1
|
Xu Z, Jiang F, Wu X, Ren B, Zhang C, Lin L, Li S. ACAA2 Protects Against Cardiac Dysfunction and Lipid Peroxidation in Renal Insufficiency with the Treatment of S-Nitroso-L-Cysteine. Biomolecules 2025; 15:364. [PMID: 40149900 PMCID: PMC11940541 DOI: 10.3390/biom15030364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/16/2025] [Accepted: 02/22/2025] [Indexed: 03/29/2025] Open
Abstract
The key fatty acid β-oxidation protein acetyl-CoA acyltransferase 2 (ACAA2) plays a significant role in myocardial lipid peroxidation and cardiac dysfunction induced by renal insufficiency. However, the mechanisms of lipid metabolism related to renal insufficiency-associated cardiac dysfunction remain poorly understood, and current clinical treatments have been largely ineffective. Through analysis of the Gene Expression Omnibus (GEO) database, we identified that the cardiac functional changes caused by renal insufficiency were primarily centered around the fatty acid β-oxidation signaling pathway, where ACAA2 plays a pivotal role in fatty acid β-oxidation, the tricarboxylic acid cycle, and ketone body metabolism. In an adenine-induced renal insufficiency mouse model, further examination with hematoxylin-eosin staining, Masson staining, and Oil Red O staining revealed alterations in the heart and kidney as well as the accumulation of lipid. Non-invasive blood pressure measurements and ultrasound images demonstrated improvements of peripheral vascular and right ventricular hemodynamic parameters with S-nitroso-L-cysteine (CSNO) inhalation therapy. In cell experiments, knocking down ACAA2 led to accumulation of lipid droplets and exacerbation of oxidative stress in cardiomyocytes, while overexpression of ACAA2 reversed these effects. The transcription factor FOXO4 was found to regulate lipid peroxidation by modulating ACAA2, and knocking down FOXO4 partially restored the expression of ACAA2, reducing oxidative stress in cardiomyocytes. Furthermore, exogenous CSNO effectively restored the expression of ACAA2 and reduced the level of FOXO4, thereby mitigating lipid peroxidation and improving cardiac function. Therefore, in the context of renal insufficiency, regulating the FOXO4-ACAA2 axis through CSNO inhalation therapy may provide a novel therapeutic strategy for alleviating myocardial lipid peroxidation and improving cardiac function.
Collapse
Affiliation(s)
- Zhengqi Xu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
| | - Feng Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
| | - Xiaofan Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
| | - Bowen Ren
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
| | - Cuntai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China;
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China;
| | - Sheng Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China;
| |
Collapse
|
2
|
Pan L, Yin C, Jin KJ, Huang CX, Wang X, Wang Y, Rinkiko S, Jia JG, Zhang GP, Zhu YZ, Dai YX, Zou YZ, Gong H. Low-density lipoprotein receptor-related protein 6 ameliorates cardiac hypertrophy by regulating CTSD/HSP90α signaling during pressure overload. Acta Pharmacol Sin 2025; 46:606-617. [PMID: 39779966 PMCID: PMC11845692 DOI: 10.1038/s41401-024-01415-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/21/2024] [Indexed: 01/11/2025]
Abstract
Pressure overload induces pathological cardiac remodeling, including cardiac hypertrophy and fibrosis, resulting in cardiac dysfunction or heart failure. Recently, we observed that the low-density lipoprotein receptor-related protein 6 (LRP6), has shown potential in enhancing cardiac function by mitigating cardiac fibrosis in a mouse model subjected to pressure overload. In this study, we investigated the role of LRP6 as a potential modulator of pressure overload-induced cardiac hypertrophy and elucidated the underlying molecular mechanisms. We performed transverse aortic constriction (TAC) to induce pressure overload in cardiomyocyte-specific LRP6 overexpression mice (LRP6-over mice) and in control mice (α-myosin heavy chain (α-MHC) Mer-Cre-Mer Tg mice or named MCM mice). Cardiac function and hypertrophy were assessed using echocardiography. LRP6-over mice showed improved cardiac function and reduced hypertrophy after TAC, compared with MCM mice. We also applied mechanical stretch to cultured neonatal rat cardiomyocytes to model pressure overload in vitro. Mass spectrometry analysis showed that LRP6 interacts with HSP90α and cathepsin D (CTSD) in cardiomyocytes under mechanical stress. Further analysis demonstrated that LRP6 facilitates CTSD-mediated degradation of HSP90α, consequently inhibiting β-catenin activation and reducing cardiac hypertrophy post-TAC. Treatment with recombinant HSP90α protein or the CTSD inhibitor, pepstatin A, partly abolished the protective effect of LRP6 overexpression on myocardial hypertrophy and cardiac function after TAC in mice. Collectively, our data suggest that LRP6 protects against pressure overload-induced myocardial remodeling and that the CTSD/HSP90α/β-catenin axis may be a potential therapeutic target.
Collapse
Affiliation(s)
- Le Pan
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chao Yin
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Ke-Jia Jin
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chen-Xing Huang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiang Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Ying Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Suguro Rinkiko
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macao, China
| | - Jian-Guo Jia
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Guo-Ping Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yi-Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macao, China
| | - Yu-Xiang Dai
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Yun-Zeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, State Key Laboratory of Cardiovascular Diseases, NHC Key Laboratory of Ischemic Heart Diseases, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Pan L, Huang C, Jin X, Wu J, Jin K, Lin J, Wang Y, Li J, Yin C, Wang X, Zhang L, Zhang G, Dong H, Guo J, Komuro I, Dai Y, Zou Y, Gong H. Cardiac secreted HSP90α exacerbates pressure overload myocardial hypertrophy and heart failure. Redox Biol 2025; 79:103466. [PMID: 39721497 PMCID: PMC11732234 DOI: 10.1016/j.redox.2024.103466] [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: 11/04/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Sustained myocardial hypertrophy or left ventricular hypertrophy (LVH) triggered by pressure overload is strongly linked to adverse cardiovascular outcomes. Here, we investigated the clinical relationship between serum HSP90α (an isoform of HSP90) levels and LVH in patients with hypertension or aortic stenosis (AS) and explored underlying mechanisms in pressure overload mouse model. We built a pressure overload mouse model via transverse aortic constriction (TAC). Compared to controls, elevated serum HSP90α levels were observed in patients with hypertension or AS, and the levels positively correlated with LVH. Similarly, HSP90α levels increased in heart tissues from patients with obstructive hypertrophic cardiomyopathy (HCM), and in mice post-TAC. TAC induced the enhanced cardiac expression and secretion of HSP90α from cardiomyocytes and cardiac fibroblasts. Knockdown of HSP90α or blockade of extracellular HSP90α (eHSP90α) attenuated cardiac hypertrophy and dysfunction by inhibition of β-catenin/TCF7 signaling under pressure overload. Further analysis revealed that eHSP90α interacted with EC1-EC2 region of N-cadherin to activate β-catenin, enhancing the transcription of hypertrophic genes by TCF7, resulting in cardiac hypertrophy and dysfunction under pressure overload. These insights suggest the therapeutic potential of targeting HSP90α-initiated signaling pathway against cardiac hypertrophy and heart failure under pressure overload.
