1
|
Chacar S, Abdrabou W, Al Hageh C, Ali L, Venkatachalam T, Zalloua P, Suleiman MS, Howarth FC, Khraibi AA, Nader M. Remodeling of the cardiac striatin interactome and its dynamics in the diabetic heart. Sci Rep 2025; 15:7384. [PMID: 40025125 PMCID: PMC11873221 DOI: 10.1038/s41598-025-91098-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 02/18/2025] [Indexed: 03/04/2025] Open
Abstract
Diabetic cardiomyopathy (DbCM) is a silent and complex condition involving numerous signaling pathways that impair cardiomyocyte metabolism and cardiac performance. Striatin (STRN) is a multifaceted protein that binds metabolic proteins, yet its role in diabetic heart remains unexplored. Here we characterized the cardiac STRN interactome by performing immunoprecipitation on left ventricle (LV) proteins from control and diabetic hearts (rats treated with streptozotocin for 24 weeks) to dissect its derivative protein complex. Diabetic rats exhibited pathological heart remodeling characterized by increased heart weight/body weight ratio, elevated levels of Atrial Natriuretic Factor (ANF), and altered expression of alpha and beta-myosin heavy chain isoforms. Notably, STRN expression mirrored that of the remodeling marker ANF across all cardiac chambers. Proteomic analysis yielded 247 proteins interacting with STRN exclusively in diabetic LV, 94 in both control and diabetic LV, and 11 only in control LV. STRN retained a higher interaction with some STRN interacting phosphatase and kinase complex (STRIPAK) proteins (i.e. protein phosphatase 2A (PP2A), and sarcolemmal associated membrane protein (SLMAP)) in diabetic LV, indicating a preserved role of this signalosome in diabetic settings. Functional enrichment and gene ontology revealed that the STRN interactome in diabetic LV carried signalosomes related to cardiac contractility, endoplasmic reticulum stress, mitochondrial function, and apoptotic processes. Western blot experiments confirmed the interaction between STRN and SLMAP in both control and diabetic heart. These data suggest a pivotal role for the STRN signalosome in cardiometabolic disorders, potentially paving the way for novel therapeutic management of DbCM. Targeting the STRN interactome in DbCM, mainly the first-line interactors SLMAP, PP2A, and Cav-1 may offer hope for patients with diabetes-induced cardiac injuries.
Collapse
Affiliation(s)
- Stephanie Chacar
- Department of Medical Sciences, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Wael Abdrabou
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Cynthia Al Hageh
- Department of Public Health and Epidemiology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Liaqat Ali
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Thenmozhi Venkatachalam
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Pierre Zalloua
- Department of Public Health and Epidemiology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Frank Christopher Howarth
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ali A Khraibi
- Department of Biomedical Engineering and Biotechnology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Moni Nader
- Department of Medical Sciences, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia.
| |
Collapse
|
2
|
Zadorozny L, Du J, Supanekar N, Annamalai K, Yu Q, Wang M. Caveolin and oxidative stress in cardiac pathology. Front Physiol 2025; 16:1550647. [PMID: 40041164 PMCID: PMC11876135 DOI: 10.3389/fphys.2025.1550647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
Caveolins interact with signaling molecules within caveolae and subcellular membranes. Dysregulation of caveolin function and protein abundance contributes to cardiac pathophysiological processes, driving the development and progression of heart disease. Reactive oxygen species (ROS) play a critical role in maintaining cellular homeostasis and are key contributors to the pathophysiological mechanisms of cardiovascular disorders. Caveolins have been shown to modulate oxidative stress and regulate redox homeostasis. However, the specific roles of caveolins, particularly caveolin-1 and caveolin-3, in regulating ROS production during cardiac pathology remain unclear. This mini-review article highlights the correlation between caveolins and oxidative stress in maintaining cardiovascular health and modulating cardiac diseases, specifically in myocardial ischemia, heart failure, diabetes-induced metabolic cardiomyopathy, and septic cardiomyopathy. A deeper understanding of caveolin-mediated mechanisms may pave the way for innovative therapeutic approaches to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Lauren Zadorozny
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jiayue Du
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Neil Supanekar
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Karthik Annamalai
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Qing Yu
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Meijing Wang
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
3
|
An Z, Tian J, Zhao X, Liu L, Yang X, Zhang M, Zhang L, Song X. Regulation of cardiovascular and cardiac functions by caveolins. FEBS J 2024; 291:3753-3761. [PMID: 37060249 DOI: 10.1111/febs.16798] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/16/2023]
Abstract
Caveolae are intracellular vesicles with diameters ranging from 50 to 100 nm. The role of caveolins in mediating oxidative stress, autophagy, apoptosis, fibrosis, and vascular remodeling has attracted increasing attention in cardiovascular therapy. Several studies have suggested that caveolin could be a therapeutic target for the treatment of cardiac and/or vascular injury via several pathophysiological mechanisms. Despite substantial advances in our understanding of the basic biology of vesicles over the past decade, the relevance and specific role of these mechanisms in cardiovascular homeostasis remains ambiguous. Here, we review the macroscopic role of caveolins in protecting cardiac function and, at the microscopic level, examine possible cardioprotective caveolar mechanisms, including their antioxidative stress, antiapoptosis, autophagy-regulatory, antifibrosis, and angiogenesis-promoting properties. We believe that the role of caveolins in cardiac functioning has not been fully elucidated and is an important line of future research with several cardioprotective implications.
Collapse
Affiliation(s)
- Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Xueyao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mingduo Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lijun Zhang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Wu Y, Peng W, Chen S, Zeng X, Zhu J, Zhu P. CAV1 Protein Encapsulated in Mouse BMSC-Derived Extracellular Vesicles Alleviates Myocardial Fibrosis Following Myocardial Infarction by Blocking the TGF-β1/SMAD2/c-JUN Axis. J Cardiovasc Transl Res 2024; 17:523-539. [PMID: 38092988 DOI: 10.1007/s12265-023-10472-9] [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: 06/28/2023] [Accepted: 11/27/2023] [Indexed: 07/03/2024]
Abstract
Extracellular vesicles (EVs) derived from mouse bone marrow mesenchymal stem cells (mBMSCs) convey the CAV1 protein, influencing the TGF-β1/SMAD2/c-JUN pathway and thus the molecular mechanisms underlying myocardial fibrosis (MF) post-myocardial infarction (MI). Through various experimental methods, including transmission electron microscopy, Nanosight analysis, Western blot, ELISA, and qRT-PCR, we isolated, purified, and identified EVs originating from mBMSCs. Bioinformatics and experimental findings show a reduced expression of CAV1 in myocardial fibrosis tissue. Furthermore, our findings suggest that mBMSC-EVs can deliver CAV1 to cardiac fibroblasts (CFs) and that silencing CAV1 in mBMSC-EVs promotes CF fibrosis. In vivo studies further corroborated these findings. In conclusion, mBMSC-EVs mitigate myocardial fibrosis in MI mice by delivering the CAV1 protein, inhibiting the TGF-β1/SMAD2/c-JUN pathway.
Collapse
Affiliation(s)
- Yijin Wu
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106 Zhongshan Er Road, Yuexiu District, Guangzhou, 510100, China
| | - Wenying Peng
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, China
| | - Siyao Chen
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, China
| | - Xiaodong Zeng
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, China
| | - Jiade Zhu
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106 Zhongshan Er Road, Yuexiu District, Guangzhou, 510100, China
| | - Ping Zhu
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106 Zhongshan Er Road, Yuexiu District, Guangzhou, 510100, China.
| |
Collapse
|
5
|
Li HX, Ma Y, Yan YX, Zhai XK, Xin MY, Wang T, Xu DC, Song YT, Song CD, Pan CX. The purified extract of steamed Panax ginseng protects cardiomyocyte from ischemic injury via caveolin-1 phosphorylation-mediating calcium influx. J Ginseng Res 2023; 47:755-765. [PMID: 38107394 PMCID: PMC10721475 DOI: 10.1016/j.jgr.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 12/19/2023] Open
Abstract
Background Caveolin-1, the scaffolding protein of cholesterol-rich invaginations, plays an important role in store-operated Ca2+ influx and its phosphorylation at Tyr14 (p-caveolin-1) is vital to mobilize protection against myocardial ischemia (MI) injury. SOCE, comprising STIM1, ORAI1 and TRPC1, contributes to intracellular Ca2+ ([Ca2+]i) accumulation in cardiomyocytes. The purified extract of steamed Panax ginseng (EPG) attenuated [Ca2+]i overload against MI injury. Thus, the aim of this study was to investigate the possibility of EPG affecting p-caveolin-1 to further mediate SOCE/[Ca2+]i against MI injury in neonatal rat cardiomyocytes and a rat model. Methods PP2, an inhibitor of p-caveolin-1, was used. Cell viability, [Ca2+]i concentration were analyzed in cardiomyocytes. In rats, myocardial infarct size, pathological damages, apoptosis and cardiac fibrosis were evaluated, p-caveolin-1 and STIM1 were detected by immunofluorescence, and the levels of caveolin-1, STIM1, ORAI1 and TRPC1 were determined by RT-PCR and Western blot. And, release of LDH, cTnI and BNP was measured. Results EPG, ginsenosides accounting for 57.96%, suppressed release of LDH, cTnI and BNP, and protected cardiomyocytes by inhibiting Ca2+ influx. And, EPG significantly relieved myocardial infarct size, cardiac apoptosis, fibrosis, and ultrastructure abnormality. Moreover, EPG negatively regulated SOCE via increasing p-caveolin-1 protein, decreasing ORAI1 mRNA and protein levels of ORAI1, TRPC1 and STIM1. More importantly, inhibition of the p-caveolin-1 significantly suppressed all of the above cardioprotection of EPG. Conclusions Caveolin-1 phosphorylation is involved in the protective effects of EPG against MI injury via increasing p-caveolin-1 to negatively regulate SOCE/[Ca2+]i.
