1
|
Zhao Y, Sakurai T, Kamiyoshi A, Tanaka M, Ichikawa-Shindo Y, Kawate H, Matsuda Y, Zhang Y, Guo Q, Li P, Hoshiyama K, Hayashi M, Li J, Shindo T. Adrenomedullin 2/Intermedin Exerts Cardioprotective Effects by Regulating Cardiomyocyte Mitochondrial Function. Hypertension 2025; 82:e6-e21. [PMID: 39817339 DOI: 10.1161/hypertensionaha.124.23666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Adrenomedullin 2 (AM2) plays critical roles in regulating blood pressure and fluid balance. However, the specific involvement of AM2 in cardiac hypertrophy has not been comprehensively elucidated, warranting further investigation into its molecular mechanisms and therapeutic implications. METHODS Cardiac hypertrophy was induced in adult mice lacking AM2 (AM2-/-) using transverse aortic constriction surgery. Comprehensive cardiac morphology, function, histology, and transcriptome/metabolome analyses were conducted. Signal transduction underlying AM2 stimulation in the cardiomyocytes was explored. RESULTS The absence of endogenous AM2 led to the development of severe heart failure after transverse aortic constriction surgery, which was characterized by alterations in the mitochondrial morphology and function associated with glycolysis and the tricarboxylic acid cycle in the heart and cardiomyocytes of transverse aortic constriction-operated AM2-/- mice. AM2 stimulation was associated with the receptor-modifying factor RAMP2 (receptor activity-modifying protein 2), which primarily transduces signals through the MAPK (mitogen-activated protein kinase) pathway and affects the expression of genes involved in glycolysis, β-oxidation, and oxidative phosphorylation. The administration of exogenous AM2 alleviated heart failure following transverse aortic constriction. CONCLUSIONS AM2 crucially regulates mitochondrial functions associated with the glycolysis and tricarboxylic acid cycles in the cardiomyocytes, thereby exerting a protective effect on the heart under pressure overload conditions.
Collapse
Affiliation(s)
- Yunlu Zhao
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takayuki Sakurai
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Nagano, Japan (T. Sakurai, A.K., M.T., T. Shindo)
| | - Akiko Kamiyoshi
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Nagano, Japan (T. Sakurai, A.K., M.T., T. Shindo)
| | - Megumu Tanaka
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Yuka Ichikawa-Shindo
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Hisaka Kawate
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Yorishige Matsuda
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Ophthalmology (Y.M., K.H.), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Yan Zhang
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Qianqian Guo
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Peixuan Li
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Ken Hoshiyama
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Ophthalmology (Y.M., K.H.), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Marina Hayashi
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Jiake Li
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takayuki Shindo
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Nagano, Japan (T. Sakurai, A.K., M.T., T. Shindo)
| |
Collapse
|
2
|
Zhao J, Han L, Zhang YR, Liu SM, Ji DR, Wang R, Yu YR, Jia MZ, Chai SB, Tang HF, Huang W, Qi YF. Intermedin Alleviates Diabetic Cardiomyopathy by Up-Regulating CPT-1β through Activation of the Phosphatidyl Inositol 3 Kinase/Protein Kinase B Signaling Pathway. Pharmaceuticals (Basel) 2024; 17:1204. [PMID: 39338366 PMCID: PMC11435185 DOI: 10.3390/ph17091204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/03/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Diabetic cardiomyopathy (DCM), one of the most serious long-term consequences of diabetes, is closely associated with myocardial fatty acid metabolism. Carnitine palmitoyltransferase-1β (CPT-1β) is the rate-limiting enzyme responsible for β-oxidation of long-chain fatty acids. Intermedin (IMD) is a pivotal bioactive small molecule peptide, participating in the protection of various cardiovascular diseases. However, the role and underlying mechanisms of IMD in DCM are still unclear. In this study, we investigated whether IMD alleviates DCM via regulating CPT-1β. A rat DCM model was established by having rats to drink fructose water for 12 weeks. A mouse DCM model was induced by feeding mice a high-fat diet for 16 weeks. We showed that IMD and its receptor complexes levels were significantly down-regulated in the cardiac tissues of DCM rats and mice. Reduced expression of IMD was also observed in neonatal rat cardiomyocytes treated with palmitic acid (PA, 300 μM) in vitro. Exogenous and endogenous IMD mitigated cardiac hypertrophy, fibrosis, dysfunction, and lipid accumulation in DCM rats and IMD-transgenic DCM mice, whereas knockout of IMD worsened these pathological processes in IMD-knockout DCM mice. In vitro, IMD alleviated PA-induced cardiomyocyte hypertrophy and cardiac fibroblast activation. We found that CPT-1β enzyme activity, mRNA and protein levels, and acetyl-CoA content were increased in T2DM patients, rats and mice. IMD up-regulated the CPT-1β levels and acetyl-CoA content in T2DM rats and mice. Knockdown of CPT-1β blocked the effects of IMD on increasing acetyl-CoA content and on inhibiting cardiomyocyte hypertrophy and cardiac fibroblast activation. IMD receptor antagonist IMD17-47 and the phosphatidyl inositol 3 kinase (PI3K)/protein kinase B (Akt) inhibitor LY294002 reversed the effects of IMD on up-regulating CPT-1β and acetyl-CoA expression and on inhibiting cardiomyocyte hypertrophy and cardiac fibroblast activation. We revealed that IMD alleviates DCM by up-regulating CPT-1β via calcitonin receptor-like receptor/receptor activity-modifying protein (CRLR/RAMP) receptor complexes and PI3K/Akt signaling. IMD may serve as a potent therapeutic target for the treatment of DCM.
Collapse
Affiliation(s)
- Jie Zhao
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Ling Han
- Department of Cardiology, Fuxing Hospital, Capital Medical University, Beijing 100038, China
| | - Ya-Rong Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Shi-Meng Liu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Deng-Ren Ji
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Rui Wang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Yan-Rong Yu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Mo-Zhi Jia
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - San-Bao Chai
- Department of Endocrinology and Metabolism, Peking University International Hospital, Beijing 102206, China
| | - Hui-Fang Tang
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Wei Huang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Yong-Fen Qi
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100083, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100083, China
| |
Collapse
|
3
|
Yang Z, Li H, Wu P, Li Q, Yu C, Wang D, Li W. Multi-biological functions of intermedin in diseases. Front Physiol 2023; 14:1233073. [PMID: 37745233 PMCID: PMC10511904 DOI: 10.3389/fphys.2023.1233073] [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: 06/02/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Intermedin (IMD) is a member of the calcitonin gene-related peptide (CGRP)/calcitonin (CT) superfamily, and it is expressed extensively throughout the body. The typical receptors for IMD are complexes composed of calcitonin receptor-like receptor (CLR) and receptor activity-modifying protein (RAMP), which leads to a biased activation towards Gαs. As a diagnostic and prognostic biomarker, IMD regulates the initiation and metastasis of multiple tumors. Additionally, IMD functions as a proangiogenic factor that can restrain excessive vascular budding and facilitate the expansion of blood vessel lumen, ultimately resulting in the fusion of blood vessels. IMD has protective roles in various diseases, including ischemia-reperfusion injury, metabolic disease, cardiovascular diseases and inflammatory diseases. This review systematically elucidates IMD's expression, structure, related receptors and signal pathway, as well as its comprehensive functions in the context of acute kidney injury, obesity, diabetes, heart failure and sepsis. However, the precise formation process of IMD short peptides in vivo and their downstream signaling pathway have not been fully elucidated yet. Further in-depth studies are need to translate IMD research into clinical applications.
