1
|
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
|
2
|
Wang Y, Wang Z, Peng Z, Feng L, Tian W, Zhang S, Cao L, Li J, Yang L, Xu Y, Gao Y, Liu J, Yan J, Ma X, Sun W, Guo L, Li X, Shen Y, Qi Z. Cocaine and amphetamine-regulated transcript improves myocardial ischemia-reperfusion injury through PI3K/AKT signalling pathway. Clin Exp Pharmacol Physiol 2024; 51:e13904. [PMID: 38923060 DOI: 10.1111/1440-1681.13904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/13/2024] [Accepted: 05/19/2024] [Indexed: 06/28/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a common clinic scenario that occurs in the context of reperfusion therapy for acute myocardial infarction. It has been shown that cocaine and amphetamine-regulated transcript (CART) can ameliorate cerebral ischemia-reperfusion (I/R) injury, but the effect of CART on MIRI has not been studied yet. Here, we revealed that CART protected the heart during I/R process by inhibiting apoptosis and excessive autophagy, indicating that CART would be a potential drug candidate for the treatment of MIRI. Further analysis showed that CART upregulated the activation of phospho-AKT, leading to downregulation of lactate dehydrogenase (LDH) release, apoptosis, oxidative stress and excessive autophagy after I/R, which was inhibited by PI3K inhibitor, LY294002. Collectively, CART attenuated MIRI through inhibition of cardiomyocytes apoptosis and excessive autophagy, and the protective effect was dependent on PI3K/AKT signalling pathway.
Collapse
Affiliation(s)
- Yachen Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Ziwei Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- NanKai University Eye Institute, Tianjin, China
| | - Zeyan Peng
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Lifeng Feng
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Wencong Tian
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, China
| | - Shengzheng Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Lei Cao
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, China
| | - Jing Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, China
| | - Yang Xu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, China
| | - Jie Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Jie Yan
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
| | - Xiaodong Ma
- Fifth People's Hospital of Dongying, Shandong, China
| | - Wangchun Sun
- Fifth People's Hospital of Dongying, Shandong, China
| | - Lihong Guo
- Shengli Oilfield Central Hospital Gastrointestinal Disease Research Institute, Shandong, China
| | - Xuan Li
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Yanna Shen
- School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, China
- NanKai University Eye Institute, Tianjin, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin, China
- Shengli Oilfield Central Hospital Gastrointestinal Disease Research Institute, Shandong, China
- Xinjiang Production and Construction Corps Hospital, Xinjiang, China
| |
Collapse
|
3
|
Wang B, Huang Y, Cai Q, Du Z, Li X. Biomaterials for diabetic bone repair: Influencing mechanisms, multi-aspect progress and future prospects. COMPOSITES PART B: ENGINEERING 2024; 274:111282. [DOI: 10.1016/j.compositesb.2024.111282] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
|
4
|
Zhang YR, Liu SM, Chen Y, Zhang LS, Ji DR, Zhao J, Yu YR, Jia MZ, Tang CS, Huang W, Zhou YB, Chai SB, Qi YF. Intermedin alleviates diabetic vascular calcification by inhibiting GLUT1 through activation of the cAMP/PKA signaling pathway. Atherosclerosis 2023; 385:117342. [PMID: 37879153 DOI: 10.1016/j.atherosclerosis.2023.117342] [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: 09/02/2022] [Revised: 09/21/2023] [Accepted: 10/10/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND AND AIMS Vascular calcification (VC) is regarded as an independent risk factor for cardiovascular events in type 2 diabetic patients. Glucose transporter 1 (GLUT1) involves VC. Intermedin/Adrenomedullin-2 (IMD/ADM2) is a cardiovascular protective peptide that can inhibit multiple disease-associated VC. However, the role and mechanism of IMD in diabetic VC remain unclear. Here, we investigated whether IMD inhibits diabetic VC by inhibiting GLUT1. METHODS AND RESULTS It was found that plasma IMD concentration was significantly decreased in type 2 diabetic patients and in fructose-induced diabetic rats compared with that in controls. Plasma IMD content was inversely correlated with fasting blood glucose level and VC severity. IMD alleviated VC in fructose-induced diabetic rats. Deficiency of Adm2 aggravated and Adm2 overexpression attenuated VC in high-fat diet-induced diabetic mice. In vitro, IMD mitigated high glucose-induced calcification of vascular smooth muscle cells (VSMCs). Mechanistically, IMD reduced advanced glycation end products (AGEs) content and the level of receptor for AGEs (RAGE). IMD decreased glucose transporter 1 (GLUT1) levels. The inhibitory effect of IMD on RAGE protein level was blocked by GLUT1 knockdown. GLUT1 knockdown abolished the effect of IMD on alleviating VSMC calcification. IMD receptor antagonist IMD17-47 and cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) inhibitor H89 abolished the inhibitory effects of IMD on GLUT1 and VSMC calcification. CONCLUSIONS These findings revealed that IMD exerted its anti-calcification effect by inhibiting GLUT1, providing a novel therapeutic target for diabetic VC.
Collapse
Affiliation(s)
- Ya-Rong Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Shi-Meng Liu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Yao Chen
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Lin-Shuang Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Deng-Ren Ji
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Jie Zhao
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Yan-Rong Yu
- Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Mo-Zhi Jia
- Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Chao-Shu Tang
- StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China
| | - Wei Huang
- StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China
| | - Ye-Bo Zhou
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China.
| | - San-Bao Chai
- Department of Endocrinology and Metabolism, Peking University International Hospital, Beijing, 102206, China.
| | - Yong-Fen Qi
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China.
| |
Collapse
|
5
|
Niu H, Hao Y, Pang Y, Shen Y, Li J, Xu X. LncRNA-adm2 targets adm2 via cid-miR-n3 and negatively regulates the inflammatory response in grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2023; 138:108800. [PMID: 37187213 DOI: 10.1016/j.fsi.2023.108800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/23/2023] [Accepted: 05/05/2023] [Indexed: 05/17/2023]
Abstract
Long non-coding RNAs (lncRNAs), which impact gene expression following pathogen infections, have garnered significant attention in recent years. Recent discoveries have revealed that lncRNAs play a crucial role in fish immune responses to pathogen infections. We investigated the influence of lncRNA-adm2 on the antibacterial immune response generated by Aeromonas hydrophila in grass carp (Ctenopharyngodon idella) through the adsorption of cid-miR-n3. Furthermore, we found that cid-miR-n3 interacts with lncRNA-adm2 and targets the 3' UTR of adm2. The upregulation of lncRNA-adm2 expression led to the suppression of pro-inflammatory cytokines (il-1β and il-6) in CIK cells, while anti-inflammatory cytokines (il-10) increased. Our research provides evidence that lncRNAs are involved in the antibacterial immune response of fish, expanding our understanding of the function of lncRNAs in teleosts.