Collapse
Affiliation(s)
- Hai-Xia Li
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, Henan Province, China
| | - Yan Ma
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Yu-Xiao Yan
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Xin-Ke Zhai
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Meng-Yu Xin
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Tian Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Dong-Cao Xu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Yu-Tong Song
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| | - Chun-Dong Song
- The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, 9 Renmin Road, Zhengzhou, Henan Province, China
| | - Cheng-Xue Pan
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan Province, China
| |
Collapse
|
6
|
Zhan C, Liu K, Zhang Y, Zhang Y, He M, Wu R, Bi C, Shen B. Myocardial infarction unveiled: Key miRNA players screened by a novel lncRNA-miRNA-mRNA network model. Comput Biol Med 2023; 160:106987. [PMID: 37141653 DOI: 10.1016/j.compbiomed.2023.106987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Myocardial infarction (MI) is a major contributor to global mortality, and microRNAs (miRNAs) are important in its pathogenesis. Identifying blood miRNAs with clinical application potential for the early detection and treatment of MI is crucial. METHODS We obtained MI-related miRNA and miRNA microarray datasets from MI Knowledge Base (MIKB) and Gene Expression Omnibus (GEO), respectively. A new feature called target regulatory score (TRS) was proposed to characterize the RNA interaction network. MI-related miRNAs were characterized using TRS, transcription factor (TF) gene proportion (TFP), and ageing-related gene (AG) proportion (AGP) via the lncRNA-miRNA-mRNA network. A bioinformatics model was then developed to predict MI-related miRNAs, which were verified by literature and pathway enrichment analysis. RESULTS The TRS-characterized model outperformed previous methods in identifying MI-related miRNAs. MI-related miRNAs had high TRS, TFP, and AGP values, and combining the three features improved prediction accuracy to 0.743. With this method, 31 candidate MI-related miRNAs were screened from the specific-MI lncRNA-miRNA-mRNA network, associated with key MI pathways like circulatory system processes, inflammatory response, and oxygen level adaptation. Most candidate miRNAs were directly associated with MI according to literature evidence, except hsa-miR-520c-3p and hsa-miR-190b-5p. Furthermore, CAV1, PPARA and VEGFA were identified as MI key genes, and were targeted by most of the candidate miRNAs. CONCLUSIONS This study proposed a novel bioinformatics model based on multivariate biomolecular network analysis to identify putative key miRNAs of MI, which deserve further experimental and clinical validation for translational applications.
Collapse
Affiliation(s)
- Chaoying Zhan
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuxin Zhang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China
| | - Yingbo Zhang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China; Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, Hainan, China
| | - Mengqiao He
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China
| | - Rongrong Wu
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China
| | - Cheng Bi
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China
| | - Bairong Shen
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
7
|
Chen Z, Wang Z, Liu D, Zhao X, Ning S, Liu X, Wang G, Zhang F, Luo F, Yao J, Tian X. Critical role of caveolin-1 in intestinal ischemia reperfusion by inhibiting protein kinase C βII. Free Radic Biol Med 2023; 194:62-70. [PMID: 36410585 DOI: 10.1016/j.freeradbiomed.2022.11.030] [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: 07/18/2022] [Revised: 10/24/2022] [Accepted: 11/17/2022] [Indexed: 11/19/2022]
Abstract
Intestinal ischemia reperfusion (I/R) is a common clinical pathological process. We previously reported that pharmacological inhibition of protein kinase C (PKC) βII with a specific inhibitor attenuated gut I/R injury. However, the endogenous regulatory mechanism of PKCβII inactivation is still unclear. Here, we explored the critical role of caveolin-1 (Cav1) in protecting against intestinal I/R injury by regulating PKCβII inactivation. PKCβII translocated to caveolae and bound with Cav1 after intestinal I/R. Cav1 was highly expressed in the intestine of mice with I/R and IEC-6 cells stimulated with hypoxia/reoxygenation (H/R). Cav1-knockout (KO) mice suffered from worse intestinal injury after I/R than wild-type (WT) mice and showed extremely low survival due to exacerbated systemic inflammatory response syndrome (SIRS) and remote organ (lung and liver) injury. Cav1 deficiency resulted in excessive PKCβII activation and increased oxidative stress and apoptosis after intestinal I/R. Full-length Cav1 scaffolding domain peptide (CSP) suppressed excessive PKCβII activation and protected the gut against oxidative stress and apoptosis due to I/R injury. In summary, Cav1 could regulate PKCβII endogenous inactivation to alleviate intestinal I/R injury. This finding may represent a novel therapeutic strategy for the prevention and treatment of intestinal I/R injury.
Collapse
Affiliation(s)
- Zhao Chen
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, 116023, Dalian, China
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Deshun Liu
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, 116023, Dalian, China
| | - Xuzi Zhao
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, 116023, Dalian, China
| | - Shili Ning
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, 116023, Dalian, China
| | - Xingming Liu
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, 116023, Dalian, China
| | - Guangzhi Wang
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, 116023, Dalian, China
| | - Feng Zhang
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, 116023, Dalian, China
| | - Fuwen Luo
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, 116023, Dalian, China
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Xiaofeng Tian
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, 116023, Dalian, China.
| |
Collapse
|
8
|
Wu SJ, He RL, Zhao L, Yu XY, Jiang YN, Guan X, Chen QY, Ren FF, Xie ZY, Wu LP, Li L. Cardiac-Specific Overexpression of Caveolin-1 in Rats With Ischemic Cardiomyopathy Improves Arrhythmogenicity and Cardiac Remodelling. Can J Cardiol 2023; 39:73-86. [PMID: 36240973 DOI: 10.1016/j.cjca.2022.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/16/2022] [Accepted: 10/04/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Ischemic cardiomyopathy (ICM) is associated with electrical and structural remodelling, leading to arrhythmias. Caveolin-1 (Cav1) is a membrane protein involved in the pathogenesis of ischemic injury. Cav1 deficiency has been associated with arrhythmogenicity. The current study aimed to determine how Cav1 overexpression inhibits arrhythmias and cardiac remodelling in ICM. METHODS ICM was modelled using left anterior descending (LAD) artery ligation for 4 weeks. Cardiac-specific Cav1 overexpression in ICM on arrhythmias, excitation-contraction coupling, and cardiac remodelling were investigated using the intramyocardial injection of an adeno-associated virus serotype 9 (AAV-9) system, carrying a specific sequence expressing Cav1 (AAVCav1) under the cardiac troponin T (cTnT) promoter. RESULTS Cav1 overexpression decreased susceptibility to arrhythmias by upregulating gap junction connexin 43 (CX43) and reducing spontaneous irregular proarrhythmogenic Ca2+ waves in ventricular cardiomyocytes. It also alleviated ischemic injury-induced contractility weakness by improving Ca2+ cycling through normalizing Ca2+-handling protein levels and improving Ca2+ homeostasis. Masson stain and immunoblotting revealed that the deposition of excessive fibrosis was attenuated by Cav1 overexpression, inhibiting the transforming growth factor-β (TGF-β)/Smad2 signalling pathway. Coimmunoprecipitation assays demonstrated that the interaction between Cav1 and cSrc modulated CX43 expression and Ca2+-handling protein levels. CONCLUSIONS Cardiac-specific overexpression of Cav1 attenuated ventricular arrhythmia, improved Ca2+ cycling, and attenuated cardiac remodelling. These effects were attributed to modulation of CX43, normalized Ca2+-handling protein levels, improved Ca2+ homeostasis, and attenuated cardiac fibrosis.
Collapse
Affiliation(s)
- Shu-Jie Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Rui-Lin He
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Lin Zhao
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Xiao-Yu Yu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Yi-Na Jiang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Xuan Guan
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Qiao-Ying Chen
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Fang-Fang Ren
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Zuo-Yi Xie
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Lian-Pin Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Lei Li
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China.
| |
Collapse
|
9
|
Xia W, Li X, Wu Q, Xu A, Zhang L, Xia Z. The importance of caveolin as a target in the prevention and treatment of diabetic cardiomyopathy. Front Immunol 2022; 13:951381. [PMID: 36405687 PMCID: PMC9666770 DOI: 10.3389/fimmu.2022.951381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/21/2022] [Indexed: 08/30/2023] Open
Abstract
The diabetic population has been increasing in the past decades and diabetic cardiomyopathy (DCM), a pathology that is defined by the presence of cardiac remodeling and dysfunction without conventional cardiac risk factors such as hypertension and coronary heart diseases, would eventually lead to fatal heart failure in the absence of effective treatment. Impaired insulin signaling, commonly known as insulin resistance, plays an important role in the development of DCM. A family of integral membrane proteins named caveolins (mainly caveolin-1 and caveolin-3 in the myocardium) and a protein hormone adiponectin (APN) have all been shown to be important for maintaining normal insulin signaling. Abnormalities in caveolins and APN have respectively been demonstrated to cause DCM. This review aims to summarize recent research findings of the roles and mechanisms of caveolins and APN in the development of DCM, and also explore the possible interplay between caveolins and APN.
Collapse
Affiliation(s)
- Weiyi Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xia Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Liangqing Zhang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
10
|
CAV-1 Overexpression Exacerbates the Manifestation in EPAC-1-Induced Chronic Postsurgical Pain in Rats. Pain Res Manag 2022; 2022:8566840. [PMID: 35958678 PMCID: PMC9357801 DOI: 10.1155/2022/8566840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 01/06/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022]
Abstract
Purpose. Caveolae (CAV) are an invaginated microcapsule with the shape of Ω on the surface of the cell membrane. Caveolin-1 (CAV-1) is involved in neuropathic pain, and adenosine monophosphate (AMP)-exchange protein directly activated by cAMP1 (EPAC-1) is a potential therapeutic target for chronic pain. However, whether EPAC-1 promotes chronic postsurgical pain (CPSP) through CAV-1 has not been reported. Here, we aim to investigate the underlying mechanism of CAV in CPSP. Methods. All the rats were divided into 9 groups, including the Naive group, Sham group, skin/muscle incision and retraction (SMIR) group, SMIR + CAV-1 siRNA group, SMIR + control siRNA group, SMIR (7 days)+Saline group, SMIR (7 days)+CE3F4 group, 8-PCPT group, and Saline group. The CPSP rat model was established after SMIR. A mechanical withdrawal threshold (MWT) was recorded to evaluate the animal’s behavior. Western blotting and immunofluorescent were performed to detect the protein expression levels of EPAC-1 and P-CAV-1. Results. EPAC-1 and CAV-1 were both overexpressed after operation, particularly in astrocytes, microglia, and neurons of spinal marrow (all
). Interestingly, CAV-1 siRNA can partly reverse the SMIR-induced hypersensitivity, but there was no effect on EPAC-1. Besides, EPAC-1 blockage partly reversed the SMIR-induced hypersensitivity and CAV-1 overexpression, and EPAC-1 activation promoted CAV-1 overexpression and hypersensitivity in normal rats (all
). Conclusion. CAV-1 mediates the functional coupling of microglia, astrocytes, and neurons, and thus EPAC-1/CAV-1 plays an important role in CPSP exacerbation.
