunoprecipitation, and mouse cardiac structure and function were evaluated using hematoxylin and eosin staining and echocardiography. The results demonstrated that, SSA inhibited IAA‑induced cardiomyocyte hypertrophy, upregulated Trim16 expression, downregulated RIP2 expression and decreased p38 phosphorylation. Furthermore, Trim16 mediated SSA‑induced degradation of RIP2 by ubiquitination. In a mouse model of IAA‑induced CKD‑associated cardiac injury, SSA upregulated the protein expression levels of Trim16 and downregulated those of RIP2. Moreover, SSA alleviated heart hypertrophy and diastolic dysfunction in IAA‑treated mice. Taken together, these results suggest that SSA is a protective agent against IAA‑induced CKD‑associated cardiac injury and that Trim16‑mediated ubiquitination‑related degradation of RIP2 and p38 phosphorylation may contribute to the development of CKD‑associated cardiac injury.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Medical Science, Yangzhou Polytechnic College, Yangzhou, Jiangsu 225127, P.R. China
| | - Xiaoyuan Hu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xinguang Chen
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
4
|
Hou H, Chen Y, Feng X, Xu G, Yan M. Tripartite motif‑containing 14 may aggravate cardiac hypertrophy via the AKT signalling pathway in neonatal rat cardiomyocytes and transgenic mice. Mol Med Rep 2023; 28:173. [PMID: 37503784 PMCID: PMC10433706 DOI: 10.3892/mmr.2023.13060] [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: 12/15/2022] [Accepted: 05/25/2023] [Indexed: 07/29/2023] Open
Abstract
Tripartite motif‑containing 14 (TRIM14) is an E3 ubiquitin ligase that primarily participates in the natural immune response and in tumour development via ubiquitination. However, the role of TRIM14 in cardiac hypertrophy is not currently clear. The present study examined the role of TRIM14 in cardiac hypertrophy and its potential molecular mechanism. TRIM14 was overexpressed in neonatal rat cardiomyocytes using adenovirus and cardiomyocyte hypertrophy was induced using phenylephrine (PE). Cardiomyocyte hypertrophy was assessed by measuring cardiomyocyte surface area and markers of hypertrophy. In addition, TRIM14‑transgenic (TRIM14‑TG) mice were created and cardiac hypertrophy was induced using transverse aortic constriction (TAC). Cardiac function, heart weight‑to‑body weight ratio (HW/BW), cardiomyocyte cross‑sectional area, cardiac fibrosis and hypertrophic markers were further examined. The expression of AKT signalling pathway‑related proteins was detected. TRIM14 overexpression in cardiomyocytes promoted PE‑induced increases in cardiomyocyte surface area and hypertrophic markers. TRIM14‑TG mice developed worse cardiac function, greater HW/BW, cross‑sectional area and cardiac fibrosis, and higher levels of hypertrophic markers in response to TAC. TRIM14 overexpression also increased the phosphorylation levels of AKT, GSK‑3β, mTOR and p70S6K in vivo and in vitro. To the best our knowledge, the present study was the first to reveal that overexpression of TRIM14 aggravated cardiac hypertrophy in vivo and in vitro, which may be related to activation of the AKT signalling pathway.
Collapse
Affiliation(s)
- Hongwei Hou
- Department of Cardiology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Yan Chen
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Xiuyuan Feng
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Guang Xu
- Department of Cardiology, Ezhou Central Hospital, Ezhou, Hubei 436000, P.R. China
| | - Min Yan
- Department of General Practice, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
5
|
Jeong SY, Choi JH, Kim J, Woo JS, Lee EH. Tripartite Motif-Containing Protein 32 (TRIM32): What Does It Do for Skeletal Muscle? Cells 2023; 12:2104. [PMID: 37626915 PMCID: PMC10453674 DOI: 10.3390/cells12162104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/07/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Tripartite motif-containing protein 32 (TRIM32) is a member of the tripartite motif family and is highly conserved from flies to humans. Via its E3 ubiquitin ligase activity, TRIM32 mediates and regulates many physiological and pathophysiological processes, such as growth, differentiation, muscle regeneration, immunity, and carcinogenesis. TRIM32 plays multifunctional roles in the maintenance of skeletal muscle. Genetic variations in the TRIM32 gene are associated with skeletal muscular dystrophies in humans, including limb-girdle muscular dystrophy type 2H (LGMD2H). LGMD2H-causing genetic variations of TRIM32 occur most frequently in the C-terminal NHL (ncl-1, HT2A, and lin-41) repeats of TRIM32. LGMD2H is characterized by skeletal muscle dystrophy, myopathy, and atrophy. Surprisingly, most patients with LGMD2H show minimal or no dysfunction in other tissues or organs, despite the broad expression of TRIM32 in various tissues. This suggests more prominent roles for TRIM32 in skeletal muscle than in other tissues or organs. This review is focused on understanding the physiological roles of TRIM32 in skeletal muscle, the pathophysiological mechanisms mediated by TRIM32 genetic variants in LGMD2H patients, and the correlations between TRIM32 and Duchenne muscular dystrophy (DMD).
Collapse
Affiliation(s)
- Seung Yeon Jeong
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jun Hee Choi
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jooho Kim
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin Seok Woo
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 10833, USA
| | - Eun Hui Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
6
|
Qian H, Lu Z, Hao C, Zhao Y, Bo X, Hu Y, Zhang Y, Yao Y, Ma G, Chen L. TRIM44 aggravates cardiac fibrosis after myocardial infarction via TAK1 stabilization. Cell Signal 2023:110744. [PMID: 37271349 DOI: 10.1016/j.cellsig.2023.110744] [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: 02/28/2023] [Revised: 05/03/2023] [Accepted: 05/29/2023] [Indexed: 06/06/2023]
Abstract
Myocardial infarction (MI) is one of the most dangerous cardiovascular events. Cardiac fibrosis is a common pathological feature of remodeling after injury that is related to adverse clinical results with no effective treatment. Previous studies have confirmed that TRIM44, an E3 ligase, can promote the proliferation and migration of various tumor cells. However, the role of TRIM44 in cardiac fibrosis remains unknown. Models of TGF-β1 stimulation and MI-induced fibrosis were established to investigate the role and potential underlying mechanism of TRIM44 in cardiac fibrosis. The results showed that cardiac fibrosis was significantly inhibited after TRIM44 knockdown in a mouse model of MI, while it was enhanced when TRIM44 was overexpressed. Furthermore, in vitro studies showed that fibrosis markers were significantly reduced in cardiac fibroblasts (CFs) with TRIM44 knockdown, whereas TRIM44 overexpression promoted the expression of fibrosis markers. Mechanistically, TRIM44 maintains TAK1 stability by inhibiting the degradation of k48-linked polyubiquitination-mediated ubiquitination, thereby increasing phosphorylated TAK1 expression in the fibrotic environment and activating MAPKs to promote fibrosis. Pharmacological inhibition of TAK1 phosphorylation reversed the fibrogenic effects of TRIM44 overexpression. Combined, these results suggest that TRIM44 is a potential therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Hao Qian
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhengri Lu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chunshu Hao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuanyuan Zhao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiangwei Bo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ya Hu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yao Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lijuan Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Nanjing 211200, China.