Collapse
Affiliation(s)
- Huiqin Niu
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Yinghu Hao
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Yifan Pang
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Yubang Shen
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China
| | - Jiale Li
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China.
| | - Xiaoyan Xu
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
6
|
Liao H, Wang Z, Zhang X, Li X, Chen X. Intermedin induces autophagy and attenuates hypoxia-induced injury in cardiomyocytes by regulation of MALAT1/ULK1. Peptides 2023; 160:170917. [PMID: 36442698 DOI: 10.1016/j.peptides.2022.170917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 10/26/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
Myocardial infarction is a predominant cause of cardiovascular diseases with high incidence and death rate worldwide. Although growing evidence has suggested that IMD has significant protective influences on the cardiovascular system, the molecular regulatory mechanism of IMD in hypoxia-induced injury caused by myocardial infarction is urgent to be elucidated. In the present study, we found hypoxia led to a noteworthy enhancement in IMD expression and IMD alleviated hypoxia-induced myocardial injury of NRCMs. Furthermore, IMD was proved to inhibit hypoxia-induced injury by regulating MALAT1. Our findings suggested MALAT1 positively regulated the mRNA and protein expression level of ULK1 and hypoxia induced autophagy of NRCMs. MALAT1 stimulated autophagy to block hypoxia-induced cell injury in NRCMs via upregulation of ULK1 expression. Autophagy suppression abolished the protective capability of IMD overexpression against hypoxia-induced myocardial injury in NRCMs. In a word, our study shed light on the central mechanism of IMD in preventing hypoxia-induced injury caused by myocardial infarction. We confirmed IMD induced autophagy and attenuated hypoxia-induced injury in cardiomyocytes via MALAT1/ULK1, which may contribute to designing effective therapeutic approaches of myocardial infarction.
Collapse
Affiliation(s)
- Hang Liao
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610015, China
| | - Ziqiong Wang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610015, China
| | - Xin Zhang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610015, China
| | - Xinran Li
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610015, China
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610015, China.
| |
Collapse
|
7
|
Liu F, Duan J, Teng X, Peng D. THE INCREASED PLASMA LEVELS OF INTERMEDIN IN PATIENTS WITH TYPE 2 DIABETES MELLITUS. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2022; 18:271-277. [PMID: 36699172 PMCID: PMC9867815 DOI: 10.4183/aeb.2022.271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Context Intermedin (IMD) is the member of calcitonin gene-related peptide family, and tightly associated with type 2 diabetes mellitus (T2DM). The change of plasma IMD levels in T2DM is still unknown. Objective We aimed to investigate the plasma levels of IMD in patients with T2DM. Design Fortyone patients with T2DM who were hospitalized in the endocrinology department of Civil Aviation General Hospital from January 2012 to June 2015 were enrolled, and 44 volunteers were selected as the control group. Subjects and Methods Plasma level of IMD was detected by ELISA. Diagnostic value of IMD was analyzed by area under the receiver operating characteristic (ROC) curve (AUC). Results The plasma level of IMD in T2DM group was higher than that in the healthy control group, whereas smoking or cardiovascular complications did no influence the IMD levels. IMD levels were correlated with BMI, DBP, triglyceride, uric acid, urea nitrogen, fasting and 2 hours postprandial blood glucose, and HbA1C. The greatest value of AUC for IMD was only 58.73%. Conclusions Although plasma levels of IMD were increased in patients with T2DM, the very low diagnostic value of IMD for T2DM might not be used for the disease diagnosis.
Collapse
Affiliation(s)
- F. Liu
- Civil Aviation General Hospital, Department of Endocrinology, Beijing, China
| | - J.T. Duan
- Civil Aviation General Hospital, Department of Endocrinology, Beijing, China
| | - X. Teng
- Hebei Medical University, Department of Physiology, Shijiazhuang, China
- Hebei Collaborative Innovation Center for Cardio–Cerebrovascular Disease, Shijiazhuang, China
| | - D.Q. Peng
- Civil Aviation General Hospital, Department of Endocrinology, Beijing, China
| |
Collapse
|
8
|
Liu Z, Liu Q, Guo H, Liang J, Zhang Y. Overview of Physical and Pharmacological Therapy in Enhancing Bone Regeneration Formation During Distraction Osteogenesis. Front Cell Dev Biol 2022; 10:837430. [PMID: 35573673 PMCID: PMC9096102 DOI: 10.3389/fcell.2022.837430] [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: 12/16/2021] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Distraction osteogenesis (DO) is a kind of bone regeneration technology. The principle is to incise the cortical bone and apply continuous and stable distraction force to the fractured end of the cortical bone, thereby promoting the proliferation of osteoblastic cells in the tension microenvironment and stimulating new bone formation. However, the long consolidation course of DO presumably lead to several complications such as infection, fracture, scar formation, delayed union and malunion. Therefore, it is of clinical significance to reduce the long treatment duration. The current treatment strategy to promote osteogenesis in DO includes gene, growth factor, stem-cell, physical and pharmacological therapies. Among these methods, pharmacological and physical therapies are considered as safe, economical, convenience and effective. Recently, several physical and pharmacological therapies have been demonstrated with a decent ability to enhance bone regeneration during DO. In this review, we have comprehensively summarized the latest evidence for physical (Photonic, Waves, Gas, Mechanical, Electrical and Electromagnetic stimulation) and pharmacological (Bisphosphonates, Hormone, Metal compounds, Biologics, Chinese medicine, etc) therapies in DO. These evidences will bring novel and significant information for the bone healing during DO in the future.
Collapse
Affiliation(s)
- Ze Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qi Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hongbin Guo
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jieyu Liang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Zhang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Zhang LS, Zhang JS, Hou YL, Lu WW, Ni XQ, Lin F, Liu XY, Wang XJ, Yu YR, Jia MZ, Tang CS, Han L, Chai SB, Qi YF. Intermedin 1-53 Inhibits NLRP3 Inflammasome Activation by Targeting IRE1α in Cardiac Fibrosis. Inflammation 2022; 45:1568-1584. [PMID: 35175495 DOI: 10.1007/s10753-022-01642-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 11/24/2022]
Abstract
Intermedin (IMD), a paracrine/autocrine peptide, protects against cardiac fibrosis. However, the underlying mechanism remains poorly understood. Previous study reports that activation of nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome contributes to cardiac fibrosis. In this study, we aimed to investigate whether IMD mitigated cardiac fibrosis by inhibiting NLRP3. Cardiac fibrosis was induced by angiotensin II (Ang II) infusion for 2 weeks in rats. Western blot, real-time PCR, histological staining, immunofluorescence assay, RNA sequencing, echocardiography, and hemodynamics were used to detect the role and the mechanism of IMD in cardiac fibrosis. Ang II infusion resulted in rat cardiac fibrosis, shown as over-deposition of myocardial interstitial collagen and cardiac dysfunction. Importantly, NLRP3 activation and endoplasmic reticulum stress (ERS) were found in Ang II-treated rat myocardium. Ang II infusion decreased the expression of IMD and increased the expression of the receptor system of IMD in the fibrotic rat myocardium. IMD treatment attenuated the cardiac fibrosis and improved cardiac function. In addition, IMD inhibited the upregulation of NLRP3 markers and ERS markers induced by Ang II. In vitro, IMD knockdown by small interfering RNA significantly promoted the Ang II-induced cardiac fibroblast and NLRP3 activation. Moreover, silencing of inositol requiring enzyme 1 α (IRE1α) blocked the effects of IMD inhibiting fibroblast and NLRP3 activation. Pre-incubation with PKA pathway inhibitor H89 blocked the effects of IMD on the anti-ERS, anti-NLRP3, and anti-fibrotic response. In conclusion, IMD alleviated cardiac fibrosis by inhibiting NLRP3 inflammasome activation through suppressing IRE1α via the cAMP/PKA pathway.