Collapse
Affiliation(s)
- Zhi Yang
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongchun Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Pengfei Wu
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingyan Li
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - ChunYan Yu
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Denian Wang
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weimin Li
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Ma S, Yan F, Hou Y. Intermedin 1-53 Ameliorates Atrial Fibrosis and Reduces Inducibility of Atrial Fibrillation via TGF-β1/pSmad3 and Nox4 Pathway in a Rat Model of Heart Failure. J Clin Med 2023; 12:jcm12041537. [PMID: 36836072 PMCID: PMC9959393 DOI: 10.3390/jcm12041537] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
OBJECTIVE New drugs to block the occurrence of atrial fibrillation (AF) based on atrial structural remodeling (ASR) are urgently needed. The purpose of this study was to study the role of intermedin 1-53 (IMD1-53) in ASR and AF formation in rats after myocardial infarction (MI). MATERIAL AND METHODS Heart failure was induced by MI in rats. Fourteen days after MI surgery, rats with heart failure were randomized into control (untreated MI group, n = 10) and IMD-treated (n = 10) groups. The MI group and sham group received saline injections. The rats in the IMD group received IMD1-53, 10 nmol/kg/day intraperitoneally for 4 weeks. The AF inducibility and atrial effective refractory period (AERP) were assessed with an electrophysiology test. Additionally, the left-atrial diameter was determined, and heart function and hemodynamic tests were performed. We detected the area changes of myocardial fibrosis in the left atrium using Masson staining. To detect the protein expression and mRNA expression of transforming growth factor-β1 (TGF-β1), α-SMA, collagen Ⅰ, collagen III, and NADPH oxidase (Nox4) in the myocardial fibroblasts and left atrium, we used the Western blot method and real-time quantitative polymerase chain reaction (PCR) assays. RESULTS Compared with the MI group, IMD1-53 treatment decreased the left-atrial diameter and improved cardiac function, while it also improved the left-ventricle end-diastolic pressure (LVEDP). IMD1-53 treatment attenuated AERP prolongation and reduced atrial fibrillation inducibility in the IMD group. In vivo, IMD1-53 reduced the left-atrial fibrosis content in the heart after MI surgery and inhibited the mRNA and protein expression of collagen type Ⅰ and III. IMD1-53 also inhibited the expression of TGF-β1, α-SMA, and Nox4 both in mRNA and protein. In vivo, we found that IMD1-53 inhibited the phosphorylation of Smad3. In vitro, we found that the downregulated expression of Nox4 was partly dependent on the TGF-β1/ALK5 pathway. CONCLUSIONS IMD1-53 decreased the duration and inducibility of AF and atrial fibrosis in the rats after MI operation. The possible mechanisms are related to the inhibition of TGF-β1/Smad3-related fibrosis and TGF-β1/Nox4 activity. Therefore, IMD1-53 may be a promising upstream treatment drug to prevent AF.
Collapse
Affiliation(s)
- Shenzhou Ma
- Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
- Cardiology Departments, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Feng Yan
- Department of Emergency Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Yinglong Hou
- Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
- Cardiology Departments, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Correspondence:
| |
Collapse
|
5
|
Jung I, Park M, Jeong MH, Park K, Kim WH, Kim GY. Transcriptional analysis of gasoline engine exhaust particulate matter 2.5-exposed human umbilical vein endothelial cells reveals the different gene expression patterns related to the cardiovascular diseases. Biochem Biophys Rep 2022; 29:101190. [PMID: 34988296 PMCID: PMC8695280 DOI: 10.1016/j.bbrep.2021.101190] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 11/28/2022] Open
Abstract
Particulate matter (PM) causes several diseases, including cardiovascular diseases (CVDs). Previous studies compared the gene expression patterns in airway epithelial cells and keratinocytes exposed to PM. However, analysis of differentially expressed gene (DEGs) in endothelial cells exposed to PM2.5 (diameter less than 2.5 μm) from fossil fuel combustion has been limited. Here, we exposed human umbilical vein endothelial cells (HUVECs) to PM2.5 from combustion of gasoline, performed RNA-seq analysis, and identified DEGs. Exposure to the IC50 concentrations of gasoline engine exhaust PM2.5 (GPM) for 24 h yielded 1081 (up-regulation: 446, down-regulation: 635) DEGs. The most highly up-regulated gene is NGFR followed by ADM2 and NUPR1. The most highly down-regulated gene is TNFSF10 followed by GDF3 and EDN1. Gene Ontology enrichment analysis revealed that GPM regulated genes involved in cardiovascular system development, tube development and circulatory system development. Kyoto Encyclopedia of Genes and Genomes and Reactome pathway analyses showed that genes related to cytokine–cytokine receptor interactions and cytokine signaling in the immune system were significantly affected by GPM. We confirmed the RNA-seq data of some highly altered genes by qRT-PCR and showed the induction of NGFR, ADM2 and IL-11 at a protein level, indicating that the observed gene expression patterns were reliable. Given the adverse effects of PM2.5 on CVDs, our findings provide new insight into the importance of several DEGs and pathways in GPM-induced CVDs.
Collapse
Affiliation(s)
- Inkyo Jung
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Minhan Park
- School of Earth Science and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Myong-Ho Jeong
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Kihong Park
- School of Earth Science and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Won-Ho Kim
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Geun-Young Kim
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| |
Collapse
|
6
|
Nagar H, Kim S, Lee I, Choi SJ, Piao S, Jeon BH, Shong M, Kim CS. CRIF1 deficiency suppresses endothelial cell migration via upregulation of RhoGDI2. PLoS One 2021; 16:e0256646. [PMID: 34437633 PMCID: PMC8389428 DOI: 10.1371/journal.pone.0256646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/11/2021] [Indexed: 11/22/2022] Open
Abstract
Rho GDP-dissociation inhibitor (RhoGDI), a downregulator of Rho family GTPases, prevents nucleotide exchange and membrane association. It is responsible for the activation of Rho GTPases, which regulate a variety of cellular processes, such as migration. Although RhoGDI2 has been identified as a tumor suppressor gene involved in cellular migration and invasion, little is known about its role in vascular endothelial cell (EC) migration. CR6-interacting factor 1 (CRIF1) is a CR6/GADD45-interacting protein with important mitochondrial functions and regulation of cell growth. We examined the expression of RhoGDI2 in CRIF1-deficient human umbilical vein endothelial cells (HUVECs) and its role in cell migration. Expression of RhoGDI2 was found to be considerably higher in CRIF1-deficient HUVECs along with suppression of cell migration. Moreover, the phosphorylation levels of Akt and CREB were decreased in CRIF1-silenced cells. The Akt-CREB signaling pathway was implicated in the changes in endothelial cell migration caused by CRIF1 downregulation. In addition to RhoGDI2, we identified another factor that promotes migration and invasion of ECs. Adrenomedullin2 (ADM2) is an autocrine/paracrine factor that regulates vascular tone and other vascular functions. Endogenous ADM2 levels were elevated in CRIF1-silenced HUVECs with no effect on cell migration. However, siRNA-mediated depletion of RhoGDI2 or exogenous ADM2 administration significantly restored cell migration via the Akt-CREB signaling pathway. In conclusion, RhoGDI2 and ADM2 play important roles in the migration of CRIF1-deficient endothelial cells.
Collapse
Affiliation(s)
- Harsha Nagar
- Department of Physiology and Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seonhee Kim
- Department of Physiology and Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of BK21 Plus CNU Integrative Biomedical Education Initiative, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Ikjun Lee
- Department of Physiology and Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of BK21 Plus CNU Integrative Biomedical Education Initiative, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Su-Jeong Choi
- Department of Physiology and Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Shuyu Piao
- Department of Physiology and Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Byeong Hwa Jeon
- Department of Physiology and Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Cuk-Seong Kim
- Department of Physiology and Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
7
|
Li Z, Guo J, Bian Y, Zhang M. Intermedin protects thapsigargin‑induced endoplasmic reticulum stress in cardiomyocytes by modulating protein kinase A and sarco/endoplasmic reticulum Ca 2+‑ATPase. Mol Med Rep 2020; 23:107. [PMID: 33300086 PMCID: PMC7723158 DOI: 10.3892/mmr.2020.11746] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022] Open
Abstract
Intermedin (IMD) is a calcitonin/calcitonin-related peptide that elicits cardioprotective effects in a variety of heart diseases, such as cardiac hypertrophy and heart failure. However, the molecular mechanism of IMD remains unclear. The present study investigated the effects of IMD on neonatal rat ventricular myocytes treated with thapsigargin. The results of the present study demonstrated that thapsigargin induced apoptosis in cardiomyocytes in a dose- and time-dependent manner. Thapsigargin induced endoplasmic reticulum stress, as determined by increased expression levels of 78-kDa glucose-regulated protein, C/EBP-homologous protein and caspase-12, which were dose-dependently attenuated by pretreatment with IMD. In addition, IMD treatment counteracted the thapsigargin-induced suppression of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) activity and protein expression levels, and cytoplasmic Ca2+ overload. IMD treatment also augmented the phosphorylation of phospholamban, which is a crucial regulator of SERCA. Additionally, treatment with the protein kinase A antagonist H-89 inhibited the IMD-mediated cardioprotective effects, including SERCA activity restoration, anti-Ca2+ overload, endoplasmic reticulum stress inhibition and antiapoptosis effects. In conclusion, the results of the present study suggested that IMD may protect cardiomyocytes against thapsigargin-induced endoplasmic reticulum stress and the associated apoptosis at least partly by activating the protein kinase A/SERCA pathway.