Collapse
|
11
|
Zhao X, Yang X, An Z, Liu L, Yong J, Xing H, Huang R, Tian J, Song X. Pathophysiology and molecular mechanism of caveolin involved in myocardial protection strategies in ischemic conditioning. Biomed Pharmacother 2022; 153:113282. [PMID: 35750009 DOI: 10.1016/j.biopha.2022.113282] [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: 04/27/2022] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 11/02/2022] Open
Abstract
Multiple pathophysiological pathways are activated during the process of myocardial injury. Various cardioprotective strategies protect the myocardium from ischemia, infarction, and ischemia/reperfusion (I/R) injury through different targets, yet the clinical translation remains limited. Caveolae and its structure protein, caveolins, have been suggested as a bridge to transmit damage-preventing signals and mediate the protection of ultrastructure in cardiomyocytes under pathological conditions. In this review, we first briefly introduce caveolae and caveolins. Then we review the cardioprotective strategies mediated by caveolins through various pathophysiological pathways. Finally, some possible research directions are proposed to provide future experiments and clinical translation perspectives targeting caveolin based on the investigative evidence.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Xueyao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Jingwen Yong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Haoran Xing
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Rongchong Huang
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, 95th Yong An Road, Xuan Wu District, Beijing 100050, PR China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China.
| |
Collapse
|
12
|
The caveolin levels in cardiovascular disease. COR ET VASA 2021. [DOI: 10.33678/cor.2021.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
13
|
Mirzababaei A, Shiraseb F, Abaj F, Khosroshahi RA, Tavakoli A, Koohdani F, Clark CCT, Mirzaei K. The effect of dietary total antioxidant capacity (DTAC) and Caveolin-1 gene variant interaction on cardiovascular risk factors among overweight and obese women: A cross-sectional investigation. Clin Nutr 2021; 40:4893-4903. [PMID: 34358834 DOI: 10.1016/j.clnu.2021.07.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/26/2021] [Accepted: 07/06/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Previous studies have shown that the Caveolin-1 (CAV-1) gene variant may be associated with Cardiovascular disease (CVD) risk. Moreover, dietary total antioxidant capacity (DTAC) has been shown to potentially elicit favorable effects on CVD risk. Therefore, this study sought to investigate the effect of DTAC and CAV-1 interaction on CVD risk factors. METHODS This cross-sectional study consisted of 352 women, with overweight and/or obesity, aged 18-48years from Iran. A food frequency questionnaire (FFQ), with 147 items, was used to assess dietary intake. The CAV-1 rs 3807992 and anthropometric data were measured by the PCR-RFLP method and bioelectrical impedance analysis (BIA), respectively. Serum profiles were measured by standard protocols. Participants were also divided into two groups based on DTAC score and rs3807992 genotype. RESULTS The mean age of the participants was 37.34 ± 9.11 and 36.01 ± 9.12 years for homozygous (GG) and minor allele carriers (AG + AA) respectively.The mean ± SD of insulin, total cholesterol (TC),high-density lipoprotein (HDL), low-density lipoprotein (LDL) and TG of participants were 1.21 ± 0.23, 185.3 ± 35.77, 46.58 ± 10.86, 95.3 ± 24.12 and 118.1 ± 58.88, respectively. There was a significant difference between genotypes for physical activity (P = 0.05), HDL (P < 0.001), insulin (P = 0.04), CRI-I (TC/HDL-C) (P = 0.01), and CRI-II (LDL-C/HDL-C) (P = 0.04). Our findings also showed, after controlling for confounding factors, significant interactions between DTAC score and the A allele carrier group on TC (Pinteraction = 0.001), LDL (Pinteraction = 0.001), insulin (Pinteraction = 0.08), HOMA-IR (Pinteraction = 0.03), AC ((TC - HDL - C)/HDL - C) (Pinteraction = 0.001), and CHOLINDEX (LDL-C-HDL-C) (Pinteraction = 0.02). CONCLUSION The results of the present study indicate that high DTAC intake may modify the odds of risk factors for CVD in AA and AG genotypes of rs 3807992. These results highlight that diet, gene variants, and their interaction, should be considered in CVD risk assessment.
Collapse
Affiliation(s)
- Atieh Mirzababaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Farideh Shiraseb
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Faezeh Abaj
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Reza Amiri Khosroshahi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Atefeh Tavakoli
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Fariba Koohdani
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Cain C T Clark
- Centre for Intelligent Healthcare, Coventry University, Coventry, CV1 5FB, UK
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
14
|
Tang Y, Fang W, Xiao Z, Song M, Zhuang D, Han B, Wu J, Sun X. Nicotinamide ameliorates energy deficiency and improves retinal function in Cav-1 -/- mice. J Neurochem 2020; 157:550-560. [PMID: 33305362 DOI: 10.1111/jnc.15266] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 01/09/2023]
Abstract
Caveolin-1(Cav-1) is involved in lipid metabolism and energy homeostasis, which is important for the energetically demanding retina. Although retinal function deficits were noted in Cav-1 knockout (Cav-1-/- ) mice, the underlying causes remain largely unknown. Here, we investigate if the disruption in energy homeostasis presents a potential mechanism for retinal function deficits in Cav-1-/- retina and if it can be ameliorated by nicotinamide (NAM). In this study, NAM was administrated orally for 2 weeks in Cav-1-/- mice before experiments. Oxidative lipidomics was conducted to detect the oxylipin changes, the retinal energy flux was measured by seahorse assay, and the retinal function was assessed by electroretinogram (ERG). Cav-1 deficiency induced the dysregulation of oxidative lipidomics and reduction in energy consumption/production in the retina by decreasing Na+ /K+ -ATPase, oxidative phosphorylation CII, cytochrome c, and oxygen consumption rate (OCR). A decrease in Sirt1 was also detected. Therapeutic administration of NAM significantly increased Sirt1 expression and improved energy deficiency by increasing Na+ /K+ -ATPase, cytochrome c, and OCR. The dysregulation of oxidative lipidomics was partially recovered, and the retinal function was improved as assessed by ERG compared to Cav-1-/- mice. Our study demonstrated the dysregulation of oxidative lipidomics in Cav-1-/- retina and established a link between energy deficiency and retinal function deficits in Cav-1-/- mice. Administration of NAM ameliorated energy deficiency, increased the expression of Sirt1, and improved retinal function, which presents a potential therapeutic strategy for Cav-1 deficiency-induced retinal function deficits.
Collapse
Affiliation(s)
- Yizhen Tang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Wangyi Fang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Zebin Xiao
- Department of Radiology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Maomao Song
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Dongli Zhuang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Binze Han
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jihong Wu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xinghuai Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
15
|
Grivas D, González-Rajal Á, Guerrero Rodríguez C, Garcia R, de la Pompa JL. Loss of Caveolin-1 and caveolae leads to increased cardiac cell stiffness and functional decline of the adult zebrafish heart. Sci Rep 2020; 10:12816. [PMID: 32733088 PMCID: PMC7393500 DOI: 10.1038/s41598-020-68802-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/05/2020] [Indexed: 01/06/2023] Open
Abstract
Caveolin-1 is the main structural protein of caveolae, small membrane invaginations involved in signal transduction and mechanoprotection. Here, we generated cav1-KO zebrafish lacking Cav1 and caveolae, and investigated the impact of this loss on adult heart function and response to cryoinjury. We found that cardiac function was impaired in adult cav1-KO fish, which showed a significantly decreased ejection fraction and heart rate. Using atomic force microscopy, we detected an increase in the stiffness of epicardial cells and cells of the cortical zone lacking Cav1/caveolae. This loss of cardiac elasticity might explain the decreased cardiac contraction and function. Surprisingly, cav1-KO mutants were able to regenerate their heart after a cryoinjury but showed a transient decrease in cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Dimitrios Grivas
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.,Ciber de Enfermedades Cardiovasculares, 28029, Madrid, Spain
| | - Álvaro González-Rajal
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.,Cell Division Lab, ANZAC Research Institute, Gate 3, Hospital Road, Concord, NSW, 2139, Australia
| | - Carlos Guerrero Rodríguez
- Materials Science Factory, Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, 28049, Madrid, Spain
| | - Ricardo Garcia
- Materials Science Factory, Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, 28049, Madrid, Spain
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain. .,Ciber de Enfermedades Cardiovasculares, 28029, Madrid, Spain.
| |
Collapse
|
16
|
Abstract
Along with the progress of global aging, the prognosis of severe ischemic heart disease (IHD) remains poor, and thus the development of effective angiogenic therapy remains an important clinical unmet need. We have developed low-energy extracorporeal cardiac shock wave therapy as an innovative minimally invasive angiogenic therapy and confirmed its efficacy in a porcine chronic myocardial ischemia model in animal experiments as well as in patients with refractory angina. Since ultrasound is more advantageous for clinical application than shock waves, we then aimed to develop ultrasound therapy for IHD. We demonstrated that specific conditions of low-intensity pulsed ultrasound (LIPUS) therapy improve myocardial ischemia in animal models through the enhancement of angiogenesis mediated by endothelial mechanotransduction. To examine the effectiveness of our LIPUS therapy in patients with severe angina pectoris, we are now conducting a prospective multicenter clinical trial in Japan. Furthermore, to overcome the current serious situation of dementia pandemic but with no effective treatments worldwide, we have recently demonstrated that our LIPUS therapy also improves cognitive impairment in mouse models of Alzheimer's disease and vascular dementia. Here, we summarize the progress in our studies to develop angiogenic therapies with sound waves.