Collapse
Affiliation(s)
- Le Pan
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenxing Huang
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xuejuan Jin
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, and Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Kejia Jin
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jingyi Lin
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Ying Wang
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jianxuan Li
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chao Yin
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiang Wang
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Lei Zhang
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Guoping Zhang
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Hangming Dong
- Department of Respiratory and Critical Care Medicine, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Junli Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, 571199, China
| | - Issei Komuro
- Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Yuxiang Dai
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, and Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, and Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China.
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, and Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China.
| |
Collapse
|
4
|
Reddy T, Puri A, Guzman-Rojas L, Thomas C, Qian W, Zhou J, Zhao H, Mahboubi B, Oo A, Cho YJ, Kim B, Thaiparambil J, Rosato R, Martinez KO, Chervo MF, Ayerbe C, Giese N, Wink D, Lockett S, Wong S, Chang J, Krishnamurthy S, Yam C, Moulder S, Piwnica-Worms H, Meric-Bernstam F, Chang J. NOS inhibition sensitizes metaplastic breast cancer to PI3K inhibition and taxane therapy via c-JUN repression. Nat Commun 2024; 15:10737. [PMID: 39737957 PMCID: PMC11685991 DOI: 10.1038/s41467-024-54651-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/19/2024] [Indexed: 01/01/2025] Open
Abstract
Metaplastic breast cancer (MpBC) is a highly chemoresistant subtype of breast cancer with no standardized therapy options. A clinical study in anthracycline-refractory MpBC patients suggested that nitric oxide synthase (NOS) inhibitor NG-monomethyl-l-arginine (L-NMMA) may augment anti-tumor efficacy of taxane. We report that NOS blockade potentiated response of human MpBC cell lines and tumors to phosphoinositide 3-kinase (PI3K) inhibitor alpelisib and taxane. Mechanistically, NOS blockade leads to a decrease in the S-nitrosylation of c-Jun NH2-terminal kinase (JNK)/c-Jun complex to repress its transcriptional output, leading to enhanced tumor differentiation and associated chemosensitivity. As a result, combined NOS and PI3K inhibition with taxane targets MpBC stem cells and improves survival in patient-derived xenograft models relative to single-/dual-agent therapy. Similarly, biopsies from MpBC tumors that responded to L-NMMA+taxane therapy showed a post-treatment reversal of epithelial-to-mesenchymal transition and decreased stemness. Our findings suggest that combined inhibition of iNOS and PI3K is a unique strategy to decrease chemoresistance and improve clinical outcomes in MpBC.
Collapse
Affiliation(s)
- Tejaswini Reddy
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | - Akshjot Puri
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | | | - Christoforos Thomas
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | - Wei Qian
- Houston Methodist Research Institute, Houston, TX, USA
| | - Jianying Zhou
- Houston Methodist Research Institute, Houston, TX, USA
| | - Hong Zhao
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | - Bijan Mahboubi
- Adams School of Dentistry, University of North Carolina, Chapel Hill, USA
| | - Adrian Oo
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Young-Jae Cho
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Baek Kim
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | | | | | | | | | - Camila Ayerbe
- McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| | - Noah Giese
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | - David Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institute of Health, Frederick, MD, USA
| | - Stephen Lockett
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Stephen Wong
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | - Jeffrey Chang
- McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Clinton Yam
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Jenny Chang
- Houston Methodist Research Institute, Houston, TX, USA.
- Houston Methodist Neal Cancer Center, Houston, TX, USA.
| |
Collapse
|
5
|
Liu Y, Liu X, Pan C. Advances in Factors Affecting ALDH2 Activity and its Mechanisms. Cardiovasc Toxicol 2024; 24:1428-1438. [PMID: 39365551 DOI: 10.1007/s12012-024-09923-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/15/2024] [Indexed: 10/05/2024]
Abstract
Aldehyde dehydrogenase 2 (ALDH2) is a mitochondrial enzyme primarily involved in the detoxification of alcohol-derived aldehyde and endogenous toxic aldehydes. It exhibits widespread expression across various organs and exerts a broad and significant impact on diverse acute cardiovascular diseases, including acute coronary syndrome, acute aortic dissection, hypoxic pulmonary hypertension, and heart failure. The ALDH2 rs671 variant represents the most prevalent genetic variant in East Asian populations, with carriage rates ranging from 30 to 50% among the Chinese population. Given its widespread presence in the body, the wide range of diseases it affects, and its high rate of variation, it can serve as a crucial tool for the precise prevention and treatment of acute cardiovascular diseases, while offering individualized medication guidance. This review aims to provide a comprehensive overview of the latest advancements in factors affecting ALDH2 activity, encompassing post-transcriptional modifications, modulators of ALDH2, and relevant clinical drugs.
Collapse
Affiliation(s)
- Yun Liu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
- Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xuemei Liu
- Department of Nephrology, The Fifth People's Hospital of Jinan, Jinan, 250022, China
| | - Chang Pan
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China.
- Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
6
|
Duong LD, West JD, Morano KA. Redox regulation of proteostasis. J Biol Chem 2024; 300:107977. [PMID: 39522946 DOI: 10.1016/j.jbc.2024.107977] [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: 07/02/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Oxidants produced through endogenous metabolism or encountered in the environment react directly with reactive sites in biological macromolecules. Many proteins, in particular, are susceptible to oxidative damage, which can lead to their altered structure and function. Such structural and functional changes trigger a cascade of events that influence key components of the proteostasis network. Here, we highlight recent advances in our understanding of how cells respond to the challenges of protein folding and metabolic alterations that occur during oxidative stress. Immediately after an oxidative insult, cells selectively block the translation of most new proteins and shift molecular chaperones from folding to a holding role to prevent wholesale protein aggregation. At the same time, adaptive responses in gene expression are induced, allowing for increased expression of antioxidant enzymes, enzymes that carry out the reduction of oxidized proteins, and molecular chaperones, all of which serve to mitigate oxidative damage and rebalance proteostasis. Likewise, concomitant activation of protein clearance mechanisms, namely proteasomal degradation and particular autophagic pathways, promotes the degradation of irreparably damaged proteins. As oxidative stress is associated with inflammation, aging, and numerous age-related disorders, the molecular events described herein are therefore major determinants of health and disease.
Collapse
Affiliation(s)
- Long Duy Duong
- Department of Microbiology & Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - James D West
- Biochemistry & Molecular Biology Program, Departments of Biology and Chemistry, The College of Wooster, Wooster, Ohio, USA.
| | - Kevin A Morano
- Department of Microbiology & Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.
| |
Collapse
|
7
|
Zhang T, Lei J, Zheng M, Wen Z, Zhou J. Nitric oxide facilitates the S-nitrosylation and deubiquitination of Notch1 protein to maintain cancer stem cells in human NSCLC. J Cell Mol Med 2024; 28:e70203. [PMID: 39523215 PMCID: PMC11550923 DOI: 10.1111/jcmm.70203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/19/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related mortality, with tumour heterogeneity, fueled by cancer stem cells (CSCs), intricately linked to treatment resistance. Therefore, it is imperative to advance therapeutic strategies targeting CSCs in NSCLC. In this study, we utilized RNA sequencing to investigate metabolic pathway alterations in NSCLC CSCs and identified a crucial role of nitric oxide (NO) metabolism in governing CSC stemness, primarily through modulation of the Notch1 protein. Mechanistically, NO-induced S-nitrosylation of Notch1 facilitated its interaction with the deubiquitylase UCHL1, leading to increased Notch1 protein stability and enhanced CSC stemness. By inhibiting NO synthesis and downregulating UCHL1 expression, we validated the impact of NO on the Notch signalling pathway and CSC stemness. Importantly, targeting NO effectively reduced CSC populations within patient-derived organoids (PDOs) during radiotherapy. This mechanism presents a promising therapeutic target to surmount radiotherapy resistance in NSCLC treatment.
Collapse
Affiliation(s)
- Tenglong Zhang
- Department of Radiation OncologyThe First Affiliated Hospital of Soochow University, Soochow UniversitySuzhouChina
- Department of OncologyQingdao Municipal HospitalQingdaoChina
| | - Jiaxin Lei
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
| | - Ming Zheng
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
| | - Zhenke Wen
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow UniversitySoochow UniversitySuzhouChina
| | - Juying Zhou
- Department of Radiation OncologyThe First Affiliated Hospital of Soochow University, Soochow UniversitySuzhouChina
| |
Collapse
|
8
|
Meng C, Su H, Shu M, Shen F, Lu Y, Wu S, Su Z, Yu M, Yang D. The functional role of m6A demethylase ALKBH5 in cardiomyocyte hypertrophy. Cell Death Dis 2024; 15:683. [PMID: 39294131 PMCID: PMC11410975 DOI: 10.1038/s41419-024-07053-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Cardiomyocyte hypertrophy is a major outcome of pathological cardiac hypertrophy. The m6A demethylase ALKBH5 is reported to be associated with cardiovascular diseases, whereas the functional role of ALKBH5 in cardiomyocyte hypertrophy remains confused. We engineered Alkbh5 siRNA (siAlkbh5) and Alkbh5 overexpressing plasmid (Alkbh5 OE) to transfect cardiomyocytes. Subsequently, RNA immunoprecipitation (RIP)-qPCR, MeRIP-qPCR analysis and the dual-luciferase reporter assays were applied to elucidate the regulatory mechanism of ALKBH5 on cardiomyocyte hypertrophy. Our study identified ALKBH5 as a new contributor of cardiomyocyte hypertrophy. ALKBH5 showed upregulation in both phenylephrine (PE)-induced cardiomyocyte hypertrophic responses in vitro and transverse aortic constriction (TAC)/high fat diet (HFD)-induced pathological cardiac hypertrophy in vivo. Knockdown or overexpression of ALKBH5 regulated the occurrence of hypertrophic responses, including the increased cardiomyocyte surface areas and elevation of the hypertrophic marker levels, such as brain natriuretic peptide (BNP) and atrial natriuretic peptide (ANP). Mechanically, we indicated that ALKBH5 activated JAK2/STAT3 signaling pathway and mediated m6A demethylation on Stat3 mRNA, but not Jak2 mRNA, resulting in the phosphorylation and nuclear translocation of STAT3, which enhances the transcription of hypertrophic genes (e.g., Nppa) and ultimately leads to the emergence of cardiomyocytes hypertrophic growth. Our work highlights the functional role of ALKBH5 in regulating the onset of cardiomyocyte hypertrophy and provides a potential target for hypertrophic heart diseases prevention and treatment. ALKBH5 activated JAK2/STAT3 signaling pathway and mediated m6A demethylation on Stat3 mRNA, but not Jak2 mRNA, resulting in the phosphorylation and nuclear translocation of STAT3, which enhances the transcription of hypertrophic genes (e.g., Nppa) and ultimately leads to the emergence of cardiomyocytes hypertrophic growth.