Collapse
Affiliation(s)
- Zhidong Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jia Guo
- Department of Cardiology, Shanxi Medical University First Hospital, Taiyuan, Shanxi 030001, P.R. China
| | - Yunfei Bian
- Department of Cardiology, Shanxi Medical University Second Hospital, Taiyuan, Shanxi 030001, P.R. China
| | - Mingsheng Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
8
|
Xu B, Xu H, Cao H, Liu X, Qin C, Zhao Y, Han X, Li H. Intermedin improves cardiac function and sympathetic neural remodeling in a rat model of post myocardial infarction heart failure. Mol Med Rep 2017. [PMID: 28627670 PMCID: PMC5562092 DOI: 10.3892/mmr.2017.6776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Emerging evidence has suggested that intermedin (IMD), a novel member of the calcitonin gene-related peptide (CGRP) family, has a wide range of cardioprotective effects. The present study investigated the effects of long-term administration of IMD on cardiac function and sympathetic neural remodeling in heart failure (HF) rats, and studied potential underlying mechanism. HF was induced in rats by myocardial infarction (MI). Male Sprague Dawley rats were randomly assigned to either saline or IMD (0.6 µg/kg/h) treatment groups for 4 weeks post-MI. Another group of sham-operated rats served as controls. Cardiac function was assessed by echocardiography, cardiac catheterization and plasma level of B-type natriuretic peptide (BNP). Cardiac sympathetic neural remodeling was assessed by immunohistochemistical study of tyrosine hydroxylase (TH) and growth associated protein 43 (GAP43) immunoreactive nerve fibers. The protein expression levels of nerve growth factor (NGF), TH and GAP43 in the ventricular myocardium were studied by western blotting. Ventricular fibrillation threshold (VFT) was determined to evaluate the incidence of ventricular arrhythmia. Oxidative stress was assessed by detecting the activity of superoxide dismutase and the level of malondialdehyde. Compared with rats administrated with saline, IMD significantly improved cardiac function, decreased the plasma BNP level, attenuated sympathetic neural remodeling, increased VFT and suppressed oxidative stress. In conclusion, these results indicated that IMD prevents ventricle remodeling and improves the performance of a failing heart. In addition, IMD attenuated sympathetic neural remodeling and reduced the incidence of ventricular arrhythmia, which may contribute to its anti-oxidative property. These results implicate IMD as a potential therapeutic agent for the treatment of HF.
Collapse
Affiliation(s)
- Bin Xu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Hao Xu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Heng Cao
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Xiaoxiao Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Chunhuan Qin
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Yanzhou Zhao
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Xiaolin Han
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Hongli Li
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| |
Collapse
|
9
|
Zhang SY, Xu MJ, Wang X. Adrenomedullin 2/intermedin: a putative drug candidate for treatment of cardiometabolic diseases. Br J Pharmacol 2017; 175:1230-1240. [PMID: 28407200 DOI: 10.1111/bph.13814] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/15/2017] [Accepted: 03/30/2017] [Indexed: 11/28/2022] Open
Abstract
Adrenomedullin (ADM) 2/intermedin (IMD) is a short peptide that belongs to the CGRP superfamily. Although it shares receptors with CGRP, ADM and amylin, ADM2 has significant and unique functions in the cardiovascular system. In the past decade, the cardiovascular effect of ADM2 has been carefully analysed. In this review, progress in understanding the effects of ADM2 on the cardiovascular system and its protective role in cardiometabolic diseases are summarized. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Song-Yang Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
| | - Ming-Jiang Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
| |
Collapse
|
10
|
Zhang H, Zhang SY, Jiang C, Li Y, Xu G, Xu MJ, Wang X. Intermedin/adrenomedullin 2 polypeptide promotes adipose tissue browning and reduces high-fat diet-induced obesity and insulin resistance in mice. Int J Obes (Lond) 2016; 40:852-60. [PMID: 26786353 DOI: 10.1038/ijo.2016.2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 11/06/2015] [Accepted: 12/06/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVES There is an urgent need to develop interventions and policies to mitigate the health effects of obesity by targeting its metabolic mediators. Adrenomedullin 2 (AM2)/intermedin (IMD) is a secreted peptide that has an important role in protecting the cardiovascular system. However, the role of AM2 in type 2 diabetes is unknown. METHODS Wild-type (WT) and aP2/AM2 transgenic (aAM2-tg) mice were fed a high-fat diet (HFD) for 8 weeks, and WT mice were treated with AM2 through mini-osmotic pumps. Indirect calorimetry, ambulatory activity and food intake, hyperinsulinemic-euglycemic clamp test, glucose and insulin tolerance tests were used for assessing insulin resistance. Rat primary adipocytes and pre-adipocyte-derived adipocytes were used for in vitro experiments. Real-time PCR and western blot were used for analyses of gene expression and protein level. RESULTS AM2 and receptor activity-modifying proteins expressions were significantly decreased in the adipose tissue of obese mice. AM2 treatment significantly reduced blood glucose, fasting serum insulin and free fatty acid levels, improved glucose tolerance and insulin sensitivity, and increased the glucose infusion rate during a hyperinsulinemic-euglycemic clamp test, indicating ameliorated HFD-induced insulin resistance. These effects were consistently observed in aAM2-tg mice under HFD conditions, whereas the aAM2-tg mice showed less weight gain and improved glucose tolerance and insulin sensitivity. More importantly, the aAM2-tg mice had increased oxygen consumption and CO2 production, reflecting more energy expenditure. These effects may be due to increased AMP-activated protein kinase phosphorylation and reduced peroxisome proliferator-activated receptor gamma co-activator 1α (PGC1α) acetylation, which result in interactions between PGC1α and PR domain containing 16 and then promote uncoupling protein 1 (UCP1) expression in adipocytes. CONCLUSIONS These results indicate that endogenous AM2 might be involved in energy metabolism in adipocytes through the upregulation of UCP1 expression.