| |
Collapse
|
7
|
Jiang XY, Guan FF, Ma JX, Dong W, Qi XL, Zhang X, Chen W, Gao S, Gao X, Pan S, Wang JZ, Ma YW, Zhang LF, Lu D. Cardiac-specific Trim44 knockout in rat attenuates isoproterenol-induced cardiac remodeling via inhibition of AKT/mTOR pathway. Dis Model Mech 2023; 16:276033. [PMID: 35855640 PMCID: PMC9441189 DOI: 10.1242/dmm.049444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
Abstract
When pathological hypertrophy progresses to heart failure (HF), the prognosis is often very poor. Therefore, it is crucial to find new and effective intervention targets. Here, myocardium-specific Trim44 knockout rats were generated using CRISPR-Cas9 technology. Cardiac phenotypic observations revealed that Trim44 knockout affected cardiac morphology at baseline. Rats with Trim44 deficiency exhibited resistance to cardiac pathological changes in response to stimulation via isoproterenol (ISO) treatment, including improvement of cardiac remodeling and dysfunction by morphological and functional observations, reduced myocardial fibrosis and reduced expression of molecular markers of cardiac stress. Furthermore, signal transduction validation associated with growth and hypertrophy development in vivo and in vitro demonstrated that Trim44 deficiency inhibited the activation of signaling pathways involved in myocardial hypertrophy, especially response to pathological stress. In conclusion, the present study indicates that Trim44 knockout attenuates ISO-induced pathological cardiac remodeling through blocking the AKT/mTOR/GSK3β/P70S6K signaling pathway. This is the first study to demonstrate the function and importance of Trim44 in the heart at baseline and under pathological stress. Trim44 could be a novel therapeutic target for prevention of cardiac hypertrophy and HF. Summary: This is the first study to demonstrate the function of Trim44 in the heart at baseline and under pathological stress. Trim44 could be a novel therapeutic target for prevention of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiao-Yu Jiang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Fei-Fei Guan
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Jia-Xin Ma
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Xiao-Long Qi
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Xu Zhang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Wei Chen
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Shan Gao
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Xiang Gao
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Shuo Pan
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Ji-Zheng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100037, China
| | - Yuan-Wu Ma
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Lian-Feng Zhang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Dan Lu
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| |
Collapse
|
8
|
Huang J, Qu Q, Dai Y, Ren D, Qian J, Ge J. Detrimental Role of PDZ-RhoGEF in Pathological Cardiac Hypertrophy. Hypertension 2023; 80:403-415. [PMID: 36448462 DOI: 10.1161/hypertensionaha.122.19142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND Postsynaptic density 95/disk-large/ZO-1 Rho guanine nucleotide exchange factor (PDZ-RhoGEF, PRG) functions as a RhoGEF for activated Gα13 and transmits activation signals to downstream signaling pathways in various pathological processes. Although the prohypertrophic effect of activated Gα13 (guanine nucleotide binding protein alpha 13; a heterotrimeric G protein) is well-established, the role of PDZ-RhoGEF in pathological cardiac hypertrophy is still obscure. METHODS Genetically engineered mice and neonatal rat ventricular myocytes were generated to investigate the function of PRG in pathological myocardial hypertrophy. The prohypertrophic stimuli-induced alternations in the morphology and intracellular signaling were measured in myocardium and neonatal rat ventricular myocytes. Furthermore, multiple molecular methodologies were used to identify the precise molecular mechanisms underlying PDZ-RhoGEF function. RESULTS Increased PDZ-RhoGEF expression was documented in both hypertrophied hearts and neonatal rat ventricular myocytes. Upon prohypertrophic stimuli, the PDZ-RhoGEF-deficient hearts displayed alleviated cardiomyocyte enlargement and attenuated collagen deposition with improved cardiac function, whereas the adverse hypertrophic responses in hearts and neonatal rat ventricular myocytes were markedly exaggerated by PDZ-RhoGEF overexpression. Mechanistically, RhoA (ras homolog family member A)-dependent signaling pathways may function as the downstream effectors of PDZ-RhoGEF in hypertrophic remodeling, as confirmed by rescue experiments using a RhoA inhibitor and dominant-negative RhoA. Furthermore, PDZ-RhoGEF is associated with activated Gα13 and contributes to Gα13-mediated activation of RhoA-dependent signaling. CONCLUSIONS Our data provide the first evidence that PDZ-RhoGEF promotes pathological cardiac hypertrophy by linking activated Gα13 to RhoA-dependent signaling pathways. Therefore, PDZ-RhoGEF has the potential to be a diagnostic marker or therapeutic target for pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Jia Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China and National Clinical Research Center for Interventional Medicine (J.H., Y.D., D.R., J.Q., J.G.)
| | - Qingrong Qu
- Department of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China and Shanghai Clinical Research Center for Tuberculosis, Shanghai, China (Q.Q.)
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China and National Clinical Research Center for Interventional Medicine (J.H., Y.D., D.R., J.Q., J.G.)
| | - Daoyuan Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China and National Clinical Research Center for Interventional Medicine (J.H., Y.D., D.R., J.Q., J.G.)
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China and National Clinical Research Center for Interventional Medicine (J.H., Y.D., D.R., J.Q., J.G.)
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China and National Clinical Research Center for Interventional Medicine (J.H., Y.D., D.R., J.Q., J.G.)
| |
Collapse
|
9
|
Lu Z, Deng M, Ma G, Chen L. TRIM38 protects H9c2 cells from hypoxia/reoxygenation injury via the TRAF6/TAK1/NF- κB signalling pathway. PeerJ 2022; 10:e13815. [PMID: 36061751 PMCID: PMC9435518 DOI: 10.7717/peerj.13815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/08/2022] [Indexed: 01/18/2023] Open
Abstract
Tripartite motif (TRIM) 38 is a ubiquitin E3 protein ligase that is involved in various intracellular physiological processes. However, the role of TRIM38 in myocardial ischaemia/reperfusion (I/R) injury remains to be elucidated. We aimed to establish an in vitro cellular hypoxia/reperfusion (H/R) model to explore the role and potential mechanisms of TRIM38 in H9c2, a rat cardiomyoblast cell line. Recombinant adenoviruses for silencing or overexpressing TRIM38 were constructed and transfected into H9c2 cells. Western blotanalysisshowed that TRIM38 expression was significantly decreased after H/R injury. Functionally, TRIM38 expression relieved inflammatory responses and oxidative stress, and inhibited H/R-induced apoptosis in H9c2 cells. Mechanistically, TRIM38 overexpression inhibited H/R-induced transforming growth factor beta-activated kinase 1 (TAK1)/nuclear factor-kappa B (NF-κB) pathway activity in H9c2 cells. The opposite results were observed after TRIM38 knockdown. Furthermore, H/R-induced injury aggravated by TRIM38 deficiency in H9c2 cells was reversed upon treatment with 5Z-7-oxozeaenol, a TAK1 inhibitor. Therefore, TRIM38 reduction attenuated the anti-apoptotic capacity and anti-inflammatory potential of H/R-stimulated H9c2 cells by activating the TAK1/NF-κB signalling pathway. Specifically, TRIM38 alleviated H/R-induced H9c2 cell injury by promoting TNF receptor-associated factor 6 degradation, which led to the inactivation of the TAK1/NF-κB signalling pathway. Thus, our study provides new insights into the molecular mechanisms underlying H/R-induced myocardial injuries.
Collapse
Affiliation(s)
- Zhengri Lu
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Mengen Deng
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Lijuan Chen
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China,Department of Cardiology, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Nanjing, Jiangsu, China
| |
Collapse
|
10
|
Tripartite Motif 38 Attenuates Cardiac Fibrosis after Myocardial Infarction by Suppressing TAK1 Activation via TAB2/3 Degradation. iScience 2022; 25:104780. [PMID: 35982795 PMCID: PMC9379576 DOI: 10.1016/j.isci.2022.104780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 06/27/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022] Open
Abstract
The role of tripartite motif (TRIM) 38, a ubiquitin E3 ligase regulating various pathophysiological processes, in cardiac fibrosis remains unclear. Here, a model of angiotensin II and myocardial infarction (MI)-induced fibrosis was established to explore its role in cardiac fibrosis and its underlying mechanisms. Cardiac fibrosis in the mouse MI model was mitigated by TRIM38 overexpression, but aggravated by its depletion. Consistently, in vitro overexpression or knockdown of TRIM38 ameliorated or aggravated the proliferation and secretion of cardiac fibroblasts (CFs) exposed to fibrotic stimulation, respectively. Mechanistically, TRIM38 suppressed cardiac fibrosis progression by attenuating TAK1/MAPK signaling. Inhibiting TAK1/MAPK signaling with a pharmacological inhibitor greatly reversed the effects of TRIM38 knockdown on CF secretion. Specifically, TRIM38 interacted with and “targeted” TAB2 and TAB3 for degradation, subsequently inhibiting TAK1 phosphorylation and negatively regulating MAPK signaling. These findings can help develop therapeutic strategies to treat and prevent cardiac fibrosis. TRIM38 expression is negatively correlated with cardiac fibrosis progression TRIM38 ameliorates the proliferation and secretion of CFs post fibrotic stimulation TRIM38 overexpression attenuates cardiac fibrosis progression in MI mice TRIM38 inhibits the TAK1/MAPK pathway by targeting the degradation of TAB2 and TAB3
Collapse
|
11
|
Liu J, Li W, Deng KQ, Tian S, Liu H, Shi H, Fang Q, Liu Z, Chen Z, Tian T, Gan S, Hu F, Hu M, Cheng X, Ji YX, Zhang P, She ZG, Zhang XJ, Chen S, Cai J, Li H. The E3 Ligase TRIM16 Is a Key Suppressor of Pathological Cardiac Hypertrophy. Circ Res 2022; 130:1586-1600. [PMID: 35437018 DOI: 10.1161/circresaha.121.318866] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Pathological cardiac hypertrophy is one of the leading causes of heart failure with highly complicated pathogeneses. The E3 ligase TRIM16 (tripartite motif-containing protein 16) has been recognized as a pivotal regulator to control cell survival, immune response, and oxidative stress. However, the role of Trim16 in cardiac hypertrophy is unknown. METHODS We generated cardiac-specific knockout mice and adeno-associated virus serotype 9-Trim16 mice to evaluate the function of Trim16 in pathological myocardial hypertrophy. The direct effect of TRIM16 on cardiomyocyte enlargement was examined using an adenovirus system. Furthermore, we combined RNA-sequencing and interactome analysis that was followed by multiple molecular biological methodologies to identify the direct target and corresponding molecular events contributing to TRIM16 function. RESULTS We found an intimate correlation of Trim16 expression with hypertrophy-related heart failure in both human and mouse. Our functional investigations and unbiased transcriptomic analyses clearly demonstrated that Trim16 deficiency markedly exacerbated cardiomyocyte enlargement in vitro and in transverse aortic constriction-induced cardiac hypertrophy mouse model, whereas Trim16 overexpression attenuated cardiac hypertrophy and remodeling. Mechanistically, Prdx1 (peroxiredoxin 1) is an essential target of Trim16 in cardiac hypertrophy. We found that Trim16 interacts with Prdx1 and inhibits its phosphorylation, leading to a robust enhancement of its downstream Nrf2 (nuclear factor-erythroid 2-related factor 2) pathway to block cardiac hypertrophy. Trim16-blocked Prdx1 phosphorylation was largely dependent on a direct interaction between Trim16 and Src and the resultant Src ubiquitinational degradation. Notably, Prdx1 knockdown largely abolished the anti-hypertrophic effects of Trim16 overexpression. CONCLUSIONS Our findings provide the first evidence supporting Trim16 as a novel suppressor of pathological cardiac hypertrophy and indicate that targeting the Trim16-Prdx1 axis represents a promising therapeutic strategy for hypertrophy-related heart failure.