Collapse
Affiliation(s)
- Lin-Shuang Zhang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China.,Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, HaidianDistrict, No. 38 Xueyuan Road, Beijing, 100083, China.,School of Nursing, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jin-Sheng Zhang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China.,Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, HaidianDistrict, No. 38 Xueyuan Road, Beijing, 100083, China
| | - Yue-Long Hou
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Wei-Wei Lu
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China.,Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, HaidianDistrict, No. 38 Xueyuan Road, Beijing, 100083, China
| | - Xian-Qiang Ni
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China.,Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, HaidianDistrict, No. 38 Xueyuan Road, Beijing, 100083, China
| | - Fan Lin
- Department of Respiratory Disease, Peking University Third Hospital, Beijing, China
| | - Xiu-Ying Liu
- Key Laboratory of Genetic Network Biology, Collaborative Innovation Center of Genetics and Development, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xiu-Jie Wang
- Key Laboratory of Genetic Network Biology, Collaborative Innovation Center of Genetics and Development, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yan-Rong Yu
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, HaidianDistrict, No. 38 Xueyuan Road, Beijing, 100083, China
| | - Mo-Zhi Jia
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, HaidianDistrict, No. 38 Xueyuan Road, Beijing, 100083, China
| | - Chao-Shu Tang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Ling Han
- Department of Cardiology, Fu Xing Hospital, Capital Medical University, A20 Fuxingmenwai Street, Xicheng District, Beijing, 100038, China.
| | - San-Bao Chai
- Department of Endocrinology, Peking University International Hospital, Life Park Road No. 1, Zhongguancun Life Science Park, Changping District, Beijing, 102206, China.
| | - Yong-Fen Qi
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China. .,Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, HaidianDistrict, No. 38 Xueyuan Road, Beijing, 100083, China.
| |
Collapse
|
10
|
Chen Y, Zhou Y, Lin J, Zhang S. Challenges to Improve Bone Healing Under Diabetic Conditions. Front Endocrinol (Lausanne) 2022; 13:861878. [PMID: 35418946 PMCID: PMC8996179 DOI: 10.3389/fendo.2022.861878] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/02/2022] [Indexed: 12/17/2022] Open
Abstract
Diabetes mellitus (DM) can affect bone metabolism and the bone microenvironment, resulting in impaired bone healing. The mechanisms include oxidative stress, inflammation, the production of advanced glycation end products (AGEs), etc. Improving bone healing in diabetic patients has important clinical significance in promoting fracture healing and improving bone integration. In this paper, we reviewed the methods of improving bone healing under diabetic conditions, including drug therapy, biochemical cues, hyperbaric oxygen, ultrasound, laser and pulsed electromagnetic fields, although most studies are in preclinical stages. Meanwhile, we also pointed out some shortcomings and challenges, hoping to provide a potential therapeutic strategy for accelerating bone healing in patients with diabetes.
Collapse
Affiliation(s)
- Yiling Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie Lin
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie Lin, ; Shiwen Zhang,
| | - Shiwen Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie Lin, ; Shiwen Zhang,
| |
Collapse
|
11
|
Study on Protection of Human Umbilical Vein Endothelial Cells from Amiodarone-Induced Damage by Intermedin through Activation of Wnt/ β-Catenin Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8889408. [PMID: 34434487 PMCID: PMC8382522 DOI: 10.1155/2021/8889408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 06/07/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
Amiodarone (AM) is one of the most effective antiarrhythmic drugs and normally administrated by intravenous infusion which is liable to cause serious phlebitis. The therapeutic drugs for preventing this complication are limited. Intermedin (IMD), a member of calcitonin family, has a broad spectrum of biological effects including anti-inflammatory effects, antioxidant activities, and antiapoptosis. But now, the protective effects of IMD against amiodarone-induced phlebitis and the underlying molecular mechanism are not well understood. In this study, the aim was to investigate the protective efficiency and potential mechanisms of IMD in amiodarone-induced phlebitis. The results of this study revealed that treatment with IMD obviously attenuated apoptosis and exfoliation of vascular endothelial cells and infiltration of inflammatory cells in the rabbit model of phlebitis induced by intravenous infusion of amiodarone compared with control. Further tests in vitro demonstrated that IMD lessened amiodarone-induced endothelial cell apoptosis, improved amiodarone-induced oxidative stress injury, reduced inflammatory reaction, and activated the Wnt/β-catenin signal pathway which was inhibited by amiodarone. And these effects could be reversed by Wnt/β-catenin inhibitor IWR-1-endo, and si-RNA knocked down the gene of Wnt pathway. These results suggested that IMD exerted the protective effects against amiodarone-induced endothelial injury via activating the Wnt/β-catenin pathway. Thus, IMD could be used as a potential agent for the treatment of phlebitis.
Collapse
|
12
|
Wang Y, Tian J, Mi Y, Ren X, Lian S, Kang J, Wang J, Zang H, Wu Z, Yang J, Qiao X, Zhou X, Wang G, Zhou Y, Li R. Experimental study on renoprotective effect of intermedin on diabetic nephropathy. Mol Cell Endocrinol 2021; 528:111224. [PMID: 33675865 DOI: 10.1016/j.mce.2021.111224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 01/17/2021] [Accepted: 01/23/2021] [Indexed: 12/16/2022]
Abstract
Intermedin(IMD) is a novel member of the calcitonin/calcitonin gene-related peptide (CT/CGRP) family that has anti-inflammatory, antioxidant and anti-apoptosis properties. This study aimed to evaluate the renoprotective effects of IMD on podocyte apoptotic loss and slit diaphragm protein deficiency the kidneys of rats with in streptozotocin (STZ) induced diabetes in high glucose-exposed podocytes. Our results showed that IMD significantly attenuated proteinuria, and alleviated the abnormal alterations in glomerular ultrastructure in vivo. IMD also improved the induction of slit diaphragm proteins, and restored the decreased Bcl-2 expression and suppressed Bax and caspase-3 induction in the diabetic glomeruli. In addition, IMD attenuated podocyte apoptosis and filamentous actin (F-actin) rearrangement in high glucose-exposed podocytes. Exposure to high glucose elevated the unfolded protein response (UPR) to endoplasmic reticulum (ER) stress in renal podocytes, and IMD treatment blocked such ER stress responses pertinent to podocyte apoptosis and reduced synthesis of slit diaphragm proteins in vivo and in vitro. These observations demonstrate that targeting ER stress is an underlying mechanism of IMD-mediated amelioration of diabetes-associated podocyte injury and dysfunction.
Collapse
Affiliation(s)
- Yanhong Wang
- Department of Nephrology, Postdoctoral Workstation of Shanxi Provincial People's Hospital, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China; Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jihua Tian
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Yang Mi
- Department of Urology, The Affiliated Bethune Hospital of Shanxi Medical University, Shanxi Bethune Hospital (Shanxi Academy of Medical Sciences), Taiyuan, Shanxi, 030032, China
| | - Xiaojun Ren
- Department of Nephrology, The Affiliated Bethune Hospital of Shanxi Medical University, Shanxi Bethune Hospital (Shanxi Academy of Medical Sciences), Taiyuan, Shanxi, 030032, China
| | - Shizhong Lian
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jing Kang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Juanjuan Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Haojing Zang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Zhijing Wu
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jia Yang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xi Qiao
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xiaoshuang Zhou
- Department of Nephrology, Postdoctoral Workstation of Shanxi Provincial People's Hospital, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China
| | - Guiqin Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Yun Zhou
- Department of Nephrology, Postdoctoral Workstation of Shanxi Provincial People's Hospital, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China.
| | - Rongshan Li
- Department of Nephrology, Postdoctoral Workstation of Shanxi Provincial People's Hospital, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, 030012, China.