Collapse
Affiliation(s)
- H Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - S-Y Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - C Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Y Li
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - G Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - M-J Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - X Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
11
|
Yamac AH, Bacaksiz A, Ismailoglu Z, Kucukbuzcu S, Sevgili E, Asoglu E, Nasifov M, Jafarov P, Erdogan E, Goktekin O. Implication of plasma intermedin levels in patients who underwent first-time diagnostic coronary angiography: a single centre, cross-sectional study. BMC Cardiovasc Disord 2014; 14:182. [PMID: 25495100 PMCID: PMC4271361 DOI: 10.1186/1471-2261-14-182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/26/2014] [Indexed: 11/10/2022] Open
Abstract
Background Intermedin (IMD) is involved in the prevention of atherosclerotic plaque progression, possessing cardioprotective effects from hypertrophy, fibrosis and ischemia-reperfusion injury. Elevated plasma IMD levels have been demonstrated in patients with acute coronary syndromes. No human study has examined the role of IMD in stable patients who underwent diagnostic coronary angiography with suspicion of coronary artery disease (CAD). Thus we investigated the role of IMD as a biomarker to discriminate patients with CAD and predict those with severe disease who require early and intensive therapeutic intervention before presenting with acute coronary syndrome. Methods Eligible two hundred and thirty-eight consecutive patients (123 males, mean age 58.4 ± 10.0 years) who underwent first-time diagnostic coronary angiography were included in this study. Plasma concentrations of IMD were measured from arterial blood samples by the enzyme-linked immunosorbent assay. Patients were divided into three groups according to the presence and degree of CAD, consisting of 48 patients with normal coronary anatomy (Group 1), 111 patients with < 50% coronary stenosis (Group 2), and 79 patients with ≥ 50% stenosis in at least one of the major coronary arteries (group 3). The severity and extent of CAD was evaluated by calculations of the vessel, Gensini, and SYNTAX scores. Results Circulating plasma IMD levels in patients with CAD were significantly higher than those in patients without CAD (157.7 ± 9.6, 134.8 ± 11.9, and 117.6 ± 7.9 pg/mL in groups 3, 2 and 1 respectively; p < 0.001). Besides, plasma IMD levels were correlated with Gensini and SYNTAX scores (rs = 0.742, and rs = 0.296, respectively; p < 0.05). The presence of ≥50% coronary artery stenosis could be predicted if a cut-off value of 147.7 pg/mL for plasma IMD was used with 88.6% sensitivity and 88.7% specificity. Moreover, a plasma IMD level of <126.6 pg/mL could discriminate a patient with normal coronary arteries from patients with angiographically proven CAD with a sensitivity and specificity of 84.7%, and 83.3% respectively. Conclusions We demonstrated that IMD might be used as a biomarker to predict CAD and its severity in patients who underwent first time diagnostic coronary angiography.
Collapse
Affiliation(s)
- Aylin Hatice Yamac
- Faculty of Medicine, Department of Cardiology, BezmiÂlem Foundation University, Adnan Menderes Avenue, Vatan Street, 34093 Fatih, Istanbul, Turkey.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Dvorakova MC, Kruzliak P, Rabkin SW. Role of neuropeptides in cardiomyopathies. Peptides 2014; 61:1-6. [PMID: 25149360 DOI: 10.1016/j.peptides.2014.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 08/09/2014] [Accepted: 08/11/2014] [Indexed: 01/19/2023]
Abstract
The role of neuropeptides in cardiomyopathy-associated heart failure has been garnering more attention. Several neuropeptides--Neuropeptide Y (NPY), vasoactive intestinal peptide (VIP), calcitonin gene related peptide (CGRP), substance P (SP) and their receptors have been studied in the various types of cardiomyopathies. The data indicate associations with the strength of the association varying depending on the kind of neuropeptide and the nature of the cardiomyopathy--diabetic, ischemic, inflammatory, stress-induced or restrictive cardiomyopathy. Several neuropeptides appear to alter regulation of genes involved in heart failure. Demonstration of an association is an essential first step in proving causality or establishing a role for a factor in a disease. Understanding the complexity of neuropeptide function should be helpful in establishing new or optimal therapeutic strategies for the treatment of heart failure in cardiomyopathies.
Collapse
Affiliation(s)
- Magdalena Chottova Dvorakova
- Department of Physiology, Charles University in Prague, Faculty of Medicine in Pilsen, Lidicka 1, 301 00 Pilsen, Czech Republic; Biomedical Centre, Faculty of Medicine in Pilsen, Charles University in Prague, Lidicka 1, 301 00 Pilsen, Czech Republic
| | - Peter Kruzliak
- Department of Cardiovascular Diseases, International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Pekarska 53, 656 91 Brno, Czech Republic.
| | - Simon W Rabkin
- Department of Medicine Division of Cardiology, University of British Columbia, 2329W Mall, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
13
|
Nagasaki S, Fukui M, Asano S, Ono K, Miki Y, Araki SI, Isobe M, Nakashima N, Takahashi K, Sasano H, Sato J. Induction of adrenomedullin 2/intermedin expression by thyroid stimulating hormone in thyroid. Mol Cell Endocrinol 2014; 395:32-40. [PMID: 25102228 DOI: 10.1016/j.mce.2014.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 10/24/2022]
Abstract
TSH is the important regulator of thyroid function but detailed molecular mechanisms have not been clarified. We first generated the iodine deficient (ID) rat in which goiter is induced by accelerated endogenous TSH secretion. The result of microarray analysis demonstrated markedly increased levels of adrenomedullin 2/intermedin (AM2/IMD) expression in the ID rat thyroid. AM2/IMD is a potent vasodilator. AM2/IMD mRNA expression was induced by TSH in a rat thyroid follicular cell line FRTL-5. Immunohistochemical analysis in human normal and Graves' disease thyroid revealed that AM2/IMD immunoreactivity was detected in follicular cells and more pronounced in Graves' disease. These results indicated that TSH induced AM2/IMD expression in the rat thyroid gland and it could locally work as a potent vasodilator, resulting in the expansion of thyroid inter-follicular capillaries. AM2/IMD could also contribute to facilitate thyroid hormone synthesis possibly via vasodilation effects and/or cAMP stimulating effects in the human thyroid gland.
Collapse
Affiliation(s)
- Shuji Nagasaki
- Drug Discovery Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan; Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Motoko Fukui
- Safety Research Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan
| | - Satoko Asano
- Drug Discovery Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan
| | - Katsuhiko Ono
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Miki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sei-ichi Araki
- Safety Research Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan
| | - Mitsui Isobe
- Safety Research Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan
| | - Noriaki Nakashima
- Department of Breast and Endocrine Surgery, Tohoku University Hospital, Sendai, Japan
| | - Kazuhiro Takahashi
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jun Sato
- Drug Discovery Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan
| |
Collapse
|
14
|
Tang B, Zhong Z, Shen HW, Wu HP, Xiang P, Hu B. Intermedin as a prognostic factor for major adverse cardiovascular events in patients with ST-segment elevation acute myocardial infarction. Peptides 2014; 58:98-102. [PMID: 24969626 DOI: 10.1016/j.peptides.2014.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 06/06/2014] [Accepted: 06/06/2014] [Indexed: 12/15/2022]
Abstract
Intermedin functions systemically as a potent vasodilator and its plasma levels have been shown to be elevated in patients with acute myocardial infarction. This study aimed to evaluate the prognostic value of plasma intermedin level in the patients with ST-segment elevation acute myocardial infarction. Plasma intermedin concentrations of 128 patients and 128 healthy controls were determined using a radioimmunoassay. Patients were followed up for 6 months for major adverse cardiovascular events (MACE) consisting of cardiovascular mortality, reinfarction, hospitalization for decompensated heart failure, and lift-threatening arrhythmia. The association of plasma intermedin levels with MACE was investigated by univariate and multivariate analyses. Plasma intermedin levels were significantly higher in patients than in healthy subjects. Elevated plasma level of intermedin was identified as an independent predictor of MACE. Receiver operating characteristic curve analysis showed that plasma intermedin levels had high predictive value for MACE. Moreover, its predictive value was similar to Global Registry of Acute Coronary Events scores' based on area under curve. Meantime, it obviously improved Global Registry of Acute Coronary Events scores' predictive value in a combined logistic-regression model. In multivariate Cox's proportional hazard analysis, plasma intermedin level emerged as an independent predictor of MACE-free survival. Thus, our results suggest that high plasma intermedin level is associated with poor outcomes of patients and may be a useful prognostic biomarker in ST-segment elevation acute myocardial infarction.