Collapse
Affiliation(s)
- Jiayi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.L.).,Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Wei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.L.).,Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Ke-Qiong Deng
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Department of Cardiology, Zhongnan Hospital of Wuhan University, China. (K.-Q.D., Z.C.)
| | - Song Tian
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Hui Liu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.).,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.)
| | - Hongjie Shi
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Qian Fang
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Zhen Liu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Ze Chen
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Department of Cardiology, Zhongnan Hospital of Wuhan University, China. (K.-Q.D., Z.C.)
| | - Tian Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.L.).,Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Shanyu Gan
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Fengjiao Hu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Medical Science Research Center, Zhongnan Hospital of Wuhan University, China. (F.H., H. Li)
| | - Manli Hu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.).,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.)
| | - Xu Cheng
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.).,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.)
| | - Yan-Xiao Ji
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Peng Zhang
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.L.).,Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Xiao-Jing Zhang
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Shaoze Chen
- Department of Cardiology, Huanggang Central Hospital, China (S.C.).,Huanggang Institute of Translational Medicine, Huanggang, China (S.C.)
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China (J.C.)
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.L.).,Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Medical Science Research Center, Zhongnan Hospital of Wuhan University, China. (F.H., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| |
Collapse
|
12
|
Chen Y, Li L, Hu C, Zhao X, Zhang P, Chang Y, Shang Y, Pang Y, Qian W, Qiu X, Zhang H, Zhang D, Zhang S, Li Y. Lingguizhugan decoction dynamically regulates MAPKs and AKT signaling pathways to retrogress the pathological progression of cardiac hypertrophy to heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153951. [PMID: 35131606 DOI: 10.1016/j.phymed.2022.153951] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/08/2022] [Accepted: 01/15/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Heart failure (HF) is a grave health concern, with high morbidity and mortality, calling for the urgent need for new and alternative pharmacotherapies. Lingguizhugan decoction (LD) is a classic Chinese formula clinically used to treat HF. However, the underlying mechanisms involved are not fully elucidated. PURPOSE Based on that, this study aims to investigate the effects and underlying mechanisms of LD on HF. METHODS After confirming the therapeutic benefits of LD in transverse aortic constriction (TAC)-induced HF mice, network pharmacology and transcriptomic analyzes were utilized to predict the potential molecular targets and pathways of LD treatment in failing hearts, which were evaluated at 3 and 9 w after TAC. UHPLC-QE-MS analysis was utilized to detect bioactive ingredients from LD and plasma of LD-treated rats. RESULTS Our results showed that LD markedly alleviated cardiac dysfunction via down-regulating CH-related genes and proteins expression in TAC mice. Significantly, cardiac hypertrophy signaling, including AKT and MAPKs signaling pathways, were identified, suggesting the pathways as likely regulatory targets for LD treatment. LD inhibited p38 and ERK phosphorylated expression levels, with the latter effect likely dependent on regulation of AMPK. Interestingly, LD exerted a dual modulatory role in the AKT-GSK3β/mTOR/P70S6K signaling pathway's regulation, which was characterized by stimulatory activity at 3 w and inhibitory effects at 9 w. Finally, 15 bioactive compounds detected from plasma were predicted as the potential regulators of the AKT-GSK3β/mTOR and MAPKs signaling pathways. CONCLUSION Our study shows LD's therapeutic efficacy in failing hearts, signifies LD as HF medication that acts dynamically by balancing AKT-GSK3β/mTOR/P70S6K and MAPKs pathways, and reveals possible bioactive compounds responsible for LD effects on HF.
Collapse
Affiliation(s)
- Yao Chen
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Cunyu Hu
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Xin Zhao
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Peng Zhang
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Yanxu Chang
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Ye Shang
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Yafen Pang
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Weiqiang Qian
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Xianzhe Qiu
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Hongxia Zhang
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, 69 Zeng Chan Road, He Bei, Tianjin 300250, China
| | - Deqin Zhang
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China
| | - Shukun Zhang
- Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, 6 Chang Jiang Road, Nan Kai, Tianjin 300100, China.
| | - Yuhong Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jing Hai, Tianjin 301617, China.
| |
Collapse
|
13
|
Chen Z, Tian L, Wang L, Ma X, Lei F, Chen X, Fu R. TRIM32 Inhibition Attenuates Apoptosis, Oxidative Stress, and Inflammatory Injury in Podocytes Induced by High Glucose by Modulating the Akt/GSK-3β/Nrf2 Pathway. Inflammation 2021; 45:992-1006. [PMID: 34783942 DOI: 10.1007/s10753-021-01597-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 11/05/2021] [Indexed: 11/27/2022]
Abstract
Hyperglycemia-induced oxidative stress in podocytes exerts a major role in the pathological process of diabetic nephropathy. Tripartite motif-containing protein 32 (TRIM32) has been reported to be a key protein in the modulation of cellular apoptosis and oxidative stress under various pathological processes. However, whether TRIM32 participates in the regulation of high glucose (HG)-induced injury in podocytes has not been investigated. This work aimed to assess the possible role of TRIM32 in mediating HG-induced apoptosis, oxidative stress, and inflammatory response in podocytes in vitro. Our results showed a marked increase in TRIM32 expression in HG-exposed podocytes and the glomeruli of diabetic mice. Loss-of-function experiments showed that TRIM32 knockdown improves the viability of HG-stimulated podocytes and suppresses HG-induced apoptosis, oxidative stress, and inflammatory responses in podocytes. Further investigation revealed that TRIM32 inhibition enhances the activation of nuclear factor erythroid 2-related factor 2 (Nrf2) signaling, which is associated with the modulation of the Akt/glycogen synthase kinase-3β (GSK-3β) axis in podocytes following HG exposure. However, Akt suppression abrogated the TRIM32 knockdown-mediated activation of Nrf2 in HG-exposed podocytes. Nrf2 knockdown also markedly abolished the protective effects induced by TRIM32 inhibition o in HG-exposed podocytes. In summary, this work demonstrated that TRIM32 inhibition protects podocytes from HG-induced injury by potentiating Nrf2 signaling through modulation of Akt/GSK-3β signaling. The findings reveal the potential role of TRIM32 in mediating podocyte injury during the progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Zhao Chen
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Lifang Tian
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Li Wang
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Xiaotao Ma
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Fuqian Lei
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Xianghui Chen
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China
| | - Rongguo Fu
- Department of Nephrology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi Province, China.
| |
Collapse
|
14
|
TRIM proteins in fibrosis. Biomed Pharmacother 2021; 144:112340. [PMID: 34678729 DOI: 10.1016/j.biopha.2021.112340] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is an outcome of tissue repair after different types of injuries. The homeostasis of extracellular matrix is broken, and excessive deposition occurs, affecting the normal function of tissues and organs, which could become prostrated in serious cases.Finding a suitable target to regulate the repair process and reduce the damage caused by fibrosis is a hot research topic at present. The TRIM family is number of one of the E3 ubiquitin ligase subfamilies and participates in various biological processes including intracellular signal transduction, apoptosis, autophagy, and immunity by regulating the ubiquitination of target proteins. For the past few years, the important role of TRIM in the occurrence and development of fibrosis has been gradually revealed. In this review, we focus on the recent emerging topics on TRIM proteins in the regulation of fibrosis, fibrosis-related cytokines and pathways.