Collapse
Affiliation(s)
- Tomohiko Shindo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
17
|
Tian J, Popal MS, Huang R, Zhang M, Zhao X, Zhang M, Song X. Caveolin as a Novel Potential Therapeutic Target in Cardiac and Vascular Diseases: A Mini Review. Aging Dis 2020; 11:378-389. [PMID: 32257548 PMCID: PMC7069461 DOI: 10.14336/ad.2019.09603] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 06/03/2019] [Indexed: 12/27/2022] Open
Abstract
Caveolin, a structural protein of caveolae, play roles in the regulation of endothelial function, cellular lipid homeostasis, and cardiac function by affecting the activity and biogenesis of nitric oxide, and by modulating signal transduction pathways that mediate inflammatory responses and oxidative stress. In this review, we present the role of caveolin in cardiac and vascular diseases and the relevant signaling pathways involved. Furthermore, we discuss a novel therapeutic perspective comprising crosstalk between caveolin and autophagy.
Collapse
Affiliation(s)
- Jinfan Tian
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mohammad Sharif Popal
- 2 Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - RongChong Huang
- 3 Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100010, China
| | - Min Zhang
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xin Zhao
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mingduo Zhang
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiantao Song
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
18
|
Haddad D, Al Madhoun A, Nizam R, Al-Mulla F. Role of Caveolin-1 in Diabetes and Its Complications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9761539. [PMID: 32082483 PMCID: PMC7007939 DOI: 10.1155/2020/9761539] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/10/2019] [Accepted: 12/26/2019] [Indexed: 12/25/2022]
Abstract
It is estimated that in 2017 there were 451 million people with diabetes worldwide. These figures are expected to increase to 693 million by 2045; thus, innovative preventative programs and treatments are a necessity to fight this escalating pandemic disorder. Caveolin-1 (CAV1), an integral membrane protein, is the principal component of caveolae in membranes and is involved in multiple cellular functions such as endocytosis, cholesterol homeostasis, signal transduction, and mechanoprotection. Previous studies demonstrated that CAV1 is critical for insulin receptor-mediated signaling, insulin secretion, and potentially the development of insulin resistance. Here, we summarize the recent progress on the role of CAV1 in diabetes and diabetic complications.
Collapse
Affiliation(s)
- Dania Haddad
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Ashraf Al Madhoun
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rasheeba Nizam
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| |
Collapse
|
19
|
Buyang Huanwu Decoction Exerts Cardioprotective Effects through Targeting Angiogenesis via Caveolin-1/VEGF Signaling Pathway in Mice with Acute Myocardial Infarction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4275984. [PMID: 31178960 PMCID: PMC6501136 DOI: 10.1155/2019/4275984] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/02/2019] [Accepted: 03/11/2019] [Indexed: 11/18/2022]
Abstract
Background Acute myocardial infarction (AMI) remains a leading cause of morbidity and mortality worldwide. The idea of therapeutic angiogenesis in ischemic myocardium is a promising strategy for MI patients. Buyang Huanwu decoction (BHD), a famous Chinese herbal prescription, exerted antioxidant, antiapoptotic, and anti-inflammatory effects, which contribute to cardio-/cerebral protection. Here, we aim to investigate the effects of BHD on angiogenesis through the caveolin-1 (Cav-1)/vascular endothelial growth factor (VEGF) pathway in MI model of mice. Materials and Methods C57BL/6 mice were randomly divided into 3 groups by the table of random number: (1) sham-operated group (sham, n = 15), (2) AMI group (AMI+sham, n = 20), and (3) BHD-treated group (AMI+BHD, n = 20). 2,3,5-Triphenyltetrazolium chloride solution stain was used to determine myocardial infarct size. Myocardial histopathology was tested using Masson staining and hematoxylin-eosin staining. CD31 immunofluorescence staining was used to analyze the angiogenesis in the infarction border zone. Western blot analysis, immunofluorescence staining, and/or real-time quantitative reverse transcription polymerase chain reaction was applied to test the expression of Cav-1, VEGF, vascular endothelial growth factor receptor 2 (VEGFR2), and/or phosphorylated extracellular signal-regulated kinase (p-ERK). All statistical analyses were performed using the SPSS 20.0 software and GraphPad Prism 6.05. Values of P < 0.05 were considered as statistically significant. Results and Conclusion Compared with the AMI group, the BHD-treated group showed a significant improvement in the heart weight/body weight ratio, echocardiography images, cardiac function, infarct size, Mason staining of the collagen deposition area, and density of microvessel in the infarction border zone (P < 0.05). Compared with the AMI group, BHD promoted the expression of Cav-1, VEGF, VEGFR2, and p-ERK in the infarction border zone after AMI. BHD could exert cardioprotective effects on the mouse model with AMI through targeting angiogenesis via Cav-1/VEGF signaling pathway.
Collapse
|
20
|
Zhong W, Huang Q, Zeng L, Hu Z, Tang X. Caveolin-1 and MLRs: A potential target for neuronal growth and neuroplasticity after ischemic stroke. Int J Med Sci 2019; 16:1492-1503. [PMID: 31673241 PMCID: PMC6818210 DOI: 10.7150/ijms.35158] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 09/03/2019] [Indexed: 12/22/2022] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality worldwide. Thrombolytic therapy, the only established treatment to reduce the neurological deficits caused by ischemic stroke, is limited by time window and potential complications. Therefore, it is necessary to develop new therapeutic strategies to improve neuronal growth and neurological function following ischemic stroke. Membrane lipid rafts (MLRs) are crucial structures for neuron survival and growth signaling pathways. Caveolin-1 (Cav-1), the main scaffold protein present in MLRs, targets many neural growth proteins and promotes growth of neurons and dendrites. Targeting Cav-1 may be a promising therapeutic strategy to enhance neuroplasticity after cerebral ischemia. This review addresses the role of Cav-1 and MLRs in neuronal growth after ischemic stroke, with an emphasis on the mechanisms by which Cav-1/MLRs modulate neuroplasticity via related receptors, signaling pathways, and gene expression. We further discuss how Cav-1/MLRs may be exploited as a potential therapeutic target to restore neuroplasticity after ischemic stroke. Finally, several representative pharmacological agents known to enhance neuroplasticity are discussed in this review.
Collapse
Affiliation(s)
- Wei Zhong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qianyi Huang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Liuwang Zeng
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
21
|
Russell J, Du Toit EF, Peart JN, Patel HH, Headrick JP. Myocyte membrane and microdomain modifications in diabetes: determinants of ischemic tolerance and cardioprotection. Cardiovasc Diabetol 2017; 16:155. [PMID: 29202762 PMCID: PMC5716308 DOI: 10.1186/s12933-017-0638-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/22/2017] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease, predominantly ischemic heart disease (IHD), is the leading cause of death in diabetes mellitus (DM). In addition to eliciting cardiomyopathy, DM induces a ‘wicked triumvirate’: (i) increasing the risk and incidence of IHD and myocardial ischemia; (ii) decreasing myocardial tolerance to ischemia–reperfusion (I–R) injury; and (iii) inhibiting or eliminating responses to cardioprotective stimuli. Changes in ischemic tolerance and cardioprotective signaling may contribute to substantially higher mortality and morbidity following ischemic insult in DM patients. Among the diverse mechanisms implicated in diabetic impairment of ischemic tolerance and cardioprotection, changes in sarcolemmal makeup may play an overarching role and are considered in detail in the current review. Observations predominantly in animal models reveal DM-dependent changes in membrane lipid composition (cholesterol and triglyceride accumulation, fatty acid saturation vs. reduced desaturation, phospholipid remodeling) that contribute to modulation of caveolar domains, gap junctions and T-tubules. These modifications influence sarcolemmal biophysical properties, receptor and phospholipid signaling, ion channel and transporter functions, contributing to contractile and electrophysiological dysfunction, cardiomyopathy, ischemic intolerance and suppression of protective signaling. A better understanding of these sarcolemmal abnormalities in types I and II DM (T1DM, T2DM) can inform approaches to limiting cardiomyopathy, associated IHD and their consequences. Key knowledge gaps include details of sarcolemmal changes in models of T2DM, temporal patterns of lipid, microdomain and T-tubule changes during disease development, and the precise impacts of these diverse sarcolemmal modifications. Importantly, exercise, dietary, pharmacological and gene approaches have potential for improving sarcolemmal makeup, and thus myocyte function and stress-resistance in this ubiquitous metabolic disorder.
Collapse
Affiliation(s)
- Jake Russell
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Eugene F Du Toit
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Hemal H Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California San Diego, San Diego, USA
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia. .,School of Medical Science, Griffith University, Southport, QLD, 4217, Australia.
| |
Collapse
|
22
|
Nah J, Yoo SM, Jung S, Jeong EI, Park M, Kaang BK, Jung YK. Phosphorylated CAV1 activates autophagy through an interaction with BECN1 under oxidative stress. Cell Death Dis 2017; 8:e2822. [PMID: 28542134 PMCID: PMC5520747 DOI: 10.1038/cddis.2017.71] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 12/19/2022]
Abstract
CAV1/Caveolin1, an integral membrane protein, is involved in caveolae function and cellular signaling pathways. Here, we report that CAV1 is a positive regulator of autophagy under oxidative stress and cerebral ischemic injury. Treatment with hydrogen peroxide enhanced autophagy flux and caused the localization of BECN1 to the mitochondria, whereas these changes were impaired in the absence of CAV1. Among many autophagy signals, only LC3 foci formation in response to hydrogen peroxide was abolished by CAV1 deficiency. Under oxidative stress, CAV1 interacted with a complex of BECN1/VPS34 through its scaffolding domain, and this interaction facilitated autophagosome formation. Interestingly, the phosphorylation of CAV1 at tyrosine-14 was essential for the interaction with BECN1 and their localization to the mitochondria, and the activation of autophagy in response to hydrogen peroxide. In addition, the expression of a phosphatase PTPN1 reduced the phosphorylation of CAV1 and inhibited autophagy. Further, compared to that in wild-type mice, autophagy was impaired and cerebral infarct damage was aggravated in the brain of Cav1 knockout mice. These results suggest that the phosphorylated CAV1 functions to activate autophagy through binding to the BECN1/VPS34 complex under oxidative stress and to protect against ischemic damage.