Collapse
Affiliation(s)
- Bei Tang
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| | - Ze Zhong
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China.
| | - Hong-Wei Shen
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| | - Hui-Ping Wu
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| | - Peng Xiang
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| | - Bin Hu
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| |
Collapse
|
15
|
Dai XY, Cai Y, Sun W, Ding Y, Wang W, Kong W, Tang C, Zhu Y, Xu MJ, Wang X. Intermedin inhibits macrophage foam-cell formation via tristetraprolin-mediated decay of CD36 mRNA. Cardiovasc Res 2013; 101:297-305. [PMID: 24253523 DOI: 10.1093/cvr/cvt254] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS CD36-mediated uptake of oxidized low-density lipoprotein (oxLDL) plays a pivotal role in macrophage foam-cell formation and atherogenesis. Previously we reported on intermedin (IMD), a novel member of the calcitonin gene-related peptide family, in atherosclerotic plaque reducing atherogenesis in apolipoprotein E-deficient (apoE(-/-)) mice. Here, we studied the role of IMD in CD36-mediated macrophage foam-cell formation. METHODS AND RESULTS In apoE(-/-) mice, 6-week IMD infusion reduced oxLDL uptake, intracellular cholesterol content, and foam-cell formation in peritoneal macrophages and reduced protein and mRNA levels of CD36. These in vivo results agreed with in vitro observations in primary peritoneal macrophages. Reduced CD36 protein and mRNA levels were due to an IMD-accelerated decay of CD36 mRNA. Tristetraprolin (TTP), which binds to AU-rich elements in the 3' untranslated regions (UTRs) of mRNA and promotes its degradation, mediated CD36 mRNA destabilization. TTP knockdown by short-hairpin RNA increased and TTP overexpression reduced CD36 expression, and TTP knockdown rescued IMD-reduced CD36 expression. Moreover, IMD repressed TTP phosphorylation, thereby activating TTP, for increased TTP binding to the 3'-UTR of CD36 mRNA. CONCLUSION Thus, IMD attenuates macrophage foam-cell formation via TTP-mediated degradation of CD36 mRNA. Our findings reveal a new mechanism of the anti-atherogenic role of IMD and a novel pattern for regulation of CD36 expression in macrophages.
Collapse
Affiliation(s)
- Xiao-Yan Dai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Li H, Bian Y, Zhang N, Guo J, Wang C, Lau WB, Xiao C. Intermedin protects against myocardial ischemia-reperfusion injury in diabetic rats. Cardiovasc Diabetol 2013; 12:91. [PMID: 23777472 PMCID: PMC3703263 DOI: 10.1186/1475-2840-12-91] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 06/14/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Diabetic patients, through incompletely understood mechanisms, endure exacerbated ischemic heart injury compared to non-diabetic patients. Intermedin (IMD) is a novel calcitonin gene-related peptide (CGRP) superfamily member with established cardiovascular protective effects. However, whether IMD protects against diabetic myocardial ischemia/reperfusion (MI/R) injury is unknown. METHODS Diabetes was induced by streptozotocin in Sprague-Dawley rats. Animals were subjected to MI via left circumflex artery ligation for 30 minutes followed by 2 hours R. IMD was administered formally 10 minutes before R. Outcome measures included left ventricular function, oxidative stress, cellular death, infarct size, and inflammation. RESULTS IMD levels were significantly decreased in diabetic rats compared to control animals. After MI/R, diabetic rats manifested elevated intermedin levels, both in plasma (64.95 ± 4.84 pmol/L, p < 0.05) and myocardial tissue (9.8 ± 0.60 pmol/L, p < 0.01) compared to pre-MI control values (43.62 ± 3.47 pmol/L and 4.4 ± 0.41). IMD administration to diabetic rats subjected to MI/R decreased oxidative stress product generation, apoptosis, infarct size, and inflammatory cytokine release (p < 0.05 or p < 0.01). CONCLUSIONS By reducing oxidative stress, inflammation, and apoptosis, IMD may represent a promising novel therapeutic target mitigating diabetic ischemic heart injury.
Collapse
MESH Headings
- Adrenomedullin/metabolism
- Adrenomedullin/pharmacology
- Animals
- Apoptosis/drug effects
- Cardiotonic Agents/pharmacology
- Cytokines/metabolism
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/prevention & control
- Inflammation Mediators/metabolism
- Male
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocardial Infarction/prevention & control
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/physiopathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Neuropeptides/metabolism
- Neuropeptides/pharmacology
- Oxidative Stress/drug effects
- Rats, Sprague-Dawley
- Streptozocin
- Time Factors
- Ventricular Function, Left/drug effects
Collapse
Affiliation(s)
- Hong Li
- Department of Cardiology, Shanxi Medical University, 030001 Taiyuan, Shanxi, China
| | - Yunfei Bian
- Department of Cardiology, Shanxi Medical University, 030001 Taiyuan, Shanxi, China
| | - Nana Zhang
- Department of Cardiology, Shanxi Medical University, 030001 Taiyuan, Shanxi, China
| | - Jia Guo
- Department of Cardiology, Shanxi Medical University, 030001 Taiyuan, Shanxi, China
| | - Cheng Wang
- Department of Cardiology, Shanxi Medical University, 030001 Taiyuan, Shanxi, China
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Chuanshi Xiao
- Department of Cardiology, Shanxi Medical University, 030001 Taiyuan, Shanxi, China
| |
Collapse
|
17
|
Lv Z, Wu K, Chen X, Zhang X, Hong B. Plasma intermedin levels in patients with acute myocardial infarction. Peptides 2013; 43:121-5. [PMID: 23499766 DOI: 10.1016/j.peptides.2013.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 03/07/2013] [Accepted: 03/07/2013] [Indexed: 11/21/2022]
Abstract
It has been shown that adrenomedullin (ADM) may function as a cardiovascular-regulatory peptide in humans. Intermedin (IMD) is a newly discovered peptide related to ADM and has a greater range of biological effects on the cardiovascular in animal experiments. The purpose of the study was to investigate the pathophysiological role of IMD in patients with acute myocardial infarction (AMI). The present study included twenty patients with acute ST-segment elevation myocardial infarction (STEMI), thirty-three with stable coronary heart disease (SCHD), and eighteen healthy controls. Plasma levels of IMD, malonaldehyde (MDA), and superoxide dismutase (SOD) and cardiac biomarkers were determined at one, two, four and seven days following AMI. Plasma IMD levels were significantly increased on day 1 in AMI patients when compared with SCHD subjects (P=0.014), and reached a peak of 181.88 ± 9.47 pg/ml at 96 h. Plasma IMD concentrations were correlated with MDA and SOD. Furthermore, patients with severe lesions in their coronary arteries tended to have higher plasma IMD levels (P<0.05) in AMI patients. A significant increase in plasma IMD following AMI may be associated with oxidative stress, and could be used as a marker to reflect the severity of the coronary stenosis.
Collapse
Affiliation(s)
- Zhengbing Lv
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | | | |
Collapse
|
18
|
Qin YW, Teng X, He JQ, Du J, Tang CS, Qi YF. Increased plasma levels of intermedin and brain natriuretic peptide associated with severity of coronary stenosis in acute coronary syndrome. Peptides 2013; 42:84-8. [PMID: 23391507 DOI: 10.1016/j.peptides.2013.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 01/21/2013] [Accepted: 01/21/2013] [Indexed: 02/07/2023]
Abstract
Intermedin (IMD) is a newly discovered peptide with increased levels in plasma and cardiac tissue in mice with ischemia/reperfusion. Continuous administration of low dose IMD markedly elevated the mRNA abundance of myocardial BNP in rats. Plasma BNP levels may reflect the severity of degree of coronary stenosis in patients with acute coronary syndrome (ACS). However, the role of circulating IMD in coronary heart disease remains unclear. We aimed to examine the plasma content of IMD and brain natriuretic peptide (BNP) and its clinical significance in patients with ACS. We collected plasma samples from 41 patients with ACS and 31 controls and measured IMD and BNP levels by radioimmunoassay. The severity of coronary artery stenosis for patients with ACS was measured by coronary angiography. Plasma IMD and BNP levels were markedly higher in ACS patients than that in controls (P<0.05). The increased plasma IMD and BNP were positively correlated with degree of coronary stenosis in ACS patients (r=0.263 and r=0.238, respectively, both P<0.05). In addition, plasma levels of IMD were positively correlated with BNP levels.