Collapse
|
15
|
Liu K, Chen Y, Ai F, Li YQ, Zhang K, Zhang WT. PHLDA3 inhibition attenuates endoplasmic reticulum stress-induced apoptosis in myocardial hypoxia/reoxygenation injury by activating the PI3K/AKT signaling pathway. Exp Ther Med 2021; 21:613. [PMID: 33936270 PMCID: PMC8082641 DOI: 10.3892/etm.2021.10045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Endoplasmic reticulum stress (ERS)-induced apoptosis serves a crucial role in the pathogenesis of myocardial ischemia/reperfusion injury (MIRI). Previous studies have confirmed that pleckstrin homology-like domain family A member 3 (PHLDA3) is an important mediator in ERS-associated apoptosis. The aim of the current study focused on whether PHLDA3 served protective effects on hypoxia/reoxygenation (H/R)-injured cardiomyocytes by inhibiting ERS-induced apoptosis. Furthermore, the molecular mechanisms associated with the PI3K/AKT signaling pathway were investigated. Primary neonatal rat cardiomyocytes were isolated and randomized into four groups: i) Control + adenovirus encoding scrambled short hairpin RNA (AdshRNA); ii) control + adenoviral vectors encoding PHLDA3 shRNA (AdshPHLDA3); iii) H/R+ AdshRNA and iv) H/R+AdshPHLDA3. AdshPHLDA3 was used to knock down PHLDA3. An H/R injury model was constructed by treatment with hypoxia for 4 h followed by reoxygenation for 6 h. A PI3K/AKT inhibitor, LY294002, was supplemented in mechanistic studies. Cell viability and LDH/CK releases were detected to evaluate myocardial damage. Flow cytometry assays were used to assess apoptotic response. Western blotting assays were used to detect protein expression. The results demonstrated that H/R induced myocardial damage and increased PHLDA3 expression. ERS-induced apoptosis was significantly increased following H/R injury, as indicated by increased apoptotic rates and ERS-associated protein expression, including those of CHOP, 78 kDa glucose-regulated protein and caspase-12. However, PHLDA3 inhibition following AdshPHLDA3 transfection reversed cell damage and ERS-associated apoptosis on H/R injury. Studies for molecular mechanisms concluded that the apoptosis-inhibition effects and cardioprotective roles of PHLDA3 inhibition were induced partly by the activation of the PI3K/AKT pathway, which was verified by LY294002 treatment. In conclusion, in the process of H/R injury, PHLDA3 inhibition reduced ERS-induced apoptosis and H/R injury by activating the PI3K/AKT pathway. PHLDA3 may be a therapeutic target for the treatment of MIRI.
Collapse
Affiliation(s)
- Kai Liu
- Department of Geriatric Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Ying Chen
- Department of Geriatric Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Fen Ai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Yun-Qian Li
- Department of Geriatric Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Kun Zhang
- Department of Geriatric Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Wei-Tong Zhang
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
16
|
Hu Z, Song Q, Ma H, Guo Y, Zhang T, Xie H, Luo X. TRIM32 inhibits the proliferation and migration of pulmonary artery smooth muscle cells through the inactivation of PI3K/Akt pathway in pulmonary arterial hypertension. J Bioenerg Biomembr 2021; 53:309-320. [PMID: 33694017 DOI: 10.1007/s10863-021-09880-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/07/2021] [Indexed: 01/27/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fetal cardiovascular disease. Tripartite motif 32 (TRIM32) is a member of TRIM family that has been found to be involved in cardiovascular disease. However, the role of TRIM32 in PAH remains unclear. Here we investigated the effects of TRIM32 on hypoxia-induced pulmonary artery smooth muscle cells (PASMCs) in vitro. Our results showed that TRIM32 protein level in the plasma samples from PAH patients was decreased as compared with healthy volunteers. Exposure to hypoxia condition caused a significant decrease in TRIM32 expression in PASMCs. Overexpression of TRIM32 inhibited hypoxia-induced proliferation and migration of PASMCs. TRIM32 overexpression elevated the increased apoptotic rate and caspase-3 activity in hypoxia-induced PASMCs. Moreover, overexpression of TRIM32 reversed hypoxia-induced down-regulation of myocardin, SM 22 and calponin, as well as up-regulation of osteopontin (OPN). Whereas, TRIM32 knockdown shwed the opposite effect. Furthermore, overexpression of TRIM32 inhibited hypoxia-induced activation of PI3K/Akt with decreased phosphorylated level of PI3K and Akt. Additionally, activation of PI3K/Akt by IGF-1 treatment reversed the effects of TRIM32 on hypoxia-induced PASMCs. In conclusion, these findings indicated that TRIM32 was involved in the development of PAH through regulating the proliferation, migration, apoptosis and dedifferentiation of PASMCs, which might be mediated by the PI3K/Akt signaling pathway. Thus, TRIM32 might be a potential target for PAH treatment.
Collapse
Affiliation(s)
- Zhi Hu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China.
| | - Qiang Song
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Hui Ma
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Yaozhang Guo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Tingting Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Hang Xie
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Xiaohui Luo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
17
|
Aweida D, Cohen S. Breakdown of Filamentous Myofibrils by the UPS-Step by Step. Biomolecules 2021; 11:biom11010110. [PMID: 33467597 PMCID: PMC7830001 DOI: 10.3390/biom11010110] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 01/08/2023] Open
Abstract
Protein degradation maintains cellular integrity by regulating virtually all biological processes, whereas impaired proteolysis perturbs protein quality control, and often leads to human disease. Two major proteolytic systems are responsible for protein breakdown in all cells: autophagy, which facilitates the loss of organelles, protein aggregates, and cell surface proteins; and the ubiquitin-proteasome system (UPS), which promotes degradation of mainly soluble proteins. Recent findings indicate that more complex protein structures, such as filamentous assemblies, which are not accessible to the catalytic core of the proteasome in vitro, can be efficiently degraded by this proteolytic machinery in systemic catabolic states in vivo. Mechanisms that loosen the filamentous structure seem to be activated first, hence increasing the accessibility of protein constituents to the UPS. In this review, we will discuss the mechanisms underlying the disassembly and loss of the intricate insoluble filamentous myofibrils, which are responsible for muscle contraction, and whose degradation by the UPS causes weakness and disability in aging and disease. Several lines of evidence indicate that myofibril breakdown occurs in a strictly ordered and controlled manner, and the function of AAA-ATPases is crucial for their disassembly and loss.
Collapse
|
18
|
Under construction: The dynamic assembly, maintenance, and degradation of the cardiac sarcomere. J Mol Cell Cardiol 2020; 148:89-102. [PMID: 32920010 DOI: 10.1016/j.yjmcc.2020.08.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/11/2022]
Abstract
The sarcomere is the basic contractile unit of striated muscle and is a highly ordered protein complex with the actin and myosin filaments at its core. Assembling the sarcomere constituents into this organized structure in development, and with muscle growth as new sarcomeres are built, is a complex process coordinated by numerous factors. Once assembled, the sarcomere requires constant maintenance as its continuous contraction is accompanied by elevated mechanical, thermal, and oxidative stress, which predispose proteins to misfolding and toxic aggregation. To prevent protein misfolding and maintain sarcomere integrity, the sarcomere is monitored by an assortment of protein quality control (PQC) mechanisms. The need for effective PQC is heightened in cardiomyocytes which are terminally differentiated and must survive for many years while preserving optimal mechanical output. To prevent toxic protein aggregation, molecular chaperones stabilize denatured sarcomere proteins and promote their refolding. However, when old and misfolded proteins cannot be salvaged by chaperones, they must be recycled via degradation pathways: the calpain and ubiquitin-proteasome systems, which operate under basal conditions, and the stress-responsive autophagy-lysosome pathway. Mutations to and deficiency of the molecular chaperones and associated factors charged with sarcomere maintenance commonly lead to sarcomere structural disarray and the progression of heart disease, highlighting the necessity of effective sarcomere PQC for maintaining cardiac function. This review focuses on the dynamic regulation of assembly and turnover at the sarcomere with an emphasis on the chaperones involved in these processes and describes the alterations to chaperones - through mutations and deficient expression - implicated in disease progression to heart failure.