Collapse
Affiliation(s)
- Chen Meng
- Human Phenome Institute, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, Zhangjiang Fudan International Innovation Center, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China
| | - Haibi Su
- Human Phenome Institute, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, Zhangjiang Fudan International Innovation Center, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China
| | - Meiling Shu
- Human Phenome Institute, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, Zhangjiang Fudan International Innovation Center, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China
| | - Feng Shen
- Department of Cardiovascular Surgery, Shanghai General Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, China
| | - Yijie Lu
- Human Phenome Institute, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, Zhangjiang Fudan International Innovation Center, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China
| | - Shishi Wu
- Human Phenome Institute, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, Zhangjiang Fudan International Innovation Center, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China
| | - Zhenghua Su
- Human Phenome Institute, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, Zhangjiang Fudan International Innovation Center, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China
| | - Mengyao Yu
- Human Phenome Institute, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, Zhangjiang Fudan International Innovation Center, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China.
| | - Di Yang
- Human Phenome Institute, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, Zhangjiang Fudan International Innovation Center, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Liu Z, Bian X, Li L, Liu L, Feng C, Wang Y, Ni J, Li S, Lu D, Li Y, Ma C, Yu T, Xiao X, Xue N, Wang Y, Zhang C, Ma X, Gao X, Fan X, Liu X, Fan G. SENP1-Mediated HSP90ab1 DeSUMOylation in Cardiomyocytes Prevents Myocardial Fibrosis by Paracrine Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400741. [PMID: 38992961 PMCID: PMC11425837 DOI: 10.1002/advs.202400741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/06/2024] [Indexed: 07/13/2024]
Abstract
Myocardial infarction (MI) triggers a poor ventricular remodeling response, but the underlying mechanisms remain unclear. Here, the authors show that sentrin-specific protease 1 (SENP1) is downregulated in post-MI mice and in patients with severe heart failure. By generating cardiomyocyte-specific SENP1 knockout and overexpression mice to assess cardiac function and ventricular remodeling responses under physiological and pathological conditions. Increased cardiac fibrosis in the cardiomyocyte-specific SENP1 deletion mice, associated with increased fibronectin (Fn) expression and secretion in cardiomyocytes, promotes fibroblast activation in response to myocardial injury. Mechanistically, SENP1 deletion in mouse cardiomyocytes increases heat shock protein 90 alpha family class B member 1 (HSP90ab1) SUMOylation with (STAT3) activation and Fn secretion after ventricular remodeling initiated. Overexpression of SENP1 or mutation of the HSP90ab1 Lys72 ameliorates adverse ventricular remodeling and dysfunction after MI. Taken together, this study identifies SENP1 as a positive regulator of cardiac repair and a potential drug target for the treatment of MI. Inhibition of HSP90ab1 SUMOylation stabilizes STAT3 to inhibit the adverse ventricular remodeling response.
Collapse
Affiliation(s)
- Zhihao Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Xiyun Bian
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Lan Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Li Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Chao Feng
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, 300051, China
| | - Ying Wang
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Sheng Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Dading Lu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Yanxia Li
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Tian Yu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Xiaolin Xiao
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Na Xue
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Yuxiang Wang
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Chunyan Zhang
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Xiaofang Ma
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Xiumei Gao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Tianjin fifth Central Hospital, Tianjin, 300450, China
- Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| |
Collapse
|
10
|
Chen Y, Li S, Guan B, Yan X, Huang C, Du Y, Yang F, Zhang N, Li Y, Lu J, Wang J, Zhang J, Chen Z, Chen C, Kong X. MAP4K4 exacerbates cardiac microvascular injury in diabetes by facilitating S-nitrosylation modification of Drp1. Cardiovasc Diabetol 2024; 23:164. [PMID: 38724987 PMCID: PMC11084109 DOI: 10.1186/s12933-024-02254-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Dynamin-related protein 1 (Drp1) is a crucial regulator of mitochondrial dynamics, the overactivation of which can lead to cardiovascular disease. Multiple distinct posttranscriptional modifications of Drp1 have been reported, among which S-nitrosylation was recently introduced. However, the detailed regulatory mechanism of S-nitrosylation of Drp1 (SNO-Drp1) in cardiac microvascular dysfunction in diabetes remains elusive. The present study revealed that mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) was consistently upregulated in diabetic cardiomyopathy (DCM) and promoted SNO-Drp1 in cardiac microvascular endothelial cells (CMECs), which in turn led to mitochondrial dysfunction and cardiac microvascular disorder. Further studies confirmed that MAP4K4 promoted SNO-Drp1 at human C644 (mouse C650) by inhibiting glutathione peroxidase 4 (GPX4) expression, through which MAP4K4 stimulated endothelial ferroptosis in diabetes. In contrast, inhibition of MAP4K4 via DMX-5804 significantly reduced endothelial ferroptosis, alleviated cardiac microvascular dysfunction and improved cardiac dysfunction in db/db mice by reducing SNO-Drp1. In parallel, the C650A mutation in mice abolished SNO-Drp1 and the role of Drp1 in promoting cardiac microvascular disorder and cardiac dysfunction. In conclusion, our findings demonstrate that MAP4K4 plays an important role in endothelial dysfunction in DCM and reveal that SNO-Drp1 and ferroptosis activation may act as downstream targets, representing potential therapeutic targets for DCM.
Collapse
Affiliation(s)
- Yuqiong Chen
- Department of Cardiology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 215000, Suzhou, Jiangsu Province, China.
| | - Su Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, 200032, Shanghai, China
| | - Bo Guan
- Department of Geriatrics, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, China
| | - Xiaopei Yan
- Department of Respiratory Medicine, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, China
| | - Chao Huang
- Ministry of Science and Technology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, 215002, Suzhou, Jiangsu, China
| | - Yingqiang Du
- Department of Cardiology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 215000, Suzhou, Jiangsu Province, China
| | - Fan Yang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, 210008, Nanjing, China
- Branch of National Clinical Research Center for Metabolic Diseases, 210008, Nanjing, China
| | - Nannan Zhang
- Department of Cardiology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 215000, Suzhou, Jiangsu Province, China
| | - Yafei Li
- Department of Cardiology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 215000, Suzhou, Jiangsu Province, China
| | - Jian Lu
- Department of Critical Care Medicine, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jiankang Wang
- Department of Cardiology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 215000, Suzhou, Jiangsu Province, China
| | - Jun Zhang
- Department of Cardiology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 215000, Suzhou, Jiangsu Province, China
| | - Zhangwei Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, 200032, Shanghai, China.
| | - Chao Chen
- Department of Cardiology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 215000, Suzhou, Jiangsu Province, China.
| | - Xiangqing Kong
- Department of Cardiology, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, 215000, Suzhou, Jiangsu Province, China.