| |
Collapse
|
13
|
Wang F, Kong L, Wang W, Shi L, Wang M, Chai Y, Xu J, Kang Q. Adrenomedullin 2 improves bone regeneration in type 1 diabetic rats by restoring imbalanced macrophage polarization and impaired osteogenesis. Stem Cell Res Ther 2021; 12:288. [PMID: 33985585 PMCID: PMC8117361 DOI: 10.1186/s13287-021-02368-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/03/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Both advanced glycation end products (AGEs) and AGE-mediated M1 macrophage polarization contribute to bone marrow mesenchymal stem cell (BMSC) dysfunction, leading to impaired bone regeneration in type 1 diabetes mellitus (T1DM). Adrenomedullin 2 (ADM2), an endogenous bioactive peptide belonging to the calcitonin gene-related peptide family, exhibits various biological activities associated with the inhibition of inflammation and reduction of insulin resistance. However, the effects and underlying mechanisms of ADM2 in AGE-induced macrophage M1 polarization, BMSC dysfunction, and impaired bone regeneration remain poorly understood. METHODS The polarization of bone marrow-derived macrophages was verified using flow cytometry analysis. Alkaline phosphatase (ALP) staining, ALP activity detection, and alizarin red staining were performed to assess the osteogenesis of BMSCs. Quantitative real-time polymerase chain reaction, enzyme-linked immunosorbent assay, western blotting, and immunofluorescence staining were used to assess polarization markers, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, and osteogenic markers. In vivo, a distraction osteogenesis (DO) rat model with T1DM was established, and tibia samples were collected at different time points for radiological, biomechanical, and histological analyses, to verify the effects of ADM2 on bone regeneration and M2 polarization under diabetic conditions. RESULTS ADM2 treatment reversed AGE-induced M1 macrophage polarization towards the M2 phenotype, which was partially achieved by the peroxisome proliferator-activated receptor γ (PPARγ)-mediated inhibition of NF-κB signaling. The PPARγ inhibitor GW9662 significantly attenuated the effects of ADM2. Besides, ADM2 treatment improved the AGE-impaired osteogenic potential of BMSCs in vitro. Furthermore, ADM2 accelerated bone regeneration, as revealed by improved radiological and histological manifestations and biomechanical parameters, accompanied by improved M2 macrophage polarization in diabetic DO rats, and these effects were partially blocked by GW9662 administration. CONCLUSIONS These results indicate that ADM2 enhances diabetic bone regeneration during DO, by attenuating AGE-induced imbalances in macrophage polarization, partly through PPARγ/NF-κB signaling, and improving AGE-impaired osteogenic differentiation of BMSCs simultaneously. These findings reveal that ADM2 may serve as a potential bioactive factor for promoting bone regeneration under diabetic conditions, and imply that management of inflammation and osteogenesis, in parallel, may present a promising therapeutic strategy for diabetic patients during DO treatment.
Collapse
Affiliation(s)
- Feng Wang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Lingchi Kong
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Wenbo Wang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Li Shi
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Mengwei Wang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Yimin Chai
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Jia Xu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| | - Qinglin Kang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| |
Collapse
|
14
|
Su RY, Geng XY, Yang Y, Yin HS. Nesfatin-1 inhibits myocardial ischaemia/reperfusion injury through activating Akt/ERK pathway-dependent attenuation of endoplasmic reticulum stress. J Cell Mol Med 2021; 25:5050-5059. [PMID: 33939297 PMCID: PMC8178279 DOI: 10.1111/jcmm.16481] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/11/2021] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
Nesfatin‐1 (encoded by NUCB2) is a cardiac peptide possessing protective activities against myocardial ischaemia/reperfusion (MI/R) injury. However, the regulation of NUCB2/nesfatin‐1 and the molecular mechanisms underlying its roles in MI/R injury are not clear. Here, by investigating a mouse MI/R injury model developed with transient myocardial ischaemia followed by reperfusion, we found that the levels of NUCB2 transcript and nesfatin‐1 amount in the heart were both decreased, suggesting a transcriptional repression of NUCB2/nesfatin‐1 in response to MI/R injury. Moreover, cardiac nesfatin‐1 restoration reduced infarct size, troponin T (cTnT) level and myocardial apoptosis, supporting its cardioprotection against MI/R injury in vivo. Mechanistically, the Akt/ERK pathway was activated, and in contrast, endoplasmic reticulum (ER) stress was attenuated by nesfatin‐1 following MI/R injury. In an in vitro system, similar results were obtained in nesfatin‐1‐treated H9c2 cardiomyocytes with hypoxia/reoxygenation (H/R) injury. More importantly, the treatment of wortmannin, an inhibitor of Akt/ERK pathway, abrogated nesfatin‐1 effects on attenuating ER stress and H/R injury in H9c2 cells. Furthermore, nesfatin‐1‐mediated protection against H/R injury also vanished in the presence of tunicamycin (TM), an ER stress inducer. Lastly, Akt/ERK inhibition reversed nesfatin‐1 effects on mouse ER stress and MI/R injury in vivo. Taken together, these findings demonstrate that NUCB2/nesfatin‐1 inhibits MI/R injury through attenuating ER stress, which relies on Akt/ERK pathway activation. Hence, our study provides a molecular basis for understanding how NUCB2/nesfatin‐1 reduces MI/R injury.
Collapse
Affiliation(s)
- Rui-Ying Su
- Department of Cardiac Function Inspection, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiao-Yong Geng
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yang Yang
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hong-Shan Yin
- Department of Cardiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
15
|
Intermedin 1-53 attenuates atherosclerotic plaque vulnerability by inhibiting CHOP-mediated apoptosis and inflammasome in macrophages. Cell Death Dis 2021; 12:436. [PMID: 33934111 PMCID: PMC8088440 DOI: 10.1038/s41419-021-03712-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/19/2022]
Abstract
Atherosclerotic plaque vulnerability and rupture increase the risk of acute coronary syndromes. Advanced lesion macrophage apoptosis plays important role in the rupture of atherosclerotic plaque, and endoplasmic reticulum stress (ERS) has been proved to be a key mechanism of macrophage apoptosis. Intermedin (IMD) is a regulator of ERS. Here, we investigated whether IMD enhances atherosclerotic plaque stability by inhibiting ERS-CHOP-mediated apoptosis and subsequent inflammasome in macrophages. We studied the effects of IMD on features of plaque vulnerability in hyperlipemia apolipoprotein E-deficient (ApoE−/−) mice. Six-week IMD1-53 infusion significantly reduced atherosclerotic lesion size. Of note, IMD1-53 lowered lesion macrophage content and necrotic core size and increased fibrous cap thickness and vascular smooth muscle cells (VSMCs) content thus reducing overall plaque vulnerability. Immunohistochemical analysis indicated that IMD1-53 administration prevented ERS activation in aortic lesions of ApoE−/− mice, which was further confirmed in oxidized low-density lipoproteins (ox-LDL) induced macrophages. Similar to IMD, taurine (Tau), a non-selective ERS inhibitor significantly reduced atherosclerotic lesion size and plaque vulnerability. Moreover, C/EBP-homologous protein (CHOP), a pro-apoptosis transcription factor involved in ERS, was significantly increased in advanced lesion macrophages, and deficiency of CHOP stabilized atherosclerotic plaques in AopE−/− mice. IMD1-53 decreased CHOP level and apoptosis in vivo and in macrophages treated with ox-LDL. In addition, IMD1-53 infusion ameliorated NLRP3 inflammasome and subsequent proinflammatory cytokines in vivo and in vitro. IMD may attenuate the progression of atherosclerotic lesions and plaque vulnerability by inhibiting ERS-CHOP-mediated macrophage apoptosis, and subsequent NLRP3 triggered inflammation. The inhibitory effect of IMD on ERS-induced macrophages apoptosis was probably mediated by blocking CHOP activation.