Collapse
|
19
|
Miao R, Lu Y, He X, Liu X, Chen Z, Wang J. Ubiquitin-specific protease 19 blunts pathological cardiac hypertrophy via inhibition of the TAK1-dependent pathway. J Cell Mol Med 2020; 24:10946-10957. [PMID: 32798288 PMCID: PMC7521154 DOI: 10.1111/jcmm.15724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 11/26/2022] Open
Abstract
Ubiquitin‐specific protease 19 (USP19) belongs to USP family and is involved in promoting skeletal muscle atrophy. Although USP19 is expressed in the heart, the role of USP19 in the heart disease remains unknown. The present study provides in vivo and in vitro data to reveal the role of USP19 in preventing pathological cardiac hypertrophy. We generated USP19‐knockout mice and isolated neonatal rat cardiomyocytes (NRCMs) that overexpressed or were deficient in USP19 to investigate the effect of USP19 on transverse aortic constriction (TAC) or phenylephrine (PE)‐mediated cardiac hypertrophy. Echocardiography, pathological and molecular analysis were used to determine the extent of cardiac hypertrophy, fibrosis, dysfunction and inflammation. USP19 expression was markedly increased in rodent hypertrophic heart or cardiomyocytes underwent TAC or PE culturing, the increase was mediated by the reduction of Seven In Absentia Homolog‐2. The extent of TAC‐induced cardiac hypertrophy, fibrosis, dysfunction and inflammation in USP19‐knockout mice was exacerbated. Consistently, gain‐of‐function and loss‐of‐function approaches that involved USP19 in cardiomyocytes suggested that the down‐regulation of USP19 promoted the hypertrophic phenotype, while the up‐regulation of USP19 improved the worsened phenotype. Mechanistically, the USP19‐elicited cardiac hypertrophy improvement was attributed to the abrogation of the transforming growth factor beta‐activated kinase 1 (TAK1)‐p38/JNK1/2 transduction. Furthermore, the inhibition of TAK1 abolished the aggravated hypertrophy induced by the loss of USP19. In conclusion, the present study revealed that USP19 and the downstream of TAK1‐p38/JNK1/2 signalling pathway might be a potential target to attenuate pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Rujia Miao
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yao Lu
- Department of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xue He
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xuelian Liu
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhiheng Chen
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiangang Wang
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
Yang H, Wang XX, Zhou CY, Xiao X, Tian C, Li HH, Yin CL, Wang HX. Tripartite motif 10 regulates cardiac hypertrophy by targeting the PTEN/AKT pathway. J Cell Mol Med 2020; 24:6233-6241. [PMID: 32343488 PMCID: PMC7294125 DOI: 10.1111/jcmm.15257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/26/2020] [Accepted: 03/18/2020] [Indexed: 12/21/2022] Open
Abstract
The pathogenesis of cardiac hypertrophy is tightly associated with activation of intracellular hypertrophic signalling pathways, which leads to the synthesis of various proteins. Tripartite motif 10 (TRIM10) is an E3 ligase with important functions in protein quality control. However, its role in cardiac hypertrophy was unclear. In this study, neonatal rat cardiomyocytes (NRCMs) and TRIM10-knockout mice were subjected to phenylephrine (PE) stimulation or transverse aortic constriction (TAC) to induce cardiac hypertrophy in vitro and in vivo, respectively. Trim10 expression was significantly increased in hypertrophied murine hearts and PE-stimulated NRCMs. Knockdown of TRIM10 in NRCMs alleviated PE-induced changes in the size of cardiomyocytes and hypertrophy gene expression, whereas TRIM10 overexpression aggravated these changes. These results were further verified in TRIM10-knockout mice. Mechanistically, we found that TRIM10 knockout or knockdown decreased AKT phosphorylation. Furthermore, we found that TRIM10 knockout or knockdown increased ubiquitination of phosphatase and tensin homolog (PTEN), which negatively regulated AKT activation. The results of this study reveal the involvement of TRIM10 in pathological cardiac hypertrophy, which may occur by prompting of PTEN ubiquitination and subsequent activation of AKT signalling. Therefore, TRIM10 may be a promising target for treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Hui Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Xiao-Xiao Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Chun-Yu Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Xue Xiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Cui Tian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated hospital of Dalian Medical University, Dalian, China
| | - Chun-Lin Yin
- Department of Cardiology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Hong-Xia Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Liu J, Liu X, Hui X, Cai L, Li X, Yang Y, Shu S, Wang F, Xia H, Li S. Novel Role for Pleckstrin Homology-Like Domain Family A, Member 3 in the Regulation of Pathological Cardiac Hypertrophy. J Am Heart Assoc 2019; 8:e011830. [PMID: 31426686 PMCID: PMC6759890 DOI: 10.1161/jaha.118.011830] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background Pleckstrin homology-like domain family A, member 3 (PHLDA3), a crucial member of the PHLDA family, is involved in tumor suppression, kidney injury, liver injury, and glucose metabolism. However, the role of PHLDA3 in pathological cardiac hypertrophy and heart failure remains unclear. Methods and Results In the present study, PHLDA3 expression was downregulated in hypertrophic murine hearts and angiotensin II-treated cardiomyocytes. Next, an in vitro study suggested, by using gain- and loss-of-function approaches, that PHLDA3 attenuates Ang II exposure-induced cardiomyocyte hypertrophy. Consistent with the cell phenotype, disruption of PHLDA3 aggravated the effects of pressure overload-induced pathological cardiac hypertrophy, fibrosis, and dysfunction. In contrast, PHLDA3 overexpression resulted in an attenuated hypertrophic phenotype. Molecular analysis revealed that PHLDA3 suppressed the activation of AKT-mTOR-GSK3β-P70S6K signaling in response to hypertrophic stress, and the blockage of AKT activation rescued these adverse pathological effects of PHLDA3 deficiency-induced by AB and Ang II, respectively, in vivo and in vitro. Conclusions Collectively, our data indicated that PHLDA3 could ameliorate pressure overload-induced cardiac remodeling mainly by blocking the AKT signaling pathway, suggesting that PHLDA3 may represent a therapeutic target for the treatment of pathological cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Jia Liu
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China.,Department of Cardiology Cang Zhou People's Hospital Cangzhou Hebei China
| | - Xiaoxiong Liu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China.,Cardiovascular Research Institute Wuhan University Wuhan China.,Hubei Key Laboratory of Cardiology Wuhan China
| | - Xuejun Hui
- Department of Cardiology Second Hospital of Jilin University Changchun Jilin China
| | - Lin Cai
- Zhongnan Hospital of Wuhan University Wuhan China.,Institute of Model Animal of Wuhan University Wuhan China
| | - Xuebo Li
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China
| | - Yang Yang
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China
| | - Shangzhi Shu
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China
| | - Fan Wang
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China
| | - Hao Xia
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China.,Cardiovascular Research Institute Wuhan University Wuhan China.,Hubei Key Laboratory of Cardiology Wuhan China
| | - Shuyan Li
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China
| |
Collapse
|
22
|
Yan K, Wang K, Li P. The role of post-translational modifications in cardiac hypertrophy. J Cell Mol Med 2019; 23:3795-3807. [PMID: 30950211 PMCID: PMC6533522 DOI: 10.1111/jcmm.14330] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/06/2019] [Accepted: 03/19/2019] [Indexed: 12/19/2022] Open
Abstract
Pathological cardiac hypertrophy involves excessive protein synthesis, increased cardiac myocyte size and ultimately the development of heart failure. Thus, pathological cardiac hypertrophy is a major risk factor for many cardiovascular diseases and death in humans. Extensive research in the last decade has revealed that post‐translational modifications (PTMs), including phosphorylation, ubiquitination, SUMOylation, O‐GlcNAcylation, methylation and acetylation, play important roles in pathological cardiac hypertrophy pathways. These PTMs potently mediate myocardial hypertrophy responses via the interaction, stability, degradation, cellular translocation and activation of receptors, adaptors and signal transduction events. These changes occur in response to pathological hypertrophy stimuli. In this review, we summarize the roles of PTMs in regulating the development of pathological cardiac hypertrophy. Furthermore, PTMs are discussed as potential targets for treating or preventing cardiac hypertrophy.