Collapse
Affiliation(s)
- Jihoon Nah
- Department of Biological Sciences, Seoul National University, 1 Gwanak-ro, Seoul, Gwanak-gu 151-747, Korea
| | - Seung-Min Yoo
- Department of Biological Sciences, Seoul National University, 1 Gwanak-ro, Seoul, Gwanak-gu 151-747, Korea
| | - Sunmin Jung
- Department of Biological Sciences, Seoul National University, 1 Gwanak-ro, Seoul, Gwanak-gu 151-747, Korea
| | - Eun Il Jeong
- Department of Biological Sciences, Seoul National University, 1 Gwanak-ro, Seoul, Gwanak-gu 151-747, Korea
| | - Moonju Park
- Department of Biological Sciences, Seoul National University, 1 Gwanak-ro, Seoul, Gwanak-gu 151-747, Korea
| | - Bong-Kiun Kaang
- Department of Biological Sciences, Seoul National University, 1 Gwanak-ro, Seoul, Gwanak-gu 151-747, Korea
| | - Yong-Keun Jung
- Department of Biological Sciences, Seoul National University, 1 Gwanak-ro, Seoul, Gwanak-gu 151-747, Korea
| |
Collapse
|
23
|
Ichikawa Y, Zemljic-Harpf AE, Zhang Z, McKirnan MD, Manso AM, Ross RS, Hammond HK, Patel HH, Roth DM. Modulation of caveolins, integrins and plasma membrane repair proteins in anthracycline-induced heart failure in rabbits. PLoS One 2017; 12:e0177660. [PMID: 28498861 PMCID: PMC5428970 DOI: 10.1371/journal.pone.0177660] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/01/2017] [Indexed: 01/01/2023] Open
Abstract
Anthracyclines are chemotherapeutic drugs known to induce heart failure in a dose-dependent manner. Mechanisms involved in anthracycline cardiotoxicity are an area of relevant investigation. Caveolins bind, organize and regulate receptors and signaling molecules within cell membranes. Caveolin-3 (Cav-3), integrins and related membrane repair proteins can function as cardioprotective proteins. Expression of these proteins in anthracycline-induced heart failure has not been evaluated. We tested the hypothesis that daunorubicin alters cardioprotective protein expression in the heart. Rabbits were administered daunorubicin (3 mg/kg, IV) weekly, for three weeks or nine weeks. Nine weeks but not three weeks of daunorubicin resulted in progressive reduced left ventricular function. Cav-3 expression in the heart was unchanged at three weeks of daunorubicin and increased in nine week treated rabbits when compared to control hearts. Electron microscopy showed caveolae in the heart were increased and mitochondrial number and size were decreased after nine weeks of daunorubicin. Activated beta-1 (β1) integrin and the membrane repair protein MG53 were increased after nine weeks of daunorubicin vs. controls with no change at the three week time point. The results suggest a potential pathophysiological role for Cav3, integrins and membrane repair in daunorubicin-induced heart failure.
Collapse
Affiliation(s)
- Yasuhiro Ichikawa
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, United States of America
| | - Alice E. Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, United States of America
| | - Zheng Zhang
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - M. Dan McKirnan
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, United States of America
| | - Ana Maria Manso
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Robert S. Ross
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - H. Kirk Hammond
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Hemal H. Patel
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, United States of America
| | - David M. Roth
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
24
|
Pleasant-Jenkins D, Reese C, Chinnakkannu P, Kasiganesan H, Tourkina E, Hoffman S, Kuppuswamy D. Reversal of maladaptive fibrosis and compromised ventricular function in the pressure overloaded heart by a caveolin-1 surrogate peptide. J Transl Med 2017; 97:370-382. [PMID: 28112757 PMCID: PMC5909408 DOI: 10.1038/labinvest.2016.153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/21/2016] [Accepted: 12/01/2016] [Indexed: 12/19/2022] Open
Abstract
Chronic ventricular pressure overload (PO) results in congestive heart failure (CHF) in which myocardial fibrosis develops in concert with ventricular dysfunction. Caveolin-1 is important in fibrosis in various tissues due to its decreased expression in fibroblasts and monocytes. The profibrotic effects of low caveolin-1 can be blocked with the caveolin-1 scaffolding domain peptide (CSD, a caveolin-1 surrogate) using both mouse models and human cells. We have studied the beneficial effects of CSD on mice in which PO was induced by trans-aortic constriction (TAC). Beneficial effects observed in TAC mice receiving CSD injections daily included: improved ventricular function (increased ejection fraction, stroke volume, and cardiac output; reduced wall thickness); decreased collagen I, collagen chaperone HSP47, fibronectin, and CTGF levels; decreased activation of non-receptor tyrosine kinases Pyk2 and Src; and decreased activation of eNOS. To determine the source of cells that contribute to fibrosis in CHF, flow cytometric studies were performed that suggested that myofibroblasts in the heart are in large part bone marrow-derived. Two CD45+ cell populations were observed. One (Zone 1) contained CD45+/HSP47-/macrophage marker+ cells (macrophages). The second (Zone 2) contained CD45moderate/HSP47+/macrophage marker- cells often defined as fibrocytes. TAC increased the number of cells in Zones 1 and 2 and the level of HSP47 in Zone 2. These studies are a first step in elucidating the mechanism of action of CSD in heart fibrosis and promoting the development of CSD as a novel treatment to reduce fibrosis and improve ventricular function in CHF patients.
Collapse
Affiliation(s)
- Dorea Pleasant-Jenkins
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| | - Charles Reese
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | | | - Harinath Kasiganesan
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| | - Elena Tourkina
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Stanley Hoffman
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Dhandapani Kuppuswamy
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| |
Collapse
|
25
|
Kaakinen M, Reichelt ME, Ma Z, Ferguson C, Martel N, Porrello ER, Hudson JE, Thomas WG, Parton RG, Headrick JP. Cavin-1 deficiency modifies myocardial and coronary function, stretch responses and ischaemic tolerance: roles of NOS over-activity. Basic Res Cardiol 2017; 112:24. [PMID: 28343262 DOI: 10.1007/s00395-017-0613-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/09/2017] [Accepted: 03/09/2017] [Indexed: 02/07/2023]
Abstract
Caveolae and associated cavin and caveolins may govern myocardial function, together with responses to mechanical and ischaemic stresses. Abnormalities in these proteins are also implicated in different cardiovascular disorders. However, specific roles of the cavin-1 protein in cardiac and coronary responses to mechanical/metabolic perturbation remain unclear. We characterised cardiovascular impacts of cavin-1 deficiency, comparing myocardial and coronary phenotypes and responses to stretch and ischaemia-reperfusion in hearts from cavin-1 +/+ and cavin-1 -/- mice. Caveolae and caveolins 1 and 3 were depleted in cavin-1 -/- hearts. Cardiac ejection properties in situ were modestly reduced in cavin-1 -/- mice. While peak contractile performance in ex vivo myocardium from cavin-1 -/- and cavin-1 +/+ mice was comparable, intrinsic beating rate, diastolic stiffness and Frank-Starling behaviour (stretch-dependent diastolic and systolic forces) were exaggerated in cavin-1 -/- hearts. Increases in stretch-dependent forces were countered by NOS inhibition (100 µM L-NAME), which exposed negative inotropy in cavin-1 -/- hearts, and were mimicked by 100 µM nitroprusside. In contrast, chronotropic differences appeared largely NOS-independent. Cavin-1 deletion also induced NOS-dependent coronary dilatation, ≥3-fold prolongation of reactive hyperaemic responses, and exaggerated pressure-dependence of coronary flow. Stretch-dependent efflux of lactate dehydrogenase and cardiac troponin I was increased and induction of brain natriuretic peptide and c-Fos inhibited in cavin-1 -/- hearts, while ERK1/2 phospho-activation was preserved. Post-ischaemic dysfunction and damage was also exaggerated in cavin-1 -/- hearts. Diverse effects of cavin-1 deletion reveal important roles in both NOS-dependent and -independent control of cardiac and coronary functions, together with governing sarcolemmal fragility and myocardial responses to stretch and ischaemia.
Collapse
Affiliation(s)
- Mika Kaakinen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland.,Institute for Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Melissa E Reichelt
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Zhibin Ma
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Charles Ferguson
- Institute for Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Nick Martel
- Institute for Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Enzo R Porrello
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - James E Hudson
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Robert G Parton
- Institute for Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - John P Headrick
- School of Medical Science, Griffith University, Southport, QLD, 4217, Australia.
| |
Collapse
|
26
|
Cuadrado I, Castejon B, Martin AM, Saura M, Reventun-Torralba P, Zamorano JL, Zaragoza C. Nitric Oxide Induces Cardiac Protection by Preventing Extracellular Matrix Degradation through the Complex Caveolin-3/EMMPRIN in Cardiac Myocytes. PLoS One 2016; 11:e0162912. [PMID: 27649573 PMCID: PMC5029905 DOI: 10.1371/journal.pone.0162912] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022] Open
Abstract
Inhibition of Extracellular Matrix degradation by nitric oxide (NO) induces cardiac protection against coronary ischemia/reperfusion (IR). Glycosylation of Extracellular Matrix Metalloproteinase Inducer (EMMPRIN) stimulates enzymatic activation of matrix metalloproteinases (MMPs) in the heart, although the mechanisms leading to EMMPRIN glycosylation are poorly understood. We sought to determine if NO may induce cardiac protection by preventing glycosylation of EMMPRIN in a mouse model of IR. Here we found that Caveolin-3 binds to low glycosylated EMMPRIN (LG-EMMPRIN) in cardiac cells and in the hearts of healthy mice, whereas IR disrupted the complex in nitric oxide synthase 2 (NOS2) knockout (KO) mice. By contrast, the binding was partially restored when mice were fed with an NO donor (DEA-NO) in the drinking water, showing a significant reduction on infarct size (NOS2KO: 34.6±5 vs NOS2KO+DEA-NO: 20.7±9), in expression of matrix metalloproteinases, and cardiac performance was improved (left ventricular ejection fraction (LVEF). NOS2KO: 31±4 vs NOS2KO+DEA-NO: 46±6). The role of Caveolin-3/EMMPRIN in NO-mediated cardiac protection was further assayed in Caveolin-3 KO mice, showing no significant improvement on infarct size (Caveolin-3 KO: 34.8±3 vs Caveolin-3 KO+DEA-NO:33.7±5), or in the expression of MMPs, suggesting that stabilization of the complex Caveolin-3/LG-EMMPRIN may play a significant role in the cardioprotective effect of NO against IR.