Collapse
Affiliation(s)
- Yan-Wen Qin
- The Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | | | | | | | | | | |
Collapse
|
19
|
Pires AL, Pinho M, Alves BS, Pinho S, Sena C, Seica RM, Leite-Moreira AF. Reverse myocardial effects of intermedin in pressure-overloaded hearts: role of endothelial nitric oxide synthase activity. J Physiol 2013; 591:677-687. [PMID: 23165766 PMCID: PMC3577549 DOI: 10.1113/jphysiol.2012.240812] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 11/14/2012] [Indexed: 11/08/2022] Open
Abstract
Intermedin (IMD) is a cardiac peptide synthesized in a prepro form, which undergoes a series of proteolytic cleavages and amidations to yield the active forms of 47 (IMD(1-47)) and 40 amino acids (IMD(8-47)). There are several lines of evidence of increased IMD expression in rat models of cardiac pathologies, including congestive heart failure and ischaemia; however, its myocardial effects upon cardiac disease remain unexplored. With this in mind, we investigated the direct effects of increasing concentrations of IMD(1-47) (10(-10) to10(-6) m) on contraction and relaxation of left ventricular (LV) papillary muscles from two rat models of chronic pressure overload, one induced by transverse aortic constriction (TAC), the other by nitric oxide (NO) deficiency due to chronic NO synthase inhibition (NG-nitro-l-arginine, l-NAME), and respective controls (Sham and Ctrl). In TAC and l-NAME rats, exogenous administration of IMD(1-47) elicited concentration-dependent positive inotropic and lusitropic effects. By contrast, in Sham and Ctrl rats, IMD(1-47) induced a negative inotropic response without a significant effect on relaxation. Both TAC and l-NAME rats presented LV hypertrophy, elevated LV systolic pressures, preserved systolic function and elevated peroxynitrite levels. In the normal myocardium (Ctrl and Sham), IMD(1-47) induced a 3-fold increase of endothelial nitric oxide synthase (eNOS) phosphorylation at Ser(1177), indicating enhanced eNOS activity. In TAC and l-NAME rats, eNOS phosphorylation was increased at baseline, and its response to IMD(1-47) was blunted. In addition, the distinct myocardial response to IMD(1-47) was accompanied by distinct subcellular mechanisms. While in Sham rats the addition of IMD(1-47) induced the phosphorylation of cardiac troponin I due to NO/cGMP activation, in TAC rats IMD(1-47) induced phospholamban phosphorylation possibly associated with cAMP/protein kinase A activation. Therefore, we demonstrated for the first time a reversed myocardial response to IMD(1-47) neurohumoral stimulation due to impairment of eNOS activation in TAC and l-NAME rats. These results not only reveal the distinct myocardial effects and subcellular mechanisms for IMD(1-47) in normal and hypertrophic hearts, but also highlight the potential pathophysiological relevance of cardiac endothelial dysfunction in neurohumoral myocardial action.
Collapse
Affiliation(s)
- Ana Luísa Pires
- Department of Physiology and Cardiothoracic Surgery, Cardiovascular R&D Unit, Faculty of Medicine, University of Porto, Portugal
| | | | | | | | | | | | | |
Collapse
|
20
|
Yang X, Zhang H, Jia Y, Ni L, Li G, Xue L, Jiang Y. Effects of intermedin1-53 on myocardial fibrosis. Acta Biochim Biophys Sin (Shanghai) 2013; 45:141-8. [PMID: 23174675 DOI: 10.1093/abbs/gms093] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intermedin (IMD) is a member of the calcitonin/calcitonin gene-related peptide (CGRP) family and has similar or more potent cardiovascular actions than adrenomedullin (ADM) and any other CGRP. The aim of the present work is to study the effects of IMD1-53 on cardiac fibroblast fibrosis in vivo and in vitro. Myocardial infarction model was prepared by ligating rats' left anterior descending coronary artery. Mesenchymal collagen contents in the left ventricle were accessed by Sirius-red stain. Heart functions were explored by hemodynamic changes. Expression of I and III type collagens, IMD1-53, receptor activity-modifying proteins (RAMP)1/2/3, and calcitonin receptor-like receptor (CRLR) in left ventricle were detected by western blot analysis. Cardiac fibroblasts (CFbs) fibrosis was induced by treating the cells with aldosterone (ALD). CFbs proliferation and the hydroxyproline contents in supernatants were determined by 3-[4,5-dimehyl-2-thiazolyl]-2,5-diphenyl-2H-tetrazolium bromide assay and enzyme-linked immunosorbent assay. Heart function was decreased in myocardial infarction model rats. Expression of type I and type III collagens in infarcted zone in myocardial rats was higher than those in the sham-operated group. IMD1-53, RAMP, and CRLR in left ventricle were also up-regulated. In vitro experiment showed that ALD was a powerful stimulator of CFbs activation. IMD1-53 decreased ALD-induced CFbs proliferation in a dose-dependent manner. Moreover, CGRP8-37 and ADM22-52 remarkably blocked the effect of IMD1-53 on ALD-induced myocardial cell fibrosis. IMD could be involved in the onset of cardiac fibrosis. Like ADM, IMD1-53 exerts an antifibrotic effect on CFbs, which might be mediated by CRLR/RAMP complex and ADM receptor.
Collapse
Affiliation(s)
- Xiaoling Yang
- Department of Pathophysiology, Basic Medical School, Ningxia Medical University, Yinchuan 750004, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Pires AL, Pinho M, Sena CM, Seica R, Leite-Moreira AF. Intermedin elicits a negative inotropic effect in rat papillary muscles mediated by endothelial-derived nitric oxide. Am J Physiol Heart Circ Physiol 2012; 302:H1131-H1137. [PMID: 22227127 DOI: 10.1152/ajpheart.00877.2011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Intermedin (IMD) is a novel vasoactive peptide from the calcitonin gene-related peptide (CGRP) implicated in cardiac regulation, yet the contractile effects of IMD remain controversial, since previous studies in vivo and isolated cardiomyocytes documented contradictory results. We hypothesized cardiac endothelial cells involvement in IMD modulation of cardiac function as an explanation for these opposing observations. With this in mind, we investigated the direct action of increasing concentrations of IMD (10(-8) to 10(-6)M) on myocardial performance parameters in rat left ventricular (LV) papillary muscles with and without endocardial endothelium (EE) and in presence of receptor antagonists and intracellular pathways inhibitors. In LV papillary muscles with intact EE, IMD induced a concentration-dependent negative inotropic action (%decrease relative to baseline, at IMD concentration of 10(-6)M, active tension of 14 ± 4%, and maximum velocity of tension rise of 10 ± 4%). These effects were blunted by EE removal, AM receptor antagonist (AM(22-52)), and CGRP receptor antagonist (CGRP(8-37)). Additionally, nitric oxide (NO) synthase inhibition with N(G)-nitro-l-arginine (l-NAME) in muscles with and without EE and guanylyl cyclase inhibition with {1H-[1,2,4]oxadiazole-[4,4-a]-quinoxalin-1-one} not only blunted the negative inotropic action of IMD but also unmasked IMD-positive inotropic effect dependent on CGRP receptor PKA activation. Western blot quantification of phosphorylated cardiac troponin I (P-cTnI) in IMD-treated papillary muscles revealed a significant increase in P-cTnI when compared with untreated muscles, while in l-NAME-pretreated papillary muscles IMD failed to increase P-cTnI. Finally, we found that stimulation of both EE and microvascular endothelial cells with IMD significantly increased NO production by 40 ± 3 and 38 ± 3%, respectively, suggesting the role of cardiac endothelial cells in NO production upon IMD stimulation. Our findings establish IMD negative inotropic effect in isolated myocardium due to NO/cGMP pathway activation with concomitant thin myofilament desensitization by increase in cTnI phosphorylation and provide a coherent explanation for the previously reported contradictory results.