Collapse
Affiliation(s)
- Kaowen Yan
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, China
| | - Kun Wang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
23
|
Chen X, Su J, Feng J, Cheng L, Li Q, Qiu C, Zheng Q. TRIM72 contributes to cardiac fibrosis via regulating STAT3/Notch-1 signaling. J Cell Physiol 2019; 234:17749-17756. [PMID: 30820965 DOI: 10.1002/jcp.28400] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/19/2022]
Abstract
Cardiac fibrosis is a pathophysiological process characterized by excessive deposition of extracellular matrix. We developed a cardiac hypertrophy model using transverse aortic constriction (TAC) to uncover mechanisms relevant to excessive deposition of extracellular matrix in mouse myocardial cells. TAC caused upregulation of Tripartite motif protein 72 (TRIM72), a tripartite motif-containing protein that is critical for proliferation and migration. Importantly, in vivo silencing of TRIM72 reversed TAC-induced cardiac fibrosis, as indicated by markedly increased left ventricular systolic pressure and decreased left ventricular end-diastolic pressure. TRIM72 knockdown also attenuated deposition of fibrosis marker collagen type I and α-smooth muscle actin (α-SMA). In an in vitro study, TRIM72 was similarly upregulated in cardiac fibroblasts. Knockdown of TRIM72 markedly suppressed collagen type I and α-SMA expression and significantly decreased the proliferation and migration of cardiac fibroblasts. However, TRIM72 overexpression markedly increased collagen type I and α-SMA expression and increased the proliferation and migration of cardiac fibroblasts. Further study demonstrated that TRIM72 increased phosphorylated STAT3 in cardiac fibroblasts. TRIM72 knockdown in cardiac fibroblasts resulted in increased expression of Notch ligand Jagged-1 and its downstream gene and Notch-1 intracellular domain. Inhibition of Notch-1 abrogated sh-TRIM72-induced cardiac fibrosis. Together, our results support a novel role for TRIM72 in maintaining fibroblast-to-myofibroblast transition and suppressing fibroblast growth by regulating the STAT3/Notch-1 pathway.
Collapse
Affiliation(s)
- Xu Chen
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Second Clinical Medical College, Jinan University, Shenzhen, China.,Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Su
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianyu Feng
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Liang Cheng
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qing Li
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Chen Qiu
- Department of Respiratory and Critical Medicine, Shenzhen People's Hospital, Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Second Clinical Medical College, Jinan University, Shenzhen, China.,Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
24
|
Editorial Commentary: Dichotomous actions of the E3-ligase Ring TRIMmers in cardiac myocytes. Trends Cardiovasc Med 2018; 29:9-11. [PMID: 30585158 DOI: 10.1016/j.tcm.2018.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 12/31/2022]
|
25
|
Gupta I, Varshney NK, Khan S. Emergence of Members of TRAF and DUB of Ubiquitin Proteasome System in the Regulation of Hypertrophic Cardiomyopathy. Front Genet 2018; 9:336. [PMID: 30186311 PMCID: PMC6110912 DOI: 10.3389/fgene.2018.00336] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/03/2018] [Indexed: 01/10/2023] Open
Abstract
The ubiquitin proteasome system (UPS) plays an imperative role in many critical cellular processes, frequently by mediating the selective degradation of misfolded and damaged proteins and also by playing a non-degradative role especially important as in many signaling pathways. Over the last three decades, accumulated evidence indicated that UPS proteins are primal modulators of cell cycle progression, DNA replication, and repair, transcription, immune responses, and apoptosis. Comparatively, latest studies have demonstrated a substantial complexity by the UPS regulation in the heart. In addition, various UPS proteins especially ubiquitin ligases and proteasome have been identified to play a significant role in the cardiac development and dynamic physiology of cardiac pathologies such as ischemia/reperfusion injury, hypertrophy, and heart failure. However, our understanding of the contribution of UPS dysfunction in the plausible development of cardiac pathophysiology and the complete list of UPS proteins regulating these afflictions is still in infancy. The recent emergence of the roles of TNF receptor-associated factor (TRAFs) and deubiquitinating enzymes (DUBs) superfamily in hypertrophic cardiomyopathy has enhanced our knowledge. In this review, we have mainly compiled the TRAF superfamily of E3 ligases and few DUBs proteins with other well-documented E3 ligases such as MDM2, MuRF-1, Atrogin-I, and TRIM 32 that are specific to myocardial hypertrophy. In this review, we also aim to highlight their expression profile following physiological and pathological stimulation leading to the onset of hypertrophic phenotype in the heart that can serve as biomarkers and the opportunity for the development of novel therapies.
Collapse
Affiliation(s)
- Ishita Gupta
- Structural Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.,Drug Discovery Research Center, Translational Health Science and Technology Institute, Faridabad, India
| | - Nishant K Varshney
- Drug Discovery Research Center, Translational Health Science and Technology Institute, Faridabad, India
| | - Sameena Khan
- Drug Discovery Research Center, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
26
|
Borlepawar A, Frey N, Rangrez AY. A systematic view on E3 ligase Ring TRIMmers with a focus on cardiac function and disease. Trends Cardiovasc Med 2018; 29:1-8. [PMID: 29880235 DOI: 10.1016/j.tcm.2018.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/05/2018] [Accepted: 05/22/2018] [Indexed: 01/01/2023]
Abstract
Ubiquitination, a post-translational modification via ubiquitin-proteasome-system, is one of the vital cellular processes involved in intracellular signaling, cell death, transcriptional control, etc. Importantly, it prevents the aggregation of non-functional, misfolded or unfolded, potentially toxic proteins to maintain cellular protein homeostasis. Ubiquitination is accomplished by the concerted action of three enzymatic steps involving E1 activating enzymes, E2 conjugating enzymes, and E3 ligases. Tripartite motif-containing (TRIM) proteins are one of the integral members of E3 ubiquitin ligases in metazoans modulating essential cellular pathways. For long, MuRFs (Muscle ring finger proteins) were the most extensively studied TRIMs for their cardiac function. Recent research advances in the field and our analysis presented here, however, demonstrated broader and ever increasing involvement of additional TRIM E3 ligases in the pathophysiology of heart. In this review, we summarize the known cardiac E3 ligases and their targets, and discuss their role and importance in cardiac proteostasis, pathophysiology and potential therapeutic implications with specific focus on TRIM E3 ligases.
Collapse
Affiliation(s)
- Ankush Borlepawar
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Norbert Frey
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Ashraf Yusuf Rangrez
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105, Kiel, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany.
| |
Collapse
|
27
|
Yang Q, Liu TT, Lin H, Zhang M, Wei J, Luo WW, Hu YH, Zhong B, Hu MM, Shu HB. TRIM32-TAX1BP1-dependent selective autophagic degradation of TRIF negatively regulates TLR3/4-mediated innate immune responses. PLoS Pathog 2017; 13:e1006600. [PMID: 28898289 PMCID: PMC5595311 DOI: 10.1371/journal.ppat.1006600] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 08/22/2017] [Indexed: 12/21/2022] Open
Abstract
Toll-like receptor (TLR)-mediated signaling are critical for host defense against pathogen invasion. However, excessive responses would cause harmful damages to the host. Here we show that deficiency of the E3 ubiquitin ligase TRIM32 increases poly(I:C)- and LPS-induced transcription of downstream genes such as type I interferons (IFNs) and proinflammatory cytokines in both primary mouse immune cells and in mice. Trim32-/- mice produced higher levels of serum inflammatory cytokines and were more sensitive to loss of body weight and inflammatory death upon Salmonella typhimurium infection. TRIM32 interacts with and mediates the degradation of TRIF, a critical adaptor protein for TLR3/4, in an E3 activity-independent manner. TRIM32-mediated as well as poly(I:C)- and LPS-induced degradation of TRIF is inhibited by deficiency of TAX1BP1, a receptor for selective autophagy. Furthermore, TRIM32 links TRIF and TAX1BP1 through distinct domains. These findings suggest that TRIM32 negatively regulates TLR3/4-mediated immune responses by targeting TRIF to TAX1BP1-mediated selective autophagic degradation. TLR3/4-mediated signaling needs to be effectively terminated to avoid excessive immune responses and harmful damages to the host. In this study, we provide genetic evidence to show that the E3 ubiquitin ligase TRIM32 negatively regulates TLR3/4-mediated innate immune and inflammatory responses. Trim32-/- mice are more sensitive to the inflammatory death upon Salmonella typhimurium infection. We found that TRIM32-TAX1BP1-dependent selective autophagic degradation of the adaptor protein TRIF effectively turned off TLR3/4-mediated innate immune and inflammatory responses. Our findings reveal a novel mechanism for terminating innate immune and inflammatory responses mediated by TLR3/4.