Collapse
Affiliation(s)
- Irene Cuadrado
- Department of Systems Biology (Physiology), University of Alcalá, School of Medicine (IRYCIS), Ctra. Madrid Barcelona, Km 3,300, 28875, Alcalá de Henares, Madrid, Spain
| | - Borja Castejon
- Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034, Madrid, Spain
| | - Ana M. Martin
- Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034, Madrid, Spain
| | - Marta Saura
- Department of Systems Biology (Physiology), University of Alcalá, School of Medicine (IRYCIS), Ctra. Madrid Barcelona, Km 3,300, 28875, Alcalá de Henares, Madrid, Spain
| | - Paula Reventun-Torralba
- Department of Systems Biology (Physiology), University of Alcalá, School of Medicine (IRYCIS), Ctra. Madrid Barcelona, Km 3,300, 28875, Alcalá de Henares, Madrid, Spain
| | - Jose Luis Zamorano
- Cardiology Department, University Hospital Ramón y Cajal (IRYCIS), Ctra Colmenar Viejo, km. 9100, 28034, Madrid, Spain
| | - Carlos Zaragoza
- Cardiology Department, University Francisco de Vitoria/Hospital Ramón y Cajal Research Unit (IRYCIS), Ctra. Colmenar Viejo, km. 9100, 28034, Madrid, Spain
- * E-mail:
| |
Collapse
|
27
|
Caveolin-1/-3: therapeutic targets for myocardial ischemia/reperfusion injury. Basic Res Cardiol 2016; 111:45. [PMID: 27282376 DOI: 10.1007/s00395-016-0561-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/05/2016] [Accepted: 05/06/2016] [Indexed: 01/20/2023]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a major cause of morbidity and mortality worldwide. Caveolae, caveolin-1 (Cav-1), and caveolin-3 (Cav-3) are essential for the protective effects of conditioning against myocardial I/R injury. Caveolins are membrane-bound scaffolding proteins that compartmentalize and modulate signal transduction. In this review, we introduce caveolae and caveolins and briefly describe the interactions of caveolins in the cardiovascular diseases. We also review the roles of Cav-1/-3 in protection against myocardial ischemia and I/R injury, and in conditioning. Finally, we suggest several potential research avenues that may be of interest to clinicians and basic scientists. The information included, herein, is potentially useful for the design of future studies and should advance the investigation of caveolins as therapeutic targets.
Collapse
|
28
|
Shindo T, Ito K, Ogata T, Hatanaka K, Kurosawa R, Eguchi K, Kagaya Y, Hanawa K, Aizawa K, Shiroto T, Kasukabe S, Miyata S, Taki H, Hasegawa H, Kanai H, Shimokawa H. Low-Intensity Pulsed Ultrasound Enhances Angiogenesis and Ameliorates Left Ventricular Dysfunction in a Mouse Model of Acute Myocardial Infarction. Arterioscler Thromb Vasc Biol 2016; 36:1220-9. [DOI: 10.1161/atvbaha.115.306477] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 04/03/2016] [Indexed: 11/16/2022]
Affiliation(s)
- Tomohiko Shindo
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Kenta Ito
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Tsuyoshi Ogata
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Kazuaki Hatanaka
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Ryo Kurosawa
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Kumiko Eguchi
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Yuta Kagaya
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Kenichiro Hanawa
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Kentaro Aizawa
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Takashi Shiroto
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Sachie Kasukabe
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Satoshi Miyata
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Hirofumi Taki
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Hideyuki Hasegawa
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Hiroshi Kanai
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Graduate School of Medicine (T. Shindo, K.T., T.O., K. Hatanaka, R.K., K.E., Y.K., K. Hanawa, K.A., T. Shiroto, S.K., S.M., H.S.) and Department of Electronic Engineering, Graduate School of Engineering and Division of Biomedical Measurements and Diagnostics, Graduate School of Biomedical Engineering (H.T., H.H., H.K.), Tohoku University, Sendai, Japan
| |
Collapse
|
29
|
Wan F, Letavernier E, Le Saux CJ, Houssaini A, Abid S, Czibik G, Sawaki D, Marcos E, Dubois-Rande JL, Baud L, Adnot S, Derumeaux G, Gellen B. Calpastatin overexpression impairs postinfarct scar healing in mice by compromising reparative immune cell recruitment and activation. Am J Physiol Heart Circ Physiol 2015; 309:H1883-93. [PMID: 26453333 DOI: 10.1152/ajpheart.00594.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/03/2015] [Indexed: 12/15/2022]
Abstract
The activation of the calpain system is involved in the repair process following myocardial infarction (MI). However, the impact of the inhibition of calpain by calpastatin, its natural inhibitor, on scar healing and left ventricular (LV) remodeling is elusive. Male mice ubiquitously overexpressing calpastatin (TG) and wild-type (WT) controls were subjected to an anterior coronary artery ligation. Mortality at 6 wk was higher in TG mice (24% in WT vs. 44% in TG, P < 0.05) driven by a significantly higher incidence of cardiac rupture during the first week post-MI, despite comparable infarct size and LV dysfunction and dilatation. Calpain activation post-MI was blunted in TG myocardium. In TG mice, inflammatory cell infiltration and activation were reduced in the infarct zone (IZ), particularly affecting M2 macrophages and CD4(+) T cells, which are crucial for scar healing. To elucidate the role of calpastatin overexpression in macrophages, we stimulated peritoneal macrophages obtained from TG and WT mice in vitro with IL-4, yielding an abrogated M2 polarization in TG but not in WT cells. Lymphopenic Rag1(-/-) mice receiving TG splenocytes before MI demonstrated decreased T-cell recruitment and M2 macrophage activation in the IZ day 5 after MI compared with those receiving WT splenocytes. Calpastatin overexpression prevented the activation of the calpain system after MI. It also impaired scar healing, promoted LV rupture, and increased mortality. Defective scar formation was associated with blunted CD4(+) T-cell and M2-macrophage recruitment.
Collapse
Affiliation(s)
- Feng Wan
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Emmanuel Letavernier
- Department of Physiology, Assistance Publique-Hôpitaux de Paris (AP-HP), Tenon Hospital, Paris, France; Inflammation-Immunopathology-Biotherapy Department (DHU i2B), F-75020, Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, Unités Mixtes de Recherche Scientifique 1155, Paris, France; and
| | - Claude Jourdan Le Saux
- Department of Medicine/Cardiology Division, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Amal Houssaini
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Shariq Abid
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Gabor Czibik
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Daigo Sawaki
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Elisabeth Marcos
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France
| | - Jean-Luc Dubois-Rande
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; Département Hospitalo-Universitairé Ageing Thorax-Vessels Blood (DHU A-TVB), Department of Physiology, AP-HP, Henri Mondor Hospital, Créteil, France; DHU A-TVB, Department of Cardiology, AP-HP, Henri Mondor Hospital, Créteil, France
| | - Laurent Baud
- Department of Physiology, Assistance Publique-Hôpitaux de Paris (AP-HP), Tenon Hospital, Paris, France; Inflammation-Immunopathology-Biotherapy Department (DHU i2B), F-75020, Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, Unités Mixtes de Recherche Scientifique 1155, Paris, France; and
| | - Serge Adnot
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; Département Hospitalo-Universitairé Ageing Thorax-Vessels Blood (DHU A-TVB), Department of Physiology, AP-HP, Henri Mondor Hospital, Créteil, France
| | - Geneviève Derumeaux
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; Département Hospitalo-Universitairé Ageing Thorax-Vessels Blood (DHU A-TVB), Department of Physiology, AP-HP, Henri Mondor Hospital, Créteil, France
| | - Barnabas Gellen
- Institut National de la Santé et de la Recherche Médicale U955, Université Paris-Est Creteil, Créteil, France; DHU A-TVB, Department of Cardiology, AP-HP, Henri Mondor Hospital, Créteil, France; Department of Cardiology, Poitiers University Hospital, F-86000, Poitiers, France
| |
Collapse
|
30
|
Burma O, Onat E, Uysal A, Ilhan N, Erol D, Ozcan M, Sahna E. Effects of rosuvastatin on ADMA, rhokinase, NADPH oxidase, caveolin-1, hsp 90 and NFkB levels in a rat model of myocardial ischaemia-reperfusion. Cardiovasc J Afr 2015; 25:212-6. [PMID: 25629537 PMCID: PMC4241597 DOI: 10.5830/cvja-2014-038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 06/25/2014] [Indexed: 11/25/2022] Open
Abstract
Aim Endothelial dysfunction, oxidative stress and inflammation are among the most important mechanisms of ischaemia–reperfusion (I/R) injury. Besides their cholesterol-lowering effects, statins are known to provide protection against myocardial dysfunction and vascular endothelial injury via nitric oxide-dependent mechanisms. The aim of this study was to investigate the effects of rosuvastatin on certain intermediates involved in the generation of nitric oxide (asymmetrical dimethyl arginin, ADMA, caveolin-1 and hsp 90), oxidative stress (rhokinase, NADPH oxidase) and inflammation (NFkB), using an in vivo model of myocardial infarction in the rat. Methods Adult male Sprague Dawley rats were divided into three groups (control, I/R and I/R after 15 days of rosuvastatin administration). Reperfusion was applied for 120 min following left anterior descending coronary artery ischaemia for 30 min. Caveolin-1, hsp 90 and NFkB levels were evaluated with the quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and ADMA, rhokinase and NADPH oxidase levels were evaluated with ELISA. Results While NFkB and hsp 90 levels were higher in the I/R group, their levels were significantly lower in the rosuvastatin group. While ADMA and NADPH oxidase levels significantly increased with I/R, they were lower in the rosuvastatin-treated group, but not statistically significant. Rhokinase levels were significantly lower in the rosuvastatin group. Caveolin-1 levels were not different between the groups. Conclusion Our results suggest that ADMA, rhokinase, NADPH oxidase, hsp 90 and NFkB could facilitate I/R injury, and rosuvastatin significantly reduced levels of these parameters. These results indicate that rosuvastatin may have a protective role in I/R injury via mechanisms targeting inflammation, endothelial dysfunction and oxidative stress.