Collapse
|
16
|
Wang F, Wang W, Kong L, Shi L, Wang M, Chai Y, Xu J, Kang Q. Accelerated Bone Regeneration by Adrenomedullin 2 Through Improving the Coupling of Osteogenesis and Angiogenesis via β-Catenin Signaling. Front Cell Dev Biol 2021; 9:649277. [PMID: 33937244 PMCID: PMC8079771 DOI: 10.3389/fcell.2021.649277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/25/2021] [Indexed: 11/13/2022] Open
Abstract
Both osteogenic differentiation and the pro-angiogenic potential of bone marrow mesenchymal stem cells (BMSCs) contribute to bone regeneration during distraction osteogenesis (DO). Adrenomedullin 2 (ADM2), an endogenous bioactive peptide belonging to the calcitonin gene-related peptide family, exhibits various biological activities associated with the inhibition of inflammation and the attenuation of ischemic-hypoxic injury. However, the effects and underlying mechanisms of ADM2 in osteogenic differentiation and the pro-angiogenic potential of BMSCs, along with bone regeneration, remain poorly understood. In the present study, we found that osteogenic induction enhanced the pro-angiogenic potential of BMSCs, and ADM2 treatment further improved the osteogenic differentiation and pro-angiogenic potential of BMSCs. Moreover, the accumulation and activation of β-catenin, which is mediated by the inhibition of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and the activation of protein kinase B (AKT), have been shown to contribute to the effects of ADM2 on BMSCs. In vivo, ADM2 accelerated vessel expansion and bone regeneration, as revealed by improved radiological and histological manifestations and the biomechanical parameters in a rat DO model. Based on the present results, we concluded that ADM2 accelerates bone regeneration during DO by enhancing the osteogenic differentiation and pro-angiogenic potential of BMSCs, partly through the NF-κB/β-catenin and AKT/β-catenin pathways. Moreover, these findings imply that BMSC-mediated coupling of osteogenesis and angiogenesis may be a promising therapeutic strategy for DO patients.
Collapse
|
17
|
Sun J, Qian P, Kang Y, Dai HB, Wang FZ, Wang HY, Zhou H, Gao Q, Zhou YB. Adrenomedullin 2 attenuates LPS-induced inflammation in microglia cells by receptor-mediated cAMP-PKA pathway. Neuropeptides 2021; 85:102109. [PMID: 33253929 DOI: 10.1016/j.npep.2020.102109] [Citation(s) in RCA: 2] [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: 03/30/2020] [Revised: 11/15/2020] [Accepted: 11/15/2020] [Indexed: 12/18/2022]
Abstract
Inflammation plays a critical role in the development of neurodegenerative diseases. Adrenomedullin 2 (AM2), a member of the calcitonin gene-related peptide family, has been known to have anti-inflammatory effects. Here, we evaluated the anti-inflammatory effects of AM2 in LPS-activated microglia and BV2 cells. The endogenous mRNA and protein expressions of AM2, calcitonin receptor-like receptor (CLR), receptor activity-modifying proteins (RAMPs) including RAMP1, RAMP2 and RAMP3 and the production of inflammatory mediators including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) were detected by RT-PCR and Western blot. Our results revealed that LPS (1 μg/mL) significantly stimulated CLR, RAMP1, RAMP2 and RAMP3 protein expressions in BV2 microglia cells, but AM2 had a significant decrease. However, the mRNA levels of AM2, CLR, and RAMP1/2/3 were all markedly increased. LPS also induced obvious increases in mRNA and protein levels of the inflammatory mediators (TNF-α, IL-1β, COX2 and iNOS). More importantly, AM2 (10 nM) administration effectively inhibited the mRNA and protein expressions of these mediators induced by LPS and increased the cAMP content in LPS-stimulated BV2 cells. Furthermore, the antagonism with AM2 receptor antagonist IMD17-47, adrenomedullin (AM) receptor antagonist by AM22-52 or the inhibition of protein kinase A (PKA) activation by P1195 effectively prevented the inhibitory role of AM2 in LPS-induced production of the above inflammatory mediators. In conclusion, AM2 inhibits LPS-induced inflammation in BV2 microglia cells that may be mainly through AM receptor-mediated cAMP-PKA pathway. Our results indicate AM2 plays an important protective role in microglia inflammation, suggesting therapeutic potential for AM2 in neuroinflammation diseases caused by activated microglia.
Collapse
Affiliation(s)
- Jing Sun
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Pei Qian
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Ying Kang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Hang-Bing Dai
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Fang-Zheng Wang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Hong-Yu Wang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Hong Zhou
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Qing Gao
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Ye-Bo Zhou
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
18
|
Zhang XY, Guo Z, Li TP, Sun T. Dietary capsaicin normalizes CGRP peptidergic DRG neurons in experimental diabetic peripheral neuropathy. Sci Rep 2021; 11:1704. [PMID: 33462325 PMCID: PMC7814129 DOI: 10.1038/s41598-021-81427-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 01/04/2021] [Indexed: 12/22/2022] Open
Abstract
Diabetic sensory neuropathy leads to impairment of peripheral sensory nerves and downregulation of calcitonin gene-related peptide (CGRP) in a functionally specific subset of peripheral sensory neurons mediating pain. Whether CGRP plays a neuroprotective role in peripheral sensory nerve is unclear. We evaluated alterations in noxious thermal sensation and downregulation of CGRP in the 8 weeks after induction of diabetes in rats. We supplemented capsaicin in the diet of the animals to upregulate CGRP and reversed the downregulation of the neuropeptide in the dorsal root ganglion (DRG) neurons dissociated from the diabetic animals, via gene transfection and exogenous CGRP, to test disease-preventing and disease-limiting effects of CGRP. Significant preservation of the nociceptive sensation, CGRP in spinal cord and DRG neurons, and number of CGRP-expressing neurons was found in the diabetic animals given capsaicin. Improvement in the survival of the neurons and the outgrowth of neurites was achieved in the neurons transfected by LV-CGRP or by exogenous CGRP, paralleling the correction of abnormalities of intracellular reactive oxygen species and mitochondrial transmembrane potentials. The results suggest that downregulation of CGRP impairs viability, regeneration and function of peripheral sensory neurons while capsaicin normalizes the CGRP peptidergic DRG neurons and function of the sensory nerves.
Collapse
Affiliation(s)
- Xiao-Yi Zhang
- Department of Anesthesiology, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Zheng Guo
- Department of Anesthesiology, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China. .,Department of Anesthesiology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China. .,Key Laboratory of Cellular Physiology (Shanxi Medical University), National Education Commission, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China.
| | - Tu-Ping Li
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Tao Sun
- Department of Anesthesiology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| |
Collapse
|
19
|
Xue Y, Fu W, Liu Y, Yu P, Sun M, Li X, Yu X, Sui D. Ginsenoside Rb2 alleviates myocardial ischemia/reperfusion injury in rats through SIRT1 activation. J Food Sci 2020; 85:4039-4049. [PMID: 33073372 DOI: 10.1111/1750-3841.15505] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/05/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022]
Abstract
The cardioprotective effects of ginsenoside Rb2 on oxidative stress, which is induced by hydrogen peroxide and myocardial ischemia/reperfusion (MI/R) injury, have been studied. The mechanisms were associated with the inhibition of cardiomyocyte apoptosis, a high concentration of antioxidant defense enzymes, and scavenging oxidative stress products. Because of the association with oxidative reaction and cardioprotection, sirtuin-1 (SIRT1) was selected as a promising target for investigating whether MI/R injury can be alleviated by ginsenoside Rb2 pretreatment through SIRT1 activation. The rats were exposed to ginsenoside Rb2 with or without SIRT1 inhibitor EX527 before ligation of coronary artery. Ginsenoside Rb2 reduced myocardial superoxide generation; downregulated gp91phox expression; and decreased the mRNA expression levels and activities of interleukin-1β, interleukin-6, and tumor necrosis factor-α. The results demonstrated that ginsenoside Rb2 significantly attenuated oxidative stress and inflammation induced by MI/R injury. In addition, ginsenoside Rb2 upregulated SIRT1 expression and downregulated Ac-p53 expression. However, EX527 blocked the protective effects, indicating that the pharmacological action of ginsenoside Rb2 involves SIRT1. Our results thus revealed that ginsenoside Rb2 alleviated MI/R injury in rats by inhibiting oxidative stress and inflammatory response through SIRT1 activation. PRACTICAL APPLICATION: Ginsenoside Rb2 has a protective effect on MI/R injury by activating SIRT1 expression, reducing myocardium inflammation, and alleviating oxidative stress. Thus, ginsenoside Rb2 is a promising novel agent for ameliorating MI/R injury in ischemic heart diseases and cardiac surgery.