- Department of Cardiology, Gulou District, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing City, Jiangsu Province, China.
| |
Collapse
|
11
|
Zhan X, Yang Y, Li Q, He F. The role of deubiquitinases in cardiac disease. Expert Rev Mol Med 2024; 26:e3. [PMID: 38525836 PMCID: PMC11062144 DOI: 10.1017/erm.2024.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/10/2023] [Accepted: 12/28/2023] [Indexed: 03/26/2024]
Abstract
Deubiquitinases are a group of proteins that identify and digest monoubiquitin chains or polyubiquitin chains attached to substrate proteins, preventing the substrate protein from being degraded by the ubiquitin-proteasome system. Deubiquitinases regulate cellular autophagy, metabolism and oxidative stress by acting on different substrate proteins. Recent studies have revealed that deubiquitinases act as a critical regulator in various cardiac diseases, and control the onset and progression of cardiac disease through a board range of mechanism. This review summarizes the function of different deubiquitinases in cardiac disease, including cardiac hypertrophy, myocardial infarction and diabetes mellitus-related cardiac disease. Besides, this review briefly recapitulates the role of deubiquitinases modulators in cardiac disease, providing the potential therapeutic targets in the future.
Collapse
Affiliation(s)
- Xiaona Zhan
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yi Yang
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Qing Li
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Fan He
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
12
|
Luo S, Ye D, Wang Y, Liu X, Wang X, Xie L, Ji Y. Roles of Protein S-Nitrosylation in Endothelial Homeostasis and Dysfunction. Antioxid Redox Signal 2024; 40:186-205. [PMID: 37742108 DOI: 10.1089/ars.2023.0406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/25/2023]
Abstract
Significance: Nitric oxide (NO) plays several distinct roles in endothelial homeostasis. Except for activating the guanylyl cyclase enzyme-dependent cyclic guanosine monophosphate signaling pathway, NO can bind reactive cysteine residues in target proteins, a process known as S-nitrosylation (SNO). SNO is proposed to explain the multiple biological functions of NO in the endothelium. Investigating the targets and mechanism of protein SNO in endothelial cells (ECs) can provide new strategies for treating endothelial dysfunction-related diseases. Recent Advances: In response to different environments, proteomics has identified multiple SNO targets in ECs. Functional studies confirm that SNO regulates NO bioavailability, inflammation, permeability, oxidative stress, mitochondrial function, and insulin sensitivity in ECs. It also influences EC proliferation, migration, apoptosis, and transdifferentiation. Critical Issues: Single-cell transcriptomic analysis of ECs isolated from different mouse tissues showed heterogeneous gene signatures. However, litter research focuses on the heterogeneous properties of SNO proteins in ECs derived from different tissues. Although metabolism reprogramming plays a vital role in endothelial functions, little is known about how protein SNO regulates metabolism reprogramming in ECs. Future Directions: Precisely deciphering the effects of protein SNO in ECs isolated from different tissues under different conditions is necessary to further characterize the relationship between protein SNO and endothelial dysfunction-related diseases. In addition, identifying SNO targets that can influence endothelial metabolic reprogramming and the underlying mechanism can offer new views on the crosstalk between metabolism and post-translational protein modification. Antioxid. Redox Signal. 40, 186-205.
Collapse
Affiliation(s)
- Shanshan Luo
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Danyu Ye
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Yu Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Xingeng Liu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Xiaoqian Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Liping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Key Laboratory of Cardiovascular Medicine Research and Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, NHC Key Laboratory of Cell Transplantation, the Central Laboratory of the First Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| |
Collapse
|
13
|
Zhang A, Li H, Song Q, Cui Y, Zhang Y, Wang X, Li Z, Hou Y. High-fat stimulation induces atrial neural remodeling by reducing NO production via the CRIF1/eNOS/P21 axi. Lipids Health Dis 2023; 22:189. [PMID: 37932729 PMCID: PMC10629039 DOI: 10.1186/s12944-023-01952-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/18/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Autonomic remodeling of the atria plays a pivotal role in the development of atrial fibrillation (AF) and exerts a substantial influence on the progression of this condition. Hyperlipidemia is a predisposing factor for AF, but its effect on atrial nerve remodeling is unclear. The primary goal of this study was to explore the possible mechanisms through which the consumption of a high-fat diet (HFD) induces remodeling of atrial nerves, and to identify novel targets for clinical intervention. METHODS Cell models were created in vitro by subjecting cells to palmitic acid (PA), while rat models were established by feeding them a high-fat diet. To investigate the interplay between cardiomyocytes and nerve cells in a co-culture system, we utilized Transwell cell culture plates featuring a pore size of 0.4 μm. The CCK-8 assay was employed to determine cell viability, fluorescent probe DCFH-DA and flow cytometry were utilized for measuring ROS levels, JC-1 was used to assess the mitochondrial membrane potential, the Griess method was employed to measure the nitric oxide (NO) level in the supernatant, a fluorescence-based method was used to measure ATP levels, and MitoTracker was utilized for assessing mitochondrial morphology. The expression of pertinent proteins was evaluated using western blotting (WB) and immunohistochemistry techniques. SNAP was used to treat nerve cells in order to replicate a high-NO atmosphere, and the level of nitroso was assessed using the iodoTMT reagent labeling method. RESULTS The study found that cardiomyocytes' mitochondrial morphology and function were impaired under high-fat stimulation, affecting nitric oxide (NO) production through the CRIF1/SIRT1/eNOS axis. In a coculture model, overexpression of eNOS in cardiomyocytes increased NO expression. Moreover, the increased Keap1 nitrosylation within neuronal cells facilitated the entry of Nrf2 into the nucleus, resulting in an augmentation of P21 transcription and a suppression of proliferation. Atrial neural remodeling occurred in the HFD rat model and was ameliorated by increasing myocardial tissue eNOS protein expression with trimetazidine (TMZ). CONCLUSIONS Neural remodeling is triggered by high-fat stimulation, which decreases the production of NO through the CRIF1/eNOS/P21 axis. Additionally, TMZ prevents neural remodeling and reduces the occurrence of AF by enhancing eNOS expression.