Collapse
Affiliation(s)
- Qing Yang
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Tian-Tian Liu
- Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Heng Lin
- Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Man Zhang
- Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jin Wei
- Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Wei-Wei Luo
- Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yun-Hong Hu
- Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Bo Zhong
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Ming-Ming Hu
- Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China
- * E-mail: (MMH); (HBS)
| | - Hong-Bing Shu
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, China
- * E-mail: (MMH); (HBS)
| |
Collapse
|
28
|
Sczesny-Kaiser M, Kowalewski R, Schildhauer TA, Aach M, Jansen O, Grasmücke D, Güttsches AK, Vorgerd M, Tegenthoff M. Treadmill Training with HAL Exoskeleton-A Novel Approach for Symptomatic Therapy in Patients with Limb-Girdle Muscular Dystrophy-Preliminary Study. Front Neurosci 2017; 11:449. [PMID: 28848377 PMCID: PMC5550721 DOI: 10.3389/fnins.2017.00449] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/24/2017] [Indexed: 11/25/2022] Open
Abstract
Purpose: Exoskeletons have been developed for rehabilitation of patients with walking impairment due to neurological disorders. Recent studies have shown that the voluntary-driven exoskeleton HAL® (hybrid assistive limb) can improve walking functions in spinal cord injury and stroke. The aim of this study was to assess safety and effects on walking function of HAL® supported treadmill therapy in patients with limb-girdle muscular dystrophy (LGMD). Materials and Methods: Three LGMD patients received 8 weeks of treadmill training with HAL® 3 times a week. Outcome parameters were 10-meter walk test (10 MWT), 6-minute walk test, and timed-up-and-go test (TUG). Parameters were assessed pre and post training and 6 weeks later (follow-up). Results: All patients completed the therapy without adverse reactions and reported about improvement in endurance. Improvements in outcome parameters after 8 weeks could be demonstrated. Persisting effects were observed after 6 weeks for the 10 MWT and TUG test (follow-up). Conclusions: HAL® treadmill training in LGMD patients can be performed safely and enables an intensive highly repetitive locomotor training. All patients benefitted from this innovative method. Upcoming controlled studies with larger cohorts should prove its effects in different types of LGMD and other myopathies.
Collapse
Affiliation(s)
- Matthias Sczesny-Kaiser
- Department of Neurology, BG-University Hospital Bergmannsheil Bochum, Ruhr University BochumBochum, Germany
| | - Rebecca Kowalewski
- Department of Neurology, BG-University Hospital Bergmannsheil Bochum, Ruhr University BochumBochum, Germany
| | - Thomas A Schildhauer
- Department of General and Trauma Surgery, BG-University Hospital Bergmannsheil Bochum, Ruhr University BochumBochum, Germany
| | - Mirko Aach
- Department of Spinal Cord Injury, BG-University Hospital Bergmannsheil Bochum, Ruhr University BochumBochum, Germany
| | - Oliver Jansen
- Department of General and Trauma Surgery, BG-University Hospital Bergmannsheil Bochum, Ruhr University BochumBochum, Germany
| | - Dennis Grasmücke
- Department of Spinal Cord Injury, BG-University Hospital Bergmannsheil Bochum, Ruhr University BochumBochum, Germany
| | - Anne-Katrin Güttsches
- Department of Neurology, BG-University Hospital Bergmannsheil Bochum, Ruhr University BochumBochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, BG-University Hospital Bergmannsheil Bochum, Ruhr University BochumBochum, Germany
| | - Martin Tegenthoff
- Department of Neurology, BG-University Hospital Bergmannsheil Bochum, Ruhr University BochumBochum, Germany
| |
Collapse
|
29
|
Borlepawar A, Rangrez AY, Bernt A, Christen L, Sossalla S, Frank D, Frey N. TRIM24 protein promotes and TRIM32 protein inhibits cardiomyocyte hypertrophy via regulation of dysbindin protein levels. J Biol Chem 2017; 292:10180-10196. [PMID: 28465353 PMCID: PMC5473223 DOI: 10.1074/jbc.m116.752543] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 04/28/2017] [Indexed: 12/27/2022] Open
Abstract
We have previously shown that dysbindin is a potent inducer of cardiomyocyte hypertrophy via activation of Rho-dependent serum-response factor (SRF) signaling. We have now performed a yeast two-hybrid screen using dysbindin as bait against a cardiac cDNA library to identify the cardiac dysbindin interactome. Among several putative binding proteins, we identified tripartite motif-containing protein 24 (TRIM24) and confirmed this interaction by co-immunoprecipitation and co-immunostaining. Another tripartite motif (TRIM) family protein, TRIM32, has been reported earlier as an E3 ubiquitin ligase for dysbindin in skeletal muscle. Consistently, we found that TRIM32 also degraded dysbindin in neonatal rat ventricular cardiomyocytes as well. Surprisingly, however, TRIM24 did not promote dysbindin decay but rather protected dysbindin against degradation by TRIM32. Correspondingly, TRIM32 attenuated the activation of SRF signaling and hypertrophy due to dysbindin, whereas TRIM24 promoted these effects in neonatal rat ventricular cardiomyocytes. This study also implies that TRIM32 is a key regulator of cell viability and apoptosis in cardiomyocytes via simultaneous activation of p53 and caspase-3/-7 and inhibition of X-linked inhibitor of apoptosis. In conclusion, we provide here a novel mechanism of post-translational regulation of dysbindin and hypertrophy via TRIM24 and TRIM32 and show the importance of TRIM32 in cardiomyocyte apoptosis in vitro.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cardiomyopathy, Hypertrophic/metabolism
- Cardiomyopathy, Hypertrophic/pathology
- Carrier Proteins/antagonists & inhibitors
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cells, Cultured
- Dysbindin
- Dystrophin-Associated Proteins/chemistry
- Dystrophin-Associated Proteins/genetics
- Dystrophin-Associated Proteins/metabolism
- HEK293 Cells
- Humans
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Peptide Fragments/chemistry
- Peptide Fragments/genetics
- Peptide Fragments/metabolism
- Protein Stability
- Proteolysis
- RNA Interference
- Rats
- Rats, Wistar
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/metabolism
- Serum Response Factor/agonists
- Serum Response Factor/antagonists & inhibitors
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Signal Transduction
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Tripartite Motif Proteins/antagonists & inhibitors
- Tripartite Motif Proteins/genetics
- Tripartite Motif Proteins/metabolism
- Ubiquitin-Protein Ligases/antagonists & inhibitors
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
Collapse
Affiliation(s)
- Ankush Borlepawar
- From the Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel and
- the DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Ashraf Yusuf Rangrez
- From the Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel and
- the DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Alexander Bernt
- From the Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel and
- the DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Lynn Christen
- From the Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel and
| | - Samuel Sossalla
- From the Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel and
- the DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Derk Frank
- From the Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel and
- the DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Norbert Frey
- From the Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel and
- the DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| |
Collapse
|
30
|
Gilda JE, Gomes AV. Proteasome dysfunction in cardiomyopathies. J Physiol 2017; 595:4051-4071. [PMID: 28181243 DOI: 10.1113/jp273607] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/13/2017] [Indexed: 12/16/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) plays a critical role in removing unwanted intracellular proteins and is involved in protein quality control, signalling and cell death. Because the heart is subject to continuous metabolic and mechanical stress, the proteasome plays a particularly important role in the heart, and proteasome dysfunction has been suggested as a causative factor in cardiac dysfunction. Proteasome impairment has been detected in cardiomyopathies, heart failure, myocardial ischaemia, and hypertrophy. Proteasome inhibition is also sufficient to cause cardiac dysfunction in healthy pigs, and patients using a proteasome inhibitor for cancer therapy have a higher incidence of heart failure. In this Topical Review we discuss the experimental data which suggest UPS dysfunction is a common feature of cardiomyopathies, with an emphasis on hypertrophic cardiomyopathy caused by sarcomeric mutations. We also propose potential mechanisms by which cardiomyopathy-causing mutations may lead to proteasome impairment, such as altered calcium handling and increased oxidative stress due to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jennifer E Gilda
- Department of Neurobiology, Physiology, and Behaviour, University of California, Davis, CA, 95616, USA
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology, and Behaviour, University of California, Davis, CA, 95616, USA.,Department of Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA
| |
Collapse
|
31
|
Chen L, Huang J, Ji YX, Mei F, Wang PX, Deng KQ, Jiang X, Ma G, Li H. Tripartite Motif 8 Contributes to Pathological Cardiac Hypertrophy Through Enhancing Transforming Growth Factor β–Activated Kinase 1–Dependent Signaling Pathways. Hypertension 2017; 69:249-258. [DOI: 10.1161/hypertensionaha.116.07741] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 05/26/2016] [Accepted: 11/26/2016] [Indexed: 11/16/2022]
Abstract
Tripartite motif (TRIM) 8 functions as an E3 ubiquitin ligase, interacting with and ubiquitinating diverse substrates, and is implicated in various pathological processes. However, the function of TRIM8 in the heart remains largely uncharacterized. This study aims to explore the role of TRIM8 in the development of pathological cardiac hypertrophy. Mice and isolated neonatal rat cardiomyocytes overexpressing or lacking TRIM8 were examined in several experiments. The effect of aortic banding–induced cardiac hypertrophy was analyzed by echocardiographic, pathological and molecular analyses. Our results indicated that the TRIM8 overexpression in hearts exacerbated the cardiac hypertrophy triggered by aortic banding. In contrast, the development of pathological cardiac hypertrophy was profoundly blocked in TRIM8-deficient hearts. Mechanistically, our study suggests that TRIM8 may elicit cardiodetrimental effects by promoting the activation of transforming growth factor β–activated kinase 1 (TAK1)-p38/JNK signaling pathways. Similar results were observed in cultured neonatal rat cardiomyocytes treated with angiotensin II. The rescue experiments using the TAK1-specific inhibitor 5z-7-ox confirmed the requirement of TAK1 activation in TRIM8-mediated pathological cardiac hypertrophy. Furthermore, TRIM8 contributed to TAK1 activation by binding to and promoting TAK1 ubiquitination. In conclusion, our study demonstrates that TRIM8 plays a deleterious role in pressure overload–induced cardiac hypertrophy by accelerating the activation of TAK1-dependent signaling pathways.