Collapse
Affiliation(s)
- Ana Luísa Pires
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | | | | | | | | |
Collapse
|
22
|
Bell D, Campbell M, Ferguson M, Sayers L, Donaghy L, O'Regan A, Jewhurst V, Harbinson M. AM₁-receptor-dependent protection by intermedin of human vascular and cardiac non-vascular cells from ischaemia-reperfusion injury. J Physiol 2011; 590:1181-97. [PMID: 22183724 DOI: 10.1113/jphysiol.2011.221895] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Intermedin (IMD) protects rodent heart and vasculature from oxidative stress and ischaemia. Less is known about distribution of IMD and its receptors and the potential for similar protection in man. Expression of IMD and receptor components were studied in human aortic endothelium cells (HAECs), smooth muscle cells (HASMCs), cardiac microvascular endothelium cells (HMVECs) and fibroblasts (v-HCFs). Receptor subtype involvement in protection by IMD against injury by hydrogen peroxide (H₂O₂, 1 mmol l⁻¹) and simulated ischaemia and reperfusion were investigated using receptor component-specific siRNAs. IMD and CRLR, RAMP1, RAMP2 and RAMP3 were expressed in all cell types.When cells were treated with 1 nmol l⁻¹ IMD during exposure to 1 mmol l⁻¹ H₂O₂ for 4 h, viability was greater vs. H2O2 alone (P<0.05 for all cell types). Viabilities under 6 h simulated ischaemia differed (P<0.05) in the absence and presence of 1 nmol l⁻¹ IMD: HAECs 63% and 85%; HMVECs 51% and 68%; v-HCFs 42% and 96%. IMD 1 nmol l⁻¹ present throughout ischaemia (3 h) and reperfusion (1 h) attenuated injury (P<0.05): viabilities were 95%, 74% and 82% for HAECs, HMVECs and v-HCFs, respectively, relative to those in the absence of IMD (62%, 35%, 32%, respectively). When IMD 1 nmol l⁻¹ was present during reperfusion only, protection was still evident (P<0.05, 79%, 55%, 48%, respectively). Cytoskeletal disruption and protein carbonyl formation followed similar patterns. Pre-treatment (4 days) of HAECs with CRLR or RAMP2, but not RAMP1 or RAMP3, siRNAs abolished protection by IMD (1 nmol l⁻¹) against ischaemia-reperfusion injury. IMD protects human vascular and cardiac non-vascular cells from oxidative stress and ischaemia-reperfusion,predominantly via AM1 receptors.
Collapse
Affiliation(s)
- David Bell
- Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Northern Ireland, UK.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Rat intermedin1-47 does not improve functional recovery in postischemic hearts. Naunyn Schmiedebergs Arch Pharmacol 2011; 384:535-42. [PMID: 21881857 DOI: 10.1007/s00210-011-0680-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 08/10/2011] [Indexed: 10/17/2022]
Abstract
Intermedin, a novel member of the calcitonin/calcitonin gene-related peptide family identified from vertebrate genomes, may directly affect cardiac function but current studies revealed no clear picture. The aims of our study were to compare direct contractile effects of intermedin on cardiomyocytes to that on the whole organ and to investigate whether intermedin improves postischemic recovery independent of an effect on acute reperfusion injury. Isolated adult rat ventricular cardiomyocytes were electrically paced and cell shortening was monitored as a readout associated to cardiac performance. Calcium transients were analyzed by Fura-2AM loading of these cells. Isolated rat hearts were investigated by Langendorff perfusion under nonischemic conditions and after 45-min no-flow ischemia followed up by 30-min reperfusion prior to drug testing. Intermedin caused a positive contractile effect on cardiomyocytes that was mediated by protein kinase A activation and accompanied by improved calcium transients. In contrast, intermedin reduced left ventricular developed pressure in Langendorff-perfused rat hearts. This negative inotropic effect was attenuated by inhibition of nitric oxide synthesis. In postischemic hearts (impaired nitric oxide synthesis), the negative inotropic effect was attenuated but no positive inotropic effect occurred. However, intermedin caused robust vasodilation in nonischemic and postischemic hearts. Our findings suggest that the peptide binds preferentially to vascular cells in the intact organ. The loss of nitric oxide induction in postischemic hearts attenuates a negative inotropic effect of intermedin but does not improve cardiac performance independent of acute reperfusion injury.
Collapse
|
24
|
Aslam M, Gündüz D, Schuler D, Li L, Sharifpanah F, Sedding D, Piper HM, Noll T. Intermedin induces loss of coronary microvascular endothelial barrier via derangement of actin cytoskeleton: role of RhoA and Rac1. Cardiovasc Res 2011; 92:276-86. [PMID: 21816966 DOI: 10.1093/cvr/cvr213] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
AIMS Intermedin (IMD) is a novel member of the calcitonin gene-related peptide family, which acts via calcitonin receptor-like receptors (CLRs), mediating activation of cAMP signalling. The main objective of the present study was to analyse the molecular mechanisms of the differential effects of IMD on the macromolecule permeability of endothelial cells of different vascular beds. METHODS AND RESULTS Here we demonstrate that IMD increases permeability of rat coronary microvascular endothelial cells (RCECs) and reduces permeability of human umbilical vein endothelial cells (HUVECs) and rat aortic endothelial cells via CLRs and cAMP. Intermedin causes a derangement of the actin cytoskeleton accompanied by loss of vascular endothelial cadherin (VE-cadherin) in RCECs, while it causes a rearrangement of the actin cytoskeleton and VE-cadherin at cell-cell junctions in HUVECs. Intermedin inactivates the RhoA/Rho-kinase (Rock) pathway in both cell types; however, it inactivates Rac1 in RCECs but not in HUVECs. Inhibition and rescue experiments demonstrate that both RhoA and Rac1 are required for the RCEC barrier stability, while in HUVECs the inhibition of RhoA/Rock signalling does not interfere with basal permeability. CONCLUSION The opposite effects of IMD on permeability of RCECs and HUVECs are due to differential regulation of actin cytoskeleton dynamics via RhoA and Rac1. Moreover, Rac1 activity is regulated by the RhoA/Rock pathway in RCECs but not in HUVECs.
Collapse
Affiliation(s)
- Muhammad Aslam
- Physiologisches Institut, Justus-Liebig-Universität, Aulweg 129, D-35392 Giessen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kuwasako K, Kitamura K, Nagata S, Hikosaka T, Takei Y, Kato J. Shared and separate functions of the RAMP-based adrenomedullin receptors. Peptides 2011; 32:1540-50. [PMID: 21645567 DOI: 10.1016/j.peptides.2011.05.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 05/20/2011] [Accepted: 05/20/2011] [Indexed: 11/25/2022]
Abstract
Adrenomedullin (AM) is a novel hypotensive peptide that exerts a variety of strongly protective effects against multiorgan damage. AM-specific receptors were first identified as heterodimers composed of calcitonin-receptor-like receptor (CLR), a G protein coupled receptor, and one of two receptor activity-modifying proteins (RAMP2 or RAMP3), which are accessory proteins containing a single transmembrane domain. RAMPs are required for the surface delivery of CLR and the determination of its phenotype. CLR/RAMP2 (AM₁ receptor) is more highly AM-specific than CLR/RAMP3 (AM₂ receptor). Although there have been no reports showing differences in intracellular signaling via the two AM receptors, in vitro studies have shed light on their distinct trafficking and functionality. In addition, the tissue distributions of RAMP2 and RAMP3 differ, and their gene expression is differentially altered under pathophysiological conditions, which is suggestive of the separate roles played by AM₁ and AM₂ receptors in vivo. Both AM and the AM₁ receptor, but not the AM₂ receptor, are crucial for the development of the fetal cardiovascular system and are able to effectively protect against various vascular diseases. However, AM₂ receptors reportedly play an important role in maintaining a normal body weight in old age and may be involved in immune function. In this review article, we focus on the shared and separate functions of the AM receptor subtypes and also discuss the potential for related drug discovery. In addition, we mention their possible function as receptors for AM2 (or intermedin), an AM-related peptide whose biological functions are similar to those of AM.