Collapse
Affiliation(s)
- An Zhang
- Department of Cardiology, Cheeloo College of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Huilin Li
- Department of Emergency Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong, China
| | - Qiyuan Song
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Yansong Cui
- Department of Cardiology, Cheeloo College of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yujiao Zhang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Ximin Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Zhan Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University, Shandong, China.
| | - Yinglong Hou
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University, Shandong, China.
- Department of Cardiology, Cheeloo College of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China.
| |
Collapse
|
14
|
Tang X, Zhao S, Liu J, Liu X, Sha X, Huang C, Hu L, Sun S, Gao Y, Chen H, Zhang Z, Wang D, Gu Y, Chen S, Wang L, Gu A, Chen F, Pu J, Chen X, Yu B, Xie L, Huang Z, Han Y, Ji Y. Mitochondrial GSNOR Alleviates Cardiac Dysfunction via ANT1 Denitrosylation. Circ Res 2023; 133:220-236. [PMID: 37377022 DOI: 10.1161/circresaha.123.322654] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND The cardiac-protective role of GSNOR (S-nitrosoglutathione reductase) in the cytoplasm, as a denitrosylase enzyme of S-nitrosylation, has been reported in cardiac remodeling, but whether GSNOR is localized in other organelles and exerts novel effects remains unknown. We aimed to elucidate the effects of mitochondrial GSNOR, a novel subcellular localization of GSNOR, on cardiac remodeling and heart failure (HF). METHODS GSNOR subcellular localization was observed by cellular fractionation assay, immunofluorescent staining, and colloidal gold particle staining. Overexpression of GSNOR in mitochondria was achieved by mitochondria-targeting sequence-directed adeno-associated virus 9. Cardiac-specific knockout of GSNOR mice was used to examine the role of GSNOR in HF. S-nitrosylation sites of ANT1 (adenine nucleotide translocase 1) were identified using biotin-switch and liquid chromatography-tandem mass spectrometry. RESULTS GSNOR expression was suppressed in cardiac tissues of patients with HF. Consistently, cardiac-specific knockout mice showed aggravated pathological remodeling induced by transverse aortic constriction. We found that GSNOR is also localized in mitochondria. In the angiotensin II-induced hypertrophic cardiomyocytes, mitochondrial GSNOR levels significantly decreased along with mitochondrial functional impairment. Restoration of mitochondrial GSNOR levels in cardiac-specific knockout mice significantly improved mitochondrial function and cardiac performance in transverse aortic constriction-induced HF mice. Mechanistically, we identified ANT1 as a direct target of GSNOR. A decrease in mitochondrial GSNOR under HF leads to an elevation of S-nitrosylation ANT1 at cysteine 160 (C160). In accordance with these findings, overexpression of either mitochondrial GSNOR or ANT1 C160A, non-nitrosylated mutant, significantly improved mitochondrial function, maintained the mitochondrial membrane potential, and upregulated mitophagy. CONCLUSIONS We identified a novel species of GSNOR localized in mitochondria and found mitochondrial GSNOR plays an essential role in maintaining mitochondrial homeostasis through ANT1 denitrosylation, which provides a potential novel therapeutic target for HF.
Collapse
Affiliation(s)
- Xin Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Shuang Zhao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Jieqiong Liu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Xiameng Liu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Xinqi Sha
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Changgao Huang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Lulu Hu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Shixiu Sun
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Yuanqing Gao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Institute of Cardiothoracic Vascular Disease, Nanjing University, China (D.W., Y.G.)
| | - Hongshan Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Zhiren Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Key Laboratory of Cardiovascular Medicine Research and Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, NHC Key Laboratory of Cell Transplantation, the Central Laboratory of the First Affiliated Hospital (Z.Z., Y.J.), Harbin Medical University, Heilongjiang, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Institute of Cardiothoracic Vascular Disease, Nanjing University, China (D.W., Y.G.)
| | - Yuexi Gu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital (S.C.), Nanjing Medical University, Jiangsu, China
| | - Liansheng Wang
- Department of Cardiology (L.W.), First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Gu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health (A.G.), Nanjing Medical University, Jiangsu, China
| | - Feng Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Forensic Medicine (F.C.), Nanjing Medical University, Jiangsu, China
| | - Jun Pu
- Division of Cardiology, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, China (J.P.)
| | - Xin Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital (X.C.), Nanjing Medical University, Jiangsu, China
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Ministry of Education (B.Y.), Harbin Medical University, Heilongjiang, China
| | - Liping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
| | - Zhengrong Huang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, China (Z.H.)
| | - Yi Han
- Department of Geriatrics (Y.H.), First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School (X.T., S.Z., J.L., X.L., X.S., C.H., L.H., S.S., Y.G., H.C., L.X., Y.J.), Nanjing Medical University, Jiangsu, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Key Laboratory of Cardiovascular Medicine Research and Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, NHC Key Laboratory of Cell Transplantation, the Central Laboratory of the First Affiliated Hospital (Z.Z., Y.J.), Harbin Medical University, Heilongjiang, China
| |
Collapse
|
15
|
Zhong Q, Xiao X, Qiu Y, Xu Z, Chen C, Chong B, Zhao X, Hai S, Li S, An Z, Dai L. Protein posttranslational modifications in health and diseases: Functions, regulatory mechanisms, and therapeutic implications. MedComm (Beijing) 2023; 4:e261. [PMID: 37143582 PMCID: PMC10152985 DOI: 10.1002/mco2.261] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Protein posttranslational modifications (PTMs) refer to the breaking or generation of covalent bonds on the backbones or amino acid side chains of proteins and expand the diversity of proteins, which provides the basis for the emergence of organismal complexity. To date, more than 650 types of protein modifications, such as the most well-known phosphorylation, ubiquitination, glycosylation, methylation, SUMOylation, short-chain and long-chain acylation modifications, redox modifications, and irreversible modifications, have been described, and the inventory is still increasing. By changing the protein conformation, localization, activity, stability, charges, and interactions with other biomolecules, PTMs ultimately alter the phenotypes and biological processes of cells. The homeostasis of protein modifications is important to human health. Abnormal PTMs may cause changes in protein properties and loss of protein functions, which are closely related to the occurrence and development of various diseases. In this review, we systematically introduce the characteristics, regulatory mechanisms, and functions of various PTMs in health and diseases. In addition, the therapeutic prospects in various diseases by targeting PTMs and associated regulatory enzymes are also summarized. This work will deepen the understanding of protein modifications in health and diseases and promote the discovery of diagnostic and prognostic markers and drug targets for diseases.