Collapse
Affiliation(s)
- Oktay Burma
- Department of Cardiovascular Surgery, Faculty of Medicine, University of Firat, Elazig, Turkey.
| | - Elif Onat
- Department of Pharmacology, Faculty of Medicine, University of Firat, Elazig, Turkey
| | - Ayhan Uysal
- Department of Cardiovascular Surgery, Faculty of Medicine, University of Firat, Elazig, Turkey
| | - Necip Ilhan
- Department of Biochemistry, Faculty of Medicine, University of Firat, Elazig, Turkey
| | - Deniz Erol
- Department of Medical Genetics, Faculty of Medicine, University of Firat, Elazig, Turkey
| | - Mete Ozcan
- Department of Biophysics, Faculty of Medicine, University of Firat, Elazig, Turkey
| | - Engin Sahna
- Department of Pharmacology, Faculty of Medicine, University of Firat, Elazig, Turkey
| |
Collapse
|
31
|
Nguyen NT, Lindsey ML, Jin YF. Systems analysis of gene ontology and biological pathways involved in post-myocardial infarction responses. BMC Genomics 2015; 16 Suppl 7:S18. [PMID: 26100218 PMCID: PMC4474415 DOI: 10.1186/1471-2164-16-s7-s18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Pathway analysis has been widely used to gain insight into essential mechanisms of the response to myocardial infarction (MI). Currently, there exist multiple pathway databases that organize molecular datasets and manually curate pathway maps for biological interpretation at varying forms of organization. However, inconsistencies among different databases in pathway descriptions, frequently due to conflicting results in the literature, can generate incorrect interpretations. Furthermore, although pathway analysis software provides detailed images of interactions among molecules, it does not exhibit how pathways interact with one another or with other biological processes under specific conditions. Methods We propose a novel method to standardize descriptions of enriched pathways for a set of genes/proteins using Gene Ontology terms. We used this method to examine the relationships among pathways and biological processes for a set of condition-specific genes/proteins, represented as a functional biological pathway-process network. We applied this algorithm to a set of 613 MI-specific proteins we previously identified. Results A total of 96 pathways from Biocarta, KEGG, and Reactome, and 448 Gene Ontology Biological Processes were enriched with these 613 proteins. The pathways were represented as Boolean functions of biological processes, delivering an interactive scheme to organize enriched information with an emphasis on involvement of biological processes in pathways. We extracted a network focusing on MI to demonstrate that tyrosine phosphorylation of Signal Transducer and Activator of Transcription (STAT) protein, positive regulation of collagen metabolic process, coagulation, and positive/negative regulation of blood coagulation have immediate impacts on the MI response. Conclusions Our method organized biological processes and pathways in an unbiased approach to provide an intuitive way to identify biological properties of pathways under specific conditions. Pathways from different databases have similar descriptions yet diverse biological processes, indicating variation in their ability to share similar functional characteristics. The coverages of pathways can be expanded with the incorporation of more biological processes, predicting involvement of protein members in pathways. Further, detailed analyses of the functional biological pathway-process network will allow researchers and scientists to explore critical routes in biological systems in the progression of disease.
Collapse
|
32
|
Sanon VP, Sawaki D, Mjaatvedt CH, Jourdan‐Le Saux C. Myocardial Tissue Caveolae. Compr Physiol 2015; 5:871-86. [DOI: 10.1002/cphy.c140050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
33
|
Xu L, Guo R, Xie Y, Ma M, Ye R, Liu X. Caveolae: molecular insights and therapeutic targets for stroke. Expert Opin Ther Targets 2015; 19:633-50. [PMID: 25639269 DOI: 10.1517/14728222.2015.1009446] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Caveolae are specialized plasma membrane micro-invaginations of most mammalian cell types. The organization and function of caveolae are carried out by their coat proteins, caveolins and adaptor proteins, cavins. Caveolae/caveolins physically interact with membrane-associated signaling molecules and function in cholesterol incorporation, signaling transduction and macromolecular transport/permeability. AREAS COVERED Recent investigations have implicated a check-and-balance role of caveolae in the pathophysiology of cerebral ischemia. Caveolin knockout mice displayed exacerbated ischemic injury, whereas caveolin peptide exerted remarkable protection against ischemia/reperfusion injury. This review attempts to provide a comprehensive synopsis of how caveolae/caveolins modulate blood-brain barrier permeability, pro-survival signaling, angiogenesis and neuroinflammation, and how this may contribute to a better understanding of the participation of caveolae in ischemic cascade. The role of caveolin in the preconditioning-induced tolerance against ischemia is also discussed. EXPERT OPINION Caveolae represent a novel target for cerebral ischemia. It remains open how to manipulate caveolin expression in a practical way to recapitulate the beneficial therapeutic outcomes. Caveolin peptides and associated antagomirs may be efficacious and deserve further investigations for their potential benefits for stroke.
Collapse
Affiliation(s)
- Lili Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University , Nanjing 210002 , China
| | | | | | | | | | | |
Collapse
|
34
|
Gao Y, Chu M, Hong J, Shang J, Xu D. Hypoxia induces cardiac fibroblast proliferation and phenotypic switch: a role for caveolae and caveolin-1/PTEN mediated pathway. J Thorac Dis 2014; 6:1458-68. [PMID: 25364523 DOI: 10.3978/j.issn.2072-1439.2014.08.31] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/05/2014] [Indexed: 01/18/2023]
Abstract
BACKGROUND Cardiac fibrosis following myocardial infarction (MI) results in heart failure. Caveolin-1, the main structural protein of caveolae, regulates signal transduction pathways controlling cell proliferation and apoptosis. Meanwhile, low phosphatase and tensin homolog (PTEN) activity enhances the PI3K/Akt signal pathway to induce cell proliferation. But whether caveolin-1 and PTEN activation regulates cardiac fibroblast proliferation and contributes to cardiac fibrosis from ischemic injury is incompletely understood. This study investigates whether hypoxia inducing cardiac fibroblast proliferation and phenotypic switch is caveolin-dependent. METHODS We used in vitro and in vivo models of ischemic injury, immunohistochemical staining, and cell proliferation assays to address this hypothesis. RESULTS We found that MI induced collagen deposition and cardiac dysfunction. After MI, mice displayed reduced caveolin-1 and PTEN expression and increased α-smooth muscle actin (α-SMA) expression in the infarct zone. Qualitative and quantitative analyses indicated that caveolin-1 expression was lowest at 7 days after MI, accompanied by increased collagen deposition and attenuated cardiac function. We cultured cardiac fibroblasts of mice were in hypoxia or normoxia conditions for 12, 24 and 48 hours. At all the time points, caveolin-1 and PTEN expression were gradually reduced, whereas, α-SMA was gradually increased. We also observed that cell viability was increased at 12 and 24 h after hypoxia then lightly decreased at 48 h. Additionally, disruption of caveolae with methyl-β-cyclodextrin (MβCD) enhanced p-Akt and α-SMA expression and fibroblast proliferation and phenotypic switch. CONCLUSIONS These findings suggest a key role for caveolae, perhaps through the caveolin-1/PTEN signaling pathway, in cardiac fibroblast proliferation and phenotypic switch under hypoxia.
Collapse
Affiliation(s)
- Yao Gao
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Ming Chu
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Jian Hong
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Jingping Shang
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Di Xu
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
35
|
Shivshankar P, Halade GV, Calhoun C, Escobar GP, Mehr AJ, Jimenez F, Martinez C, Bhatnagar H, Mjaatvedt CH, Lindsey ML, Le Saux CJ. Caveolin-1 deletion exacerbates cardiac interstitial fibrosis by promoting M2 macrophage activation in mice after myocardial infarction. J Mol Cell Cardiol 2014; 76:84-93. [PMID: 25128086 PMCID: PMC4533121 DOI: 10.1016/j.yjmcc.2014.07.020] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 02/08/2023]
Abstract
Adverse remodeling following myocardial infarction (MI) leading to heart failure is driven by an imbalanced resolution of inflammation. The macrophage cell is an important control of post-MI inflammation, as macrophage subtypes secrete mediators to either promote inflammation and extend injury (M1 phenotype) or suppress inflammation and promote scar formation (M2 phenotype). We have previously shown that the absence of caveolin-1 (Cav1), a membrane scaffolding protein, is associated with adverse cardiac remodeling in mice, but the mechanisms responsible remain to be elucidated. We explore here the role of Cav1 in the activation of macrophages using wild type C57BL6/J (WT) and Cav1(tm1Mls/J) (Cav1(-/-)) mice. By echocardiography, cardiac function was comparable between WT and Cav1(-/-) mice at 3days post-MI. In the absence of Cav1, there were a surprisingly higher percentage of M2 macrophages (arginase-1 positive) detected in the infarcted zone. Conversely, restoring Cav1 function after MI in WT mice by adding back the Cav1 scaffolding domain reduced the M2 activation profile. Further, adoptive transfer of Cav1 null macrophages into WT mice on d3 post-MI exacerbated adverse cardiac remodeling at d14 post-MI. In vitro studies revealed that Cav1 null macrophages had a more pronounced M2 profile activation in response to IL-4 stimulation. In conclusion, Cav1 deletion promotes an array of maladaptive repair processes after MI, including increased TGF-β signaling, increased M2 macrophage infiltration and dysregulation of the M1/M2 balance. Our data also suggest that cardiac remodeling can be improved by therapeutic intervention regulating Cav1 function during the inflammatory response phase.