Collapse
Affiliation(s)
- Kenji Kuwasako
- Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, Miyazaki 889-1692, Japan.
| | | | | | | | | | | |
Collapse
|
26
|
Affiliation(s)
- Daniela Tirziu
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8017, USA
| | | | | |
Collapse
|
27
|
Adrenomedullin 2/Intermedin in the Hypothalamo–Pituitary–Adrenal Axis. J Mol Neurosci 2010; 43:182-92. [DOI: 10.1007/s12031-010-9413-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Accepted: 06/11/2010] [Indexed: 10/19/2022]
|
28
|
Hirose T, Totsune K, Mori N, Mori T, Morimoto R, Metoki H, Asayama K, Kikuya M, Ohkubo T, Kohzuki M, Takahashi K, Imai Y. Expression of adrenomedullin 2/intermedin, a possible reno-protective peptide, is decreased in the kidneys of rats with hypertension or renal failure. Am J Physiol Renal Physiol 2010; 299:F128-34. [PMID: 20462970 DOI: 10.1152/ajprenal.00679.2009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adrenomedullin 2/intermedin (AM2/IMD) is a potent vasodilator peptide with organ-protective effects and is abundantly expressed in the kidney. We examined the expression of AM2/IMD in the kidneys of rats with hypertension or chronic renal impairment using quantitative RT-PCR, radioimmunoassay, and immunohistochemistry. Kidneys of 8-wk-old male spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats were dissected into inner medulla, outer medulla, cortex, and glomerulus fractions. A rat renal impairment model was prepared by 5/6 nephrectomy in WKY rats. AM2/IMD mRNA levels were the highest in the cortex among four renal portions, and significantly lower in SHR than WKY rats in all renal portions. In the remnant kidneys of 5/6 nephrectomized rats, AM2/IMD mRNA levels were significantly decreased on days 3 and 56, whereas mRNA levels of calcitonin receptor-like receptor, receptor activity-modifying proteins-1 and -2, which form receptor for AM and AM2/IMD, were increased, compared with that in sham-operated rats. AM mRNA levels were decreased on day 3, but increased on day 56, after nephrectomy. Decreased immunoreactive AM2/IMD levels in the remnant kidneys of 5/6 nephrectomized rats on day 56 were confirmed by radioimmunoassay. The renal tubules were immunostained with anti-AM2/IMD antibody, with a decreased AM2/IMD immunostaining found in proximal tubular cells of 5/6 nephrectomized rats compared with sham-operated rats. In conclusion, intrarenal AM2/IMD expression is decreased in SHR and 5/6 nephrectomized rats. Given the organ-protective effects of AM2/IMD, the downregulation of AM2/IMD as an endogenous regulatory peptide may have a role in the progression of renal impairment.
Collapse
Affiliation(s)
- Takuo Hirose
- Dept. of Clinical Pharmacology and Therapeutics, Tohoku Univ. Graduate School of Pharmaceutical Sciences and Medicine, 6-3 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-8578, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Hirose T, Mori N, Totsune K, Morimoto R, Maejima T, Kawamura T, Metoki H, Asayama K, Kikuya M, Ohkubo T, Kohzuki M, Takahashi K, Imai Y. Increased expression of (pro)renin receptor in the remnant kidneys of 5/6 nephrectomized rats. ACTA ACUST UNITED AC 2010; 159:93-9. [PMID: 19896985 DOI: 10.1016/j.regpep.2009.11.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 09/27/2009] [Accepted: 11/01/2009] [Indexed: 12/26/2022]
Abstract
Recent studies have revealed that (pro)renin receptor ((P)RR), a newly identified member of the renin-angiotensin system, is associated with renal organ damage. However, there is little information regarding the regulation of (P)RR expression in various pathophysiological conditions. We therefore examined the expression of (P)RR in the remnant kidneys of rats with renal mass ablation due to 5/6 nephrectomy by quantitative RT-PCR, Western blot analysis and immunohistochemistry. Expression levels of (P)RR mRNA were significantly increased in the remnant kidneys at day 56 after nephrectomy, when compared with sham operation (about 1.6-fold, P=0.001). Western blot analysis showed that expression levels of (P)RR protein were greatly increased in the remnant kidneys at day 56, compared with sham operation (about 7.9-fold, P=0.02). The renal tubular cells were immunostained with anti-(P)RR antibody in both 5/6 nephrectomized rats and sham operated rats. The glomeruli were sporadically immunostained in 5/6 nephrectomized rats, but not in sham operated rats. These findings indicate that the intra-renal (P)RR expression is increased in the remnant kidneys of 5/6 nephrectomized rats, and suggest that (P)RR may contribute to the renal injury.
Collapse
Affiliation(s)
- Takuo Hirose
- Department of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Medicine, 6-3 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-8578, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hirose T, Mori N, Totsune K, Morimoto R, Maejima T, Kawamura T, Metoki H, Asayama K, Kikuya M, Ohkubo T, Kohzuki M, Takahashi K, Imai Y. Gene expression of (pro)renin receptor is upregulated in hearts and kidneys of rats with congestive heart failure. Peptides 2009; 30:2316-22. [PMID: 19765626 DOI: 10.1016/j.peptides.2009.09.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Revised: 09/10/2009] [Accepted: 09/10/2009] [Indexed: 12/23/2022]
Abstract
Recent studies have revealed that (pro)renin receptor ((P)RR), a newly identified member of the renin-angiotensin system, was associated with organ damage in the kidney. However, there has been little information for (P)RR in hearts. To investigate the regulation of (P)RR in heart failure, we examined the expression of (P)RR in hearts and kidneys of rats with congestive heart failure (CHF) due to coronary ligation by quantitative RT-PCR and immunohistochemistry. Significantly increased levels of (P)RR mRNA were found in the atrium, right ventricle, non-infarcted part of left ventricle, infarcted part of left ventricle and kidney of CHF rats, when compared with sham operated rats (about 1.6-fold, 1.4-fold, 1.6-fold, 1.7-fold and 1.5-fold, respectively). Expression levels of mRNAs encoding renin and angiotensinogen in these heart and kidney tissues were also increased in the CHF rats. Immunohistochemistry showed positive (P)RR immunostaining in the myocardium, the renal tubular cells, and vascular smooth muscle and endothelial cells in the heart and the kidney. The renal tubular cells were more intensely immunostained in CHF rats than in sham operated rats. These findings suggest that the expression of (P)RR is increased in the hearts and kidneys of rats with heart failure, and that (P)RR may contribute to heart failure.
Collapse
Affiliation(s)
- Takuo Hirose
- Department of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Medicine, Sendai 980-8578, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Evans JJ. Toward revealing the roles for intermedin in the community of vasoactive peptides. Hypertens Res 2009; 32:824-5. [DOI: 10.1038/hr.2009.127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
32
|
Intermedin is upregulated and has protective roles in a mouse ischemia/reperfusion model. Hypertens Res 2009; 32:861-8. [DOI: 10.1038/hr.2009.120] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
33
|
Hirose T, Takahashi K, Mori N, Nakayama T, Kikuya M, Ohkubo T, Kohzuki M, Totsune K, Imai Y. Increased expression of urotensin II, urotensin II-related peptide and urotensin II receptor mRNAs in the cardiovascular organs of hypertensive rats: comparison with endothelin-1. Peptides 2009; 30:1124-9. [PMID: 19463745 DOI: 10.1016/j.peptides.2009.02.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 02/09/2009] [Accepted: 02/11/2009] [Indexed: 02/07/2023]
Abstract
Urotensin II (UII) and urotensin II-related peptide (URP) are novel vasoactive peptides that share urotensin II receptor (UT). We have recently reported that expressions of URP and UT were up-regulated in kidneys of rats with renal failure or hypertension. To clarify possible changes of the UII system expression in cardiovascular organs with hypertension, we examined the gene expression of UII, URP and UT in hearts and aortae of hypertensive rats. Furthermore, the expression was compared with that of endothelin-1 (ET-1). Quantitative reverse transcription polymerase chain reaction analysis showed that expression levels of UII mRNA and UT mRNA were significantly elevated in the atrium of 11-12-week-old spontaneously hypertensive rats (SHR) compared with age-matched Wistar-Kyoto rats (WKY). Moreover, UT mRNA expression was elevated in the ventricle of 11-12-week-old SHR. In the aorta, expression levels of URP mRNA and UT mRNA were significantly elevated in 11-12-week-old SHR compared with age-matched WKY, similarly to those in the kidney. In contrast, expression levels of ET-1 were significantly decreased in both the heart and the kidney of 11-12-week-old SHR compared with age-matched WKY. Immunohistochemistry showed that URP and UT were immunostained in cardiomyocytes, with weaker immunostaining in vascular endothelial and smooth muscle cells, in both SHR and WKY. These findings indicate that the gene expression of the UII system components (UII, URP and UT) and ET-1 is differently regulated in hypertension, and that the UII system in the heart and aortae may have certain pathophysiological roles in hypertension.
Collapse
Affiliation(s)
- Takuo Hirose
- Department of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|