Collapse
Affiliation(s)
- Qian Zhong
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xina Xiao
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Yijie Qiu
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Zhiqiang Xu
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Chunyu Chen
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Baochen Chong
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xinjun Zhao
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Shan Hai
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Shuangqing Li
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Zhenmei An
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Lunzhi Dai
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
16
|
Cai Y, Jiang S, Huang C, Shen A, Zhang X, Yang W, Xiao Y, Gao S, Du R, Zheng G, Yan T, Craig Wan C. Baicalin inhibits pressure overload-induced cardiac hypertrophy by regulating the SIRT3-dependent signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154747. [PMID: 36931095 DOI: 10.1016/j.phymed.2023.154747] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/06/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The conserved sirtuin protein sirtuin 3 (SIRT3) is a vital protective protein for cardiac hypertrophy. Inhibition of SIRT3 accelerated the development of heart hypertrophy. On the other hand, myocardial hypertrophy was prevented by overexpressing SIRT3. SIRT3 has been proposed as a potential therapeutic target for managing or averting heart hypertrophy. Baicalin, a flavonoid extracted from the Scutellaria baicalensis plant, has anti-cardiovascular properties, including protection against cardiac hypertrophy. However, the molecular mechanism of the anti-hypertrophic effect of baicalin is not well known. PURPOSE In this study, we aim to investigate the effect of baicalin on cardiac hypertrophy and explored its underlying molecular mechanisms. STUDY-DESIGN/METHODS Abdominal aortic constriction (AAC)-induced mouse cardiac hypertrophy and angiotensin II (Ang II)-induced cardiomyocyte hypertrophy models were established. After baicalin treatment, cardiac hypertrophy was monitored by detecting the expression of hypertrophic genes and cell surface area. Echocardiogram was performed to check the heart function in vivo. Moreover, the protein expression of the SIRT3-dependent pathway was detected by Western blotting. RESULTS In this work, we demonstrated that baicalin might suppress the cell surface area and the expression of the Ang II -induced myosin heavy chain β (β-MHC), brain natriuretic polypeptide (BNP), and atrial natriuretic factor (ANF). Additionally, it reduced the AAC rats' hypertrophic impact. We also found that baicalin prevents cardiac hypertrophy by regulating SIRT3/LKB1/AMPK signaling pathway. Moreover, we showed that baicalin upregulated the SIRT3 protein expression by inhibiting proteasome and by the activation of 20 S proteasome subunit beta type-5 (PSMB5). CONCLUSION These results offer the first proof that baicalin inhibits cardiac hypertrophy due to its effect on the SIRT3-dependent signaling pathway, indicating its potential for treating cardiac hypertrophy and heart failure. The present study provides a preliminary experimental basis for the clinical application of baicalin and baicalin-like compounds.
Collapse
Affiliation(s)
- Yi Cai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Shisheng Jiang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Chaoming Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Ao Shen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xuan Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Wanling Yang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Yichuan Xiao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Shuhan Gao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Rong Du
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Guodong Zheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| | - Tingdong Yan
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China.
| | - Chunpeng Craig Wan
- Research Center of Tea and Tea Culture, College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
17
|
Sumi MP, Tupta B, Roychowdhury S, Comhair S, Asosingh K, Stuehr DJ, Erzurum SC, Ghosh A. Hemoglobin resident in the lung epithelium is protective for smooth muscle soluble guanylate cyclase function. Redox Biol 2023; 63:102717. [PMID: 37120930 PMCID: PMC10172757 DOI: 10.1016/j.redox.2023.102717] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/02/2023] Open
Abstract
Hemoglobin (Hb) present in the lung epithelium is of unknown significance. However Hb being an nitric oxide (NO) scavenger can bind to NO and reduce its deleterious effects. Hence we postulated an NO scavenging role for this lung Hb. Doing transwell co-culture with bronchial epithelial cells, A549/16-HBE (apical) and human airway smooth muscle cells (HASMCs as basal), we found that Hb can protect the smooth muscle soluble guanylyl cyclase (sGC) from excess NO. Inducing the apical A549/16-HBE cells with cytokines to trigger iNOS expression and NO generation caused a time dependent increase in SNO-sGC and this was accompanied with a concomitant drop in sGC-α1β1 heterodimerization. Silencing Hbαβ in the apical cells further increased the SNO on sGC with a faster drop in the sGC heterodimer and these effects were additive along with further silencing of thioredoxin 1 (Trx1). Since heme of Hb is critical for NO scavenging we determined the Hb heme in a mouse model of allergic asthma (OVA) and found that Hb in the inflammed OVA lungs was low in heme or heme-free relative to those of naïve lungs. Further we established a direct correlation between the status of the sGC heterodimer and the Hb heme from lung samples of human asthma, iPAH, COPD and cystic fibrosis. These findings present a new mechanism of protection of lung sGC by the epithelial Hb, and suggests that this protection maybe lost in asthma or COPD where lung Hb is unable to scavenge the NO due to it being heme-deprived.
Collapse
Affiliation(s)
- Mamta P Sumi
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Blair Tupta
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Sanjoy Roychowdhury
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Suzy Comhair
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Kewal Asosingh
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Serpil C Erzurum
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Arnab Ghosh
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
18
|
Jiao K, Su P, Feng Y, Li C. Bioinformatics analysis and identification of hub genes associated with female acute myocardial infarction patients by using weighted gene co-expression networks. Medicine (Baltimore) 2023; 102:e33634. [PMID: 37115066 PMCID: PMC10145720 DOI: 10.1097/md.0000000000033634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
To explore potential biomarkers of acute myocardial infarction (AMI) in females by using bioinformatics analysis. In this study, we explored potential biomarkers of AMI in females using bioinformatics analysis. We screened a total of 186 differentially expressed genes from the Gene Expression Omnibus. In the study, we found that weighted gene co-expression network analysis explored the co-expression network of genes and identified key modules. Simultaneously, we chose brown modules as key modules related to AMI. In this study, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis revealed that genes in the brown module were mainly enriched in "heparin" and 'complementation and coagulation cascade. Based on the protein-protein interaction network, we identified S100A9, mitogen-activated protein kinase (MAPK) 3, MAPK1, MMP3, interleukin (IL)-17A, and HSP90AB1 as hub gene sets. Whereas, polymerase chain reaction results showed that S100A9, MAPK3, MAPK1, MMP3, IL-17A, and HSP90AB1 were highly expressed compared with the control group. The IL-17 signaling pathway associated with an inflammatory response may be a potential biomarker and target for the treatment of women with myocardial infarction.