Collapse
Affiliation(s)
- Yan Xue
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, PR China.,Department of Burn Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, PR China
| | - Wenwen Fu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, PR China
| | - Yanzhe Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, PR China
| | - Ping Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, PR China
| | - Mingyang Sun
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, PR China
| | - Xin Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, PR China
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, PR China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, PR China
| |
Collapse
|
20
|
Gong J, Zhou F, Wang SXX, Xu J, Xiao F. Caveolin-3 protects diabetic hearts from acute myocardial infarction/reperfusion injury through β2AR, cAMP/PKA, and BDNF/TrkB signaling pathways. Aging (Albany NY) 2020; 12:14300-14313. [PMID: 32692723 PMCID: PMC7425465 DOI: 10.18632/aging.103469] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 05/27/2020] [Indexed: 12/11/2022]
Abstract
Diabetes mellitus (DM) might increase the incidence and mortality of cardiac failure after acute myocardial infarction (AMI) in patients. We attempted to investigate whether Caveolin-3 showed beneficial effects in DM patient post-MI injury through the cAMP/PKA and BDNF/TrkB signaling pathways. The activity of ADRB2 and cAMP/PKA signaling were impaired in nondiabetic ischemia-reperfusion (I/R) group compared with the sham and DM groups and were more impaired in diabetic I/R group than in the I/R group. In H9C2 cells, high-glucose (HG) stimulation further enhanced H/R injury by promoting cell apoptosis, inhibiting cell viability, and suppressing TrkB and Akt signaling; in contrast, the ADRB2 agonist isoprenaline (ISO) significantly attenuated the above-described effects of HG stimulation. Caveolin-3 overexpression promoted the localization of ADRB2 on the membrane of the HG-stimulated H9C2 cells, subsequently inhibiting apoptosis and promoting cell viability. Under HG stimulation, Caveolin-3 overexpression enhanced the activity of the cAMP/PKA and BDNF/TrkB signaling pathways, whereas ADRB2 silencing reversed the effects of Caveolin-3 overexpression. In conclusion, ADRB2 agonist promoted the activity of the BDNF/TrkB and cAMP/PKA signaling pathways, mitigating the HG-aggravated H/R injuries in H9C2 cells. Caveolin-3 exerts a protective effect on diabetic hearts against I/R damage through the β2AR, cAMP/PKA, and BDNF/TrkB signaling pathways.
Collapse
Affiliation(s)
- Jiaji Gong
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Fan Zhou
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Simin Xie Xin Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Junmei Xu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Feng Xiao
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
21
|
Protective effect of intermediate doses of hydrogen sulfide against myocardial ischemia-reperfusion injury in obese type 2 diabetic rats. Life Sci 2020; 256:117855. [PMID: 32473245 DOI: 10.1016/j.lfs.2020.117855] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/17/2020] [Accepted: 05/25/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Subjects with type 2 diabetes (T2D) have lower circulating hydrogen sulfide (H2S) levels following myocardial ischemia and a higher risk of mortality. The aim of this study was to determine the dose-dependent favorable effects of sodium hydrosulfide (NaSH) on myocardial ischemia-reperfusion (IR) injury in rats with T2D. METHODS T2D was induced using a high-fat diet (HFD) and low-dose of streptozotocin. Rats were divided into control, T2D, and T2D + NaSH groups. NaSH (0.28, 0.56, 1.6, 2.8, and 5.6 mg/kg) was administered intraperitoneally for 9 weeks. At the end of the study, heart from all rats were isolated and left ventricular developed pressure (LVDP) and the peak rates of positive and negative changes in LV pressure (±dp/dt) were recorded during baseline and following myocardial IR injury. In addition, infarct size as well as mRNA expression of H2S- and nitric oxide (NO)-producing enzymes were measured. RESULTS In diabetic rats, NaSH only at doses of 0.56 and 1.6 mg/kg increased recovery of LVDP (16% and 42%), +dp/dt (25% and 35%) and -dp/dt (23% and 32%) as well as decreased infarct size (44% and 35%). At these doses, NaSH increased expressions of cystathionine γ-lyase (CSE) (440% and 271%) and endothelial NO synthase (eNOS) (232% and 148%) but it decreased the expressions of inducible NOS (iNOS) (55% and 71%). NaSH at 0.28, 2.8 and 5.6 mg/kg had no significant effects on these parameters. CONCLUSION NaSH had a bell-shaped cardioprotective effect against myocardial IR injury in rats with T2D. Higher tolerance to IR injury in heart isolated from type 2 diabetic rats treated with intermediate doses of NaSH is associated with higher CSE-derived H2S and eNOS-derived NO as well as lower iNOS-derived NO.
Collapse
|
22
|
Impairment of maturation of BMP-6 (35 kDa) correlates with delayed fracture healing in experimental diabetes. J Orthop Surg Res 2020; 15:186. [PMID: 32448307 PMCID: PMC7245805 DOI: 10.1186/s13018-020-01705-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/13/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Although it is known that diabetes interferes with fracture healing, the mechanisms remain poorly understood. The aim of this study was to investigate the correlation of BMP-6 and BMP-9 with the impairment in fracture healing in diabetes, by analyses of the difference in size and calcification of the callus, mechanical endurance, and expressing BMP-6 and BMP-9 in the callus, using a clinical related diabetic rodent model. METHODS We evaluated femur fracture healing by quantification of size and calcification of the callus by X-ray, histological and histochemical images, loading capacity of the fractured bone, and amount of BMP-6 in the callus and the bones using Western blot assay. RESULTS Significant upregulation of BMP-6 in the callus and the fractured bones of both non-diabetic and the diabetic animals was observed, at the end of the second and the fourth weeks after fracture. However, significantly lower levels of BMP-6 at 35 kDa with smaller sizes of calcified callus and poor loading capacity of the healing bones were detected in the diabetic animals, compared to the non-diabetic controls. The impairment of the maturation procedure of BMP-6 (35 kDa) from precursors may be underlying the downregulation of the BMP-6 in diabetic animals. CONCLUSIONS It could be concluded that the delayed fracture healing in the diabetic animals is correlated with deficiency of BMP-6 (35 kDa), which may be caused by impairment of maturation procedure of BMP-6 from precursors to functioning format. This is a primary study but an important step to explore the molecular pathogenesis of impairment of fracture healing in diabetes and to molecular therapeutic approach for the impairment of fracture healing.