Collapse
Affiliation(s)
- Lijuan Chen
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University Jiangsu, China (L.C., J.H., G.M.); Animal Experiment Center/Animal Biosafety Level-III Laboratory (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.) and Medical Research Institute, School of Medicine (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.), Wuhan University, China
| | - Jia Huang
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University Jiangsu, China (L.C., J.H., G.M.); Animal Experiment Center/Animal Biosafety Level-III Laboratory (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.) and Medical Research Institute, School of Medicine (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.), Wuhan University, China
| | - Yan-xiao Ji
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University Jiangsu, China (L.C., J.H., G.M.); Animal Experiment Center/Animal Biosafety Level-III Laboratory (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.) and Medical Research Institute, School of Medicine (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.), Wuhan University, China
| | - Fanghua Mei
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University Jiangsu, China (L.C., J.H., G.M.); Animal Experiment Center/Animal Biosafety Level-III Laboratory (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.) and Medical Research Institute, School of Medicine (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.), Wuhan University, China
| | - Pi-xiao Wang
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University Jiangsu, China (L.C., J.H., G.M.); Animal Experiment Center/Animal Biosafety Level-III Laboratory (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.) and Medical Research Institute, School of Medicine (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.), Wuhan University, China
| | - Ke-qiong Deng
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University Jiangsu, China (L.C., J.H., G.M.); Animal Experiment Center/Animal Biosafety Level-III Laboratory (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.) and Medical Research Institute, School of Medicine (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.), Wuhan University, China
| | - Xi Jiang
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University Jiangsu, China (L.C., J.H., G.M.); Animal Experiment Center/Animal Biosafety Level-III Laboratory (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.) and Medical Research Institute, School of Medicine (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.), Wuhan University, China
| | - Genshan Ma
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University Jiangsu, China (L.C., J.H., G.M.); Animal Experiment Center/Animal Biosafety Level-III Laboratory (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.) and Medical Research Institute, School of Medicine (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.), Wuhan University, China
| | - Hongliang Li
- From the Department of Cardiology, Zhongda Hospital Affiliated to Southeast University Jiangsu, China (L.C., J.H., G.M.); Animal Experiment Center/Animal Biosafety Level-III Laboratory (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.) and Medical Research Institute, School of Medicine (Y.-x.J., F.M., P.-x.W., K.-q.D., X.J., H.L.), Wuhan University, China
| |
Collapse
|
32
|
Myofibril breakdown during atrophy is a delayed response requiring the transcription factor PAX4 and desmin depolymerization. Proc Natl Acad Sci U S A 2017; 114:E1375-E1384. [PMID: 28096335 DOI: 10.1073/pnas.1612988114] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A hallmark of muscle atrophy is the excessive degradation of myofibrillar proteins primarily by the ubiquitin proteasome system. In mice, during the rapid muscle atrophy induced by fasting, the desmin cytoskeleton and the attached Z-band-bound thin filaments are degraded after ubiquitination by the ubiquitin ligase tripartite motif-containing protein 32 (Trim32). To study the order of events leading to myofibril destruction, we investigated the slower atrophy induced by denervation (disuse). We show that myofibril breakdown is a two-phase process involving the initial disassembly of desmin filaments by Trim32, which leads to the later myofibril breakdown by enzymes, whose expression is increased by the paired box 4 (PAX4) transcription factor. After denervation of mouse tibialis anterior muscles, phosphorylation and Trim32-dependent ubiquitination of desmin filaments increased rapidly and stimulated their gradual depolymerization (unlike their rapid degradation during fasting). Trim32 down-regulation attenuated the loss of desmin and myofibrillar proteins and reduced atrophy. Although myofibrils and desmin filaments were intact at 7 d after denervation, inducing the dissociation of desmin filaments caused an accumulation of ubiquitinated proteins and rapid destruction of myofibrils. The myofibril breakdown normally observed at 14 d after denervation required not only dissociation of desmin filaments, but also gene induction by PAX4. Down-regulation of PAX4 or its target gene encoding the p97/VCP ATPase reduced myofibril disassembly and degradation on denervation or fasting. Thus, during atrophy, the initial loss of desmin is critical for the subsequent myofibril destruction, and over time, myofibrillar proteins become more susceptible to PAX4-induced enzymes that promote proteolysis.
Collapse
|
33
|
Lazzari E, Meroni G. TRIM32 ubiquitin E3 ligase, one enzyme for several pathologies: From muscular dystrophy to tumours. Int J Biochem Cell Biol 2016; 79:469-477. [PMID: 27458054 DOI: 10.1016/j.biocel.2016.07.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 01/01/2023]
Abstract
TRIM32 is a member of the TRIpartite Motif family characterised by the presence of an N-terminal three-domain-module that includes a RING domain, which confers E3 ubiquitin ligase activity, one or two B-box domains and a Coiled-Coil region that mediates oligomerisation. Several TRIM32 substrates were identified including muscular proteins and proteins involved in cell cycle regulation and cell motility. As ubiquitination is a versatile post-translational modification that can affect target turnover, sub-cellular localisation or activity, it is likely that diverse substrates may be differentially affected by TRIM32-mediated ubiquitination, reflecting its multi-faceted roles in muscle physiology, cancer and immunity. With particular relevance for muscle physiology, mutations in TRIM32 are associated with autosomal recessive Limb-Girdle Muscular Dystrophy 2H, a muscle-wasting disease with variable clinical spectrum ranging from almost asymptomatic to wheelchair-bound patients. In this review, we will focus on the ability of TRIM32 to mark specific substrates for proteasomal degradation discussing how the TRIM32-proteasome axis may (i) be important for muscle homeostasis and for the pathogenesis of muscular dystrophy; and (ii) define either an oncogenic or tumour suppressive role for TRIM32 in the context of different types of cancer.
Collapse
Affiliation(s)
- Elisa Lazzari
- Department of Life Sciences, University of Trieste and Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Germana Meroni
- Department of Life Sciences, University of Trieste and Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy.
| |
Collapse
|