Collapse
Affiliation(s)
- Pooja Shivshankar
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ganesh V Halade
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Cheresa Calhoun
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Gladys P Escobar
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ali J Mehr
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Fabio Jimenez
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Cindy Martinez
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Harshita Bhatnagar
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Corey H Mjaatvedt
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Merry L Lindsey
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Claude Jourdan Le Saux
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
36
|
Vandermeulen E, Ruttens D, Verleden SE, Vos R, Van Raemdonck DE, Kastelijn EA, Wauters E, Lambrechts D, Nawrot TS, Cox B, Verleden GM, Vanaudenaerde BM. Genetic Variation in Caveolin-1 Affects Survival After Lung Transplantation. Transplantation 2014; 98:354-9. [DOI: 10.1097/tp.0000000000000058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
37
|
Niesman IR, Schilling JM, Shapiro LA, Kellerhals SE, Bonds JA, Kleschevnikov AM, Cui W, Voong A, Krajewski S, Ali SS, Roth DM, Patel HH, Patel PM, Head BP. Traumatic brain injury enhances neuroinflammation and lesion volume in caveolin deficient mice. J Neuroinflammation 2014; 11:39. [PMID: 24593993 PMCID: PMC3975903 DOI: 10.1186/1742-2094-11-39] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 02/10/2014] [Indexed: 11/30/2022] Open
Abstract
Background Traumatic brain injury (TBI) enhances pro-inflammatory responses, neuronal loss and long-term behavioral deficits. Caveolins (Cavs) are regulators of neuronal and glial survival signaling. Previously we showed that astrocyte and microglial activation is increased in Cav-1 knock-out (KO) mice and that Cav-1 and Cav-3 modulate microglial morphology. We hypothesized that Cavs may regulate cytokine production after TBI. Methods Controlled cortical impact (CCI) model of TBI (3 m/second; 1.0 mm depth; parietal cortex) was performed on wild-type (WT; C57Bl/6), Cav-1 KO, and Cav-3 KO mice. Histology and immunofluorescence microscopy (lesion volume, glia activation), behavioral tests (open field, balance beam, wire grip, T-maze), electrophysiology, electron paramagnetic resonance, membrane fractionation, and multiplex assays were performed. Data were analyzed by unpaired t tests or analysis of variance (ANOVA) with post-hoc Bonferroni’s multiple comparison. Results CCI increased cortical and hippocampal injury and decreased expression of MLR-localized synaptic proteins (24 hours), enhanced NADPH oxidase (Nox) activity (24 hours and 1 week), enhanced polysynaptic responses (1 week), and caused hippocampal-dependent learning deficits (3 months). CCI increased brain lesion volume in both Cav-3 and Cav-1 KO mice after 24 hours (P < 0.0001, n = 4; one-way ANOVA). Multiplex array revealed a significant increase in expression of IL-1β, IL-9, IL-10, KC (keratinocyte chemoattractant), and monocyte chemoattractant protein 1 (MCP-1) in ipsilateral hemisphere and IL-9, IL-10, IL-17, and macrophage inflammatory protein 1 alpha (MIP-1α) in contralateral hemisphere of WT mice after 4 hours. CCI increased IL-2, IL-6, KC and MCP-1 in ipsilateral and IL-6, IL-9, IL-17 and KC in contralateral hemispheres in Cav-1 KO and increased all 10 cytokines/chemokines in both hemispheres except for IL-17 (ipsilateral) and MIP-1α (contralateral) in Cav-3 KO (versus WT CCI). Cav-3 KO CCI showed increased IL-1β, IL-9, KC, MCP-1, MIP-1α, and granulocyte-macrophage colony-stimulating factor in ipsilateral and IL-1β, IL-2, IL-9, IL-10, and IL-17 in contralateral hemispheres (P = 0.0005, n = 6; two-way ANOVA) compared to Cav-1 KO CCI. Conclusion CCI caused astrocyte and microglial activation and hippocampal neuronal injury. Cav-1 and Cav-3 KO exhibited enhanced lesion volume and cytokine/chemokine production after CCI. These findings suggest that Cav isoforms may regulate neuroinflammatory responses and neuroprotection following TBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Brian P Head
- Veterans Affairs San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92161, USA.
| |
Collapse
|
38
|
Yi SL, Liu XJ, Zhong JQ, Zhang Y. Role of caveolin-1 in atrial fibrillation as an anti-fibrotic signaling molecule in human atrial fibroblasts. PLoS One 2014; 9:e85144. [PMID: 24454806 PMCID: PMC3891766 DOI: 10.1371/journal.pone.0085144] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 11/23/2013] [Indexed: 12/31/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia in the general population; yet, the precise mechanisms resulting in AF are not fully understood. Caveolin-1 (Cav-1), the principal structural component of caveolae organelles in cardiac fibroblasts, is involved in several cardiovascular conditions; however, the study on its function in atrium, in particular, in AF, is still lacking. This report examines the hypothesis that Cav-1 confers an anti-AF effect by mediating atrial structural remodeling through its anti-fibrotic action. We evaluated the expression of Cav-1, transforming growth factor-β1 (TGF-β1), and fibrosis in atrial specimens of 13 patients with AF and 10 subjects with sinus rhythm, and found that the expression of Cav-1 was significantly downregulated, whereas TGF-β1 level, collagens I/III contents and atrial fibrosis were markedly increased, in AF. Western blot analysis demonstrated that treatment of human atrial fibroblasts (HAFs) with TGF-β1 resulted in a concentration- and time-dependent repression of Cav-1. Downregulation of Cav-1 with siRNA increased the TGF-β1-induced activation of Smad signal pathway and collagens production in HAFs. Furthermore, incubation of HAFs with the peptides derived from Cav-1 to achieve Cav-1 gain-of-function abolished the TGF-β1-induced production of collagens I/III and decreases of MMP-2/-9 expression. Therefore it was concluded that Cav-1 is an important anti-AF signaling mediator by conferring its anti-fibrotic effects in atrium.
Collapse
Affiliation(s)
- Shao-lei Yi
- Key Laboratory of cardiovascular remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- School of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiao-jun Liu
- Key Laboratory of cardiovascular remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Jing-quan Zhong
- Key Laboratory of cardiovascular remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- * E-mail:
| | - Yun Zhang
- Key Laboratory of cardiovascular remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
39
|
Mutation in integrin-linked kinase (ILK(R211A)) and heat-shock protein 70 comprise a broadly cardioprotective complex. PLoS One 2013; 8:e77331. [PMID: 24260102 PMCID: PMC3832499 DOI: 10.1371/journal.pone.0077331] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/31/2013] [Indexed: 12/14/2022] Open
Abstract
Rationale Integrin-linked kinase (ILK) has been proposed as a novel molecular target that has translational potential in diverse cardiac diseases, since its upregulation promotes a broadly cardioprotective phenotype. However, ILK has been implicated as both a cardioprotective and oncogenic target, which imposes therapeutic constraints that are generally relevant to the translational potential of many kinases. Objective To study the cardioprotective properties of the activation-resistant, non-oncogenic, mutation of ILK (ILKR211A) against experimental MI invivo and Doxorubicin induced apoptosis invitro and it’s relationships to stress induced heat shock proteins. Methods/Results The transgenic mouse heart over-expressing a point mutation in the ILK pleckstrin homology (PH) domain (TgR211A) exhibits a highly cardioprotective phenotype based on LAD-ligation-induced MI reduction invivo, and on protection against doxorubicin (DOX)-induced cardiomyocyte apoptosis when overexpressed in human induced pluripotent stem cell (iPS)-derived cardiomyocytes invitro. Intriguingly, the degree of cardioprotection seen with the ILKR211A mutation exceeded that with the ILKS343D mutation. Microarray and immunoprecipitation analyses revealed upregulation of expression levels and specific binding of ILKWT, ILKS343D and ILKR211A to both constitutively active heat-shock protein 70 (Hsc70) and inducible Hsp70 in response to MI, and to acute ILK overexpression in iPSC-cardiomyocytes. ILK-mediated cardioprotection was shown to depend upon Hsp70 ATPase activity. Conclusions These findings indicate that wild type ILK and the non-oncogenic ILKR211A mutation comprise a cardioprotective module with Hsp/c70. These results advance a novel target discovery theme in which kinase mutations can be safely engineered to enhance cardioprotective effects.
Collapse
|
40
|
Fridolfsson HN, Patel HH. Caveolin and caveolae in age associated cardiovascular disease. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2013; 10:66-74. [PMID: 23610576 PMCID: PMC3627709 DOI: 10.3969/j.issn.1671-5411.2013.01.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 12/15/2012] [Accepted: 12/18/2012] [Indexed: 12/13/2022]
Abstract
It is estimated that the elderly (> 65 years of age) will increase from 13%−14% to 25% by 2035. If this trend continues, > 50% of the United States population and more than two billion people worldwide will be “aged” in the next 50 years. Aged individuals face formidable challenges to their health, as aging is associated with a myriad of diseases. Cardiovascular disease is the leading cause of morbidity and mortality in the United States with > 50% of mortality attributed to coronary artery disease and > 80% of these deaths occurring in those age 65 and older. Therefore, age is an important predictor of cardiovascular disease. The efficiency of youth is built upon cellular signaling scaffolds that provide tight and coordinated signaling. Lipid rafts are one such scaffold of which caveolae are a subset. In this review, we consider the importance of caveolae in common cardiovascular diseases of the aged and as potential therapeutic targets. We specifically address the role of caveolin in heart failure, myocardial ischemia, and pulmonary hypertension.
Collapse
Affiliation(s)
- Heidi N Fridolfsson
- Departments of Anesthesiology, University of California, San Diego, La Jolla, California 92093, USA
| | | |
Collapse
|
41
|
Effect of Ischemia Reperfusion Injury and Epoxyeicosatrienoic Acids on Caveolin Expression in Mouse Myocardium. J Cardiovasc Pharmacol 2013; 61:258-63. [DOI: 10.1097/fjc.0b013e31827afcee] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
42
|
Sellers SL, Trane AE, Bernatchez PN. Caveolin as a potential drug target for cardiovascular protection. Front Physiol 2012; 3:280. [PMID: 22934034 PMCID: PMC3429054 DOI: 10.3389/fphys.2012.00280] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 06/28/2012] [Indexed: 01/12/2023] Open
Abstract
Caveolae and caveolin are key players in a number of disease processes. Current research indicates that caveolins play a significant role in cardiovascular disease and dysfunction. The far-reaching roles of caveolins in disease and dysfunction make them particularly notable therapeutic targets. In particular, caveolin-1 (Cav-1) and caveolin-3 (Cav-3) have been identified as potential regulators of vascular dysfunction and heart disease and might even confer cardiac protection in certain settings. Such a central role in vascular health therefore makes manipulation of Cav-1/3 function or expression levels clear therapeutic targets in a variety of cardiovascular related disease states. Here, we highlight the role of Cav-1 and Cav-3 in cardiovascular health and explore the potential of Cav-1 and Cav-3 derived experimental therapeutics.
Collapse
Affiliation(s)
- Stephanie L Sellers
- Department of Anesthesiology, Pharmacology and Therapeutics and The James Hogg Research Centre, University of British Columbia Vancouver, BC, Canada
| | | | | |
Collapse
|