Collapse
|
23
|
Ahmed N, Laghari AH, AlBkhoor B, Tabassum S, Meo SA, Muhammad N, Linardi D, Al-Masri AA, Fumagalli G, Luciani GB, Faggian G, Rungatscher A. Fingolimod Plays Role in Attenuation of Myocardial Injury Related to Experimental Model of Cardiac Arrest and Extracorporeal Life Support Resuscitation. Int J Mol Sci 2019; 20:ijms20246237. [PMID: 31835656 PMCID: PMC6940876 DOI: 10.3390/ijms20246237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/22/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Sudden cardiac arrest is a major global health concern, and survival of patients with ischemia–reperfusion injury is a leading cause of myocardial dysfunction. The mechanism of this phenomenon is not well understood because of the complex pathophysiological nature of the disease. Aim of the study was to investigate the cardioprotective role of fingolimod in an in vivo model of cardiac arrest and resuscitation. Methods: In this study, an in vivo rat model of cardiac arrest using extracorporeal membrane oxygenation resuscitation monitored by invasive hemodynamic measurement was developed. At the beginning of extracorporeal life support (ECLS), animals were randomly treated with fingolimod (Group A, n = 30) or saline (Group B, n = 30). Half of the animals in each group (Group A1 and B1, n = 15 each) were sacrificed after 1 h, and the remaining animals (Group A2 and B2) after 24 h of reperfusion. Blood and myocardial tissues were collected for analysis of cardiac features, inflammatory biomarkers, and cell signaling pathways. Results: Treatment with fingolimod resulted in activation of survival pathways resulting into reduced inflammation, myocardial oxidative stress and apoptosis of cardiomyocytes. This led to significant improvement in systolic and diastolic functions of the left ventricle and improved contractility index. Conclusions: Sphingosine1phosphate receptor activation with fingolimod improved cardiac function after cardiac arrest supported with ECLS. Present study findings strongly support a cardioprotective role of fingolimod through sphingosine-1-phosphate receptor activation during reperfusion after circulatory arrest.
Collapse
Affiliation(s)
- Naseer Ahmed
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi 74800, Pakistan
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
- Correspondence:
| | - Abid H. Laghari
- Department of Medicine, section of Cardiology, Aga Khan University, Karachi 74800, Pakistan;
| | | | - Sobia Tabassum
- Department of Biological Sciences, International Islamic University, Islamabad 44000, Pakistan;
| | - Sultan Ayoub Meo
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (S.A.M.); (A.A.A.-M.)
| | - Nazeer Muhammad
- COMSATS University Islamabad, Wah Campus, Rawalpindi 47040, Pakistan;
| | - Daniele Linardi
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| | - Abeer A. Al-Masri
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (S.A.M.); (A.A.A.-M.)
| | - Guido Fumagalli
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona Medical School, 37134 Verona, Italy;
| | - Giovanni Battista Luciani
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| | - Giuseppe Faggian
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| | - Alessio Rungatscher
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| |
Collapse
|
24
|
Yang Y, Zhao M, Yu XJ, Liu LZ, He X, Deng J, Zang WJ. Pyridostigmine regulates glucose metabolism and mitochondrial homeostasis to reduce myocardial vulnerability to injury in diabetic mice. Am J Physiol Endocrinol Metab 2019; 317:E312-E326. [PMID: 31211620 DOI: 10.1152/ajpendo.00569.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diabetic patients are more susceptible to myocardial ischemia damage than nondiabetic patients, with worse clinical outcomes and greater mortality. The mechanism may be related to glucose metabolism, mitochondrial homeostasis, and oxidative stress. Pyridostigmine may improve vagal activity to protect cardiac function in cardiovascular diseases. Researchers have not determined whether pyridostigmine regulates glucose metabolism and mitochondrial homeostasis to reduce myocardial vulnerability to injury in diabetic mice. In the present study, autonomic imbalance, myocardial damage, mitochondrial dysfunction, and oxidative stress were exacerbated in isoproterenol-stimulated diabetic mice, revealing the myocardial vulnerability of diabetic mice to injury compared with mice with diabetes or exposed to isoproterenol alone. Compared with normal mice, the expression of glucose transporters (GLUT)1/4 phosphofructokinase (PFK) FB3, and pyruvate kinase isoform (PKM) was decreased in diabetic mice, but increased in isoproterenol-stimulated normal mice. Following exposure to isoproterenol, the expression of (GLUT)1/4 phosphofructokinase (PFK) FB3, and PKM decreased in diabetic mice compared with normal mice. The downregulation of SIRT3/AMPK and IRS-1/Akt in isoproterenol-stimulated diabetic mice was exacerbated compared with that in diabetic mice or isoproterenol-stimulated normal mice. Pyridostigmine improved vagus activity, increased GLUT1/4, PFKFB3, and PKM expression, and ameliorated mitochondrial dysfunction and oxidative stress to reduce myocardial damage in isoproterenol-stimulated diabetic mice. Based on these results, it was found that pyridostigmine may reduce myocardial vulnerability to injury via the SIRT3/AMPK and IRS-1/Akt pathways in diabetic mice with isoproterenol-induced myocardial damage. This study may provide a potential therapeutic target for myocardial damage in diabetic patients.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Ming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Xiao-Jiang Yu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Long-Zhu Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Xi He
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Juan Deng
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| | - Wei-Jin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi, People's Republic of China
| |
Collapse
|
25
|
Ahmed N, Mehmood A, Linardi D, Sadiq S, Tessari M, Meo SA, Rehman R, Hajjar WM, Muhammad N, Iqbal MP, Gilani AUH, Faggian G, Rungatscher A. Cardioprotective Effects of Sphingosine-1-Phosphate Receptor Immunomodulator FTY720 in a Clinically Relevant Model of Cardioplegic Arrest and Cardiopulmonary Bypass. Front Pharmacol 2019; 10:802. [PMID: 31379576 PMCID: PMC6656862 DOI: 10.3389/fphar.2019.00802] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/21/2019] [Indexed: 12/28/2022] Open
Abstract
Objective: FTY720, an immunomodulator derived from sphingosine-1-phosphate, has recently demonstrated its immunomodulatory, anti-inflammatory, anti-oxidant, anti-apoptotic and anti-inflammatory properties. Furthermore, FTY720 might be a key pharmacological target for preconditioning. In this preclinical model, we have investigated the effects of FTY720 on myocardium during reperfusion in an experimental model of cardioplegic arrest (CPA) and cardiopulmonary bypass. Methods: 30 Sprague–Dawley rats (300–350 g) were randomized into two groups: Group-A, treated with FTY720 1 mg/kg via intravenous cannulation, and Group-B, as control. After 15 min of treatment, rats underwent CPA for 30 min followed by initiation of extracorporeal life support for 2 h. Support weaning was done, and blood and myocardial tissues were collected for analysis. Hemodynamic parameters, inflammatory mediators, nitro-oxidative stress, neutrophil infiltration, immunoblotting analysis, and immunohistochemical staining were analyzed and compared between groups. Results: FTY720 treatment activated the Akt/Erk1/2 signaling pathways, reduced the level of inflammatory mediators, activated antiapoptotic proteins, and inhibited proapoptotic proteins, leading to reduced nitro-oxidative stress and cardiomyocyte apoptosis. Moreover, significant preservation of high-energy phosphates were observed in the FTY720-treated group. This resulted in improved recovery of left ventricular systolic and diastolic functions. Conclusion: The cardioprotective mechanism in CPA is associated with activation of prosurvival cell signaling pathways that prevents myocardial damage. FTY720 preserves high-energy phosphates attenuates myocardial inflammation and oxidative stress, and improves cardiac function.