Collapse
Affiliation(s)
- Kun Jiao
- Ordos Central Hospital Cardiology Department, Ordos, China
| | - Ping Su
- Ordos Central Hospital Cardiology Department, Ordos, China
| | - Yubao Feng
- Ordos Central Hospital Cardiology Department, Ordos, China
| | - Changqing Li
- Ordos Central Hospital Cardiology Department, Ordos, China
| |
Collapse
|
19
|
Muronetz VI, Kudryavtseva SS, Leisi EV, Kurochkina LP, Barinova KV, Schmalhausen EV. Regulation by Different Types of Chaperones of Amyloid Transformation of Proteins Involved in the Development of Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23052747. [PMID: 35269889 PMCID: PMC8910861 DOI: 10.3390/ijms23052747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/28/2022] [Indexed: 02/06/2023] Open
Abstract
The review highlights various aspects of the influence of chaperones on amyloid proteins associated with the development of neurodegenerative diseases and includes studies conducted in our laboratory. Different sections of the article are devoted to the role of chaperones in the pathological transformation of alpha-synuclein and the prion protein. Information about the interaction of the chaperonins GroE and TRiC as well as polymer-based artificial chaperones with amyloidogenic proteins is summarized. Particular attention is paid to the effect of blocking chaperones by misfolded and amyloidogenic proteins. It was noted that the accumulation of functionally inactive chaperones blocked by misfolded proteins might cause the formation of amyloid aggregates and prevent the disassembly of fibrillar structures. Moreover, the blocking of chaperones by various forms of amyloid proteins might lead to pathological changes in the vital activity of cells due to the impaired folding of newly synthesized proteins and their subsequent processing. The final section of the article discusses both the little data on the role of gut microbiota in the propagation of synucleinopathies and prion diseases and the possible involvement of the bacterial chaperone GroE in these processes.
Collapse
Affiliation(s)
- Vladimir I. Muronetz
- Belozersky Institute of Physico Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.P.K.); (K.V.B.); (E.V.S.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia;
- Correspondence:
| | - Sofia S. Kudryavtseva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia;
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Evgeniia V. Leisi
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Lidia P. Kurochkina
- Belozersky Institute of Physico Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.P.K.); (K.V.B.); (E.V.S.)
| | - Kseniya V. Barinova
- Belozersky Institute of Physico Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.P.K.); (K.V.B.); (E.V.S.)
| | - Elena V. Schmalhausen
- Belozersky Institute of Physico Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.P.K.); (K.V.B.); (E.V.S.)
| |
Collapse
|
20
|
Zhao S, Tang X, Miao Z, Chen Y, Cao J, Song T, You D, Zhong Y, Lin Z, Wang D, Shi Z, Tang X, Wang D, Chen S, Wang L, Gu A, Chen F, Xie L, Huang Z, Wang H, Ji Y. Hsp90 S-nitrosylation at Cys521, as a conformational switch, modulates cycling of Hsp90-AHA1-CDC37 chaperone machine to aggravate atherosclerosis. Redox Biol 2022; 52:102290. [PMID: 35334246 PMCID: PMC8942817 DOI: 10.1016/j.redox.2022.102290] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/04/2022] [Accepted: 03/14/2022] [Indexed: 12/20/2022] Open
Abstract
Endothelial dysfunction is the initial process of atherosclerosis. Heat shock protein 90 (Hsp90), as a molecular chaperone, plays a crucial role in various cardiovascular diseases. Hsp90 function is regulated by S-nitrosylation (SNO). However, the precise role of SNO-Hsp90 in endothelial dysfunction during atherosclerosis remains unclear. We here identified Hsp90 as a highly S-nitrosylated target in endothelial cells (ECs) by biotin switch assay combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS). The elevation of SNO-Hsp90 was observed in atherosclerotic human and rodent aortas as well as in oxidized LDL (oxLDL)-treated ECs. Inhibition of inducible nitric oxide synthase (iNOS) or transfection with Hsp90 cysteine 521 (Cys521) mutation plasmid decreased the level of SNO-Hsp90 in oxLDL-cultured ECs. Coimmunoprecipitation and proximity ligation assay demonstrated that SNO-Hsp90 at Cys521 suppressed the interaction between Hsp90 and activator of Hsp90 ATPase activity 1 (AHA1), but promoted the association of Hsp90 and cell division cycle 37 (CDC37). Hsp90 Cys521 mutation increased endothelial nitric oxide synthase (eNOS) activity and inhibited nuclear factor kappa-B (NF-κB) signaling, thereby increasing nitric oxide (NO) bioavailability and alleviating endothelial adhesion, inflammation and oxidative stress in oxLDL-treated ECs. Also, administration of endothelial-specific adeno-associated viruses of Cys521-mutated Hsp90 significantly mitigated vascular oxidative stress, macrophage infiltration and atherosclerosis lesion areas in high fat diet-fed ApoE-/- mice. In conclusion, SNO-Hsp90 at Cys521, that serves as a conformational switch, disrupts Hsp90/AHA1 interaction but promotes recruitment of CDC37 to exacerbate atherosclerosis. Hsp90 S-nitrosylation at Cys521 acts as a conformational switch to modulate Hsp90/AHA1 and Hsp90/CDC37 interaction. SNO-Hsp90 induces endothelial adhesion, inflammation and oxidative stress. SNO-Hsp90 mediates endothelial dysfunction to exacerbate atherosclerosis.
Collapse
|