Collapse
Affiliation(s)
- Naseer Ahmed
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan.,Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, Verona, Italy
| | - Adeela Mehmood
- Department of Pharmacology, Liaqat National Medical College, Karachi, Pakistan
| | - Daniele Linardi
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, Verona, Italy
| | - Soban Sadiq
- Pharmacology and Molecular Lab, University of Liverpool, United Kingdom
| | - Maddalena Tessari
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, Verona, Italy
| | - Sultan Ayoub Meo
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Rehana Rehman
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Waseem M Hajjar
- Department of Thoracic Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Nazeer Muhammad
- Department of Mathematics, COMSATS University Islamabad, Wah Campus, Pakistan
| | - Muhammad Perwaiz Iqbal
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Anwar-Ul-Hassan Gilani
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Giuseppe Faggian
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, Verona, Italy
| | - Alessio Rungatscher
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, Verona, Italy
| |
Collapse
|
26
|
Tawfik MK, Ameen AM. Cardioprotective effect of ranolazine in nondiabetic and diabetic male rats subjected to isoprenaline-induced acute myocardial infarction involves modulation of AMPK and inhibition of apoptosis. Can J Physiol Pharmacol 2019; 97:661-674. [DOI: 10.1139/cjpp-2018-0571] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Diabetes increases the sensitivity of myocardium to ischemic damage and impairs response of the myocardium to cardioprotective interventions. The present study aimed to elucidate the potential cardioprotective effect provided by ranolazine during myocardial infarction in nondiabetic and diabetic male rats. As AMP-activated protein kinase (AMPK) has been shown to be involved in the cellular response to ischemic injury, in this context, the present animal study evaluated the modulating role of ranolazine in the AMPK expression in isoprenaline-induced myocardial ischemic rat model. Male rats were divided into 2 experiments: experiment I and II (nondiabetic and diabetic rats) and assigned to normal control, saline control for isoprenaline, isoprenaline control, and ranolazine-treated groups. Ranolazine administration revealed effectiveness in attenuating the severity of isoprenaline-induced myocardial injury in both nondiabetic and diabetic rats as revealed by ECG signs, histopathological score, and apoptotic markers via abrogating the increments in the inflammatory and oxidative stress markers and modulating AMPK expression. Therefore, the current cardioprotective effect of ranolazine was, at least in part, mediated through inhibition of apoptosis and modulation of AMPK expression, encouraging considering the utility of ranolazine in protection from acute myocardial infarction.
Collapse
Affiliation(s)
- Mona K. Tawfik
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Angie M. Ameen
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
27
|
Cardioprotective Mechanisms of Exenatide in Isoprenaline-induced Myocardial Infarction: Novel Effects on Myocardial α-Estrogen Receptor Expression and IGF-1/IGF-2 System. J Cardiovasc Pharmacol 2019; 71:160-173. [PMID: 29256971 DOI: 10.1097/fjc.0000000000000557] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Myocardial infarction (MI) is one of the main causes of morbidity and mortality in diabetic patients. The antidiabetic glucagon-like polypeptide-1 receptor (GLP-1R) agonists, such as exenatide, proved to confer cardioprotection; however, their exact mechanisms are not fully elucidated. Although the cardioprotective effect of α-estrogen receptor (ERα) activation is well established, its involvement in exenatide-induced cardioprotection has never been investigated. Moreover, modulation of insulin-like growth factor-1/2 (IGF-1/IGF-2) system by exenatide, and the consequent effect on cardiomyocyte apoptosis, is yet to be established. Current study aimed to investigate the cardioprotective potential of exenatide versus the standard cardioprotective agent, 17β-estradiol, against isoprenaline (ISO)-induced MI in rats. MI-insulted group showed electrocardiographic abnormalities, elevated serum cardiac markers, higher serum IGF-2 level along with histopathological abnormalities. Treatment with exenatide and/or 17β-estradiol, commenced 8 weeks before ISO insult, ameliorated these anomalies with maximum cardioprotection achieved with combined treatment. This was associated with upregulation of both ERα and IGF-1R, and downregulation of IGF-2R in left ventricles. Inhibition of ERs in Langendorff preparations confirmed their involvement in mediating exenatide-induced cardioprotective effect. Current study showed that the GLP-1R agonist exenatide exerted cardioprotection associated with upregulation of ERα and modulation of IGF-1/IGF-2 signaling in favor of antiapoptosis.
Collapse
|
28
|
Sun J, Ren XS, Kang Y, Dai HB, Ding L, Tong N, Zhu GQ, Zhou YB. Intermedin in Paraventricular Nucleus Attenuates Sympathoexcitation and Decreases TLR4-Mediated Sympathetic Activation via Adrenomedullin Receptors in Rats with Obesity-Related Hypertension. Neurosci Bull 2019; 35:34-46. [PMID: 30276527 PMCID: PMC6357274 DOI: 10.1007/s12264-018-0292-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/14/2018] [Indexed: 01/11/2023] Open
Abstract
Intermedin/adrenomedullin-2 (IMD/AM2), a member of the calcitonin gene-related peptide/AM family, plays an important role in protecting the cardiovascular system. However, its role in the enhanced sympathoexcitation in obesity-related hypertension is unknown. In this study, we investigated the effects of IMD in the paraventricular nucleus (PVN) of the hypothalamus on sympathetic nerve activity (SNA), and lipopolysaccharide (LPS)-induced sympathetic activation in obesity-related hypertensive (OH) rats induced by a high-fat diet for 12 weeks. Acute experiments were performed under anesthesia. The dynamic alterations of sympathetic outflow were evaluated as changes in renal SNA and mean arterial pressure (MAP) in response to specific drugs. Male rats were fed a control diet (12% kcal as fat) or a high-fat diet (42% kcal as fat) for 12 weeks to induce OH. The results showed that IMD protein in the PVN was downregulated, but Toll-like receptor 4 (TLR4) and plasma norepinephrine (NE, indicating sympathetic hyperactivity) levels, and systolic blood pressure were increased in OH rats. LPS (0.5 µg/50 nL)-induced enhancement of renal SNA and MAP was greater in OH rats than in obese or control rats. Bilateral PVN microinjection of IMD (50 pmol) caused greater decreases in renal SNA and MAP in OH rats than in control rats, and inhibited LPS-induced sympathetic activation, and these were effectively prevented in OH rats by pretreatment with the AM receptor antagonist AM22-52. The mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) inhibitor U0126 in the PVN partially reversed the LPS-induced enhancement of SNA. However, IMD in the PVN decreased the LPS-induced ERK activation, which was also effectively prevented by AM22-52. Chronic IMD administration resulted in significant reductions in the plasma NE level and blood pressure in OH rats. Moreover, IMD lowered the TLR4 protein expression and ERK activation in the PVN, and decreased the LPS-induced sympathetic overactivity. These results indicate that IMD in the PVN attenuates SNA and hypertension, and decreases the ERK activation implicated in the LPS-induced enhancement of SNA in OH rats, and this is mediated by AM receptors.
Collapse
Affiliation(s)
- Jing Sun
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Xing-Sheng Ren
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Ying Kang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Hang-Bing Dai
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Lei Ding
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
- Department of Pathophysiology, Xuzhou Medical College, Xuzhou, 221004, China
| | - Ning Tong
- Department of Neurology, Heze Municipal Hospital, Heze, 274000, China
| | - Guo-Qing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 211166, China
| | - Ye-Bo Zhou
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
29
|
Yang X, Shao H, Chen Y, Ding N, Yang A, Tian J, Jiang Y, Li G, Jiang Y. In renal hypertension, Cirsium japonicum strengthens cardiac function via the intermedin/nitric oxide pathway. Biomed Pharmacother 2018. [DOI: 10.1016/j.biopha.2018.02.126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
30
|
Wang M, Wang J, Liu Z, Guo X, Wang N, Jia N, Zhang Y, Yuan J. Effects of intermedin on autophagy in cerebral ischemia/reperfusion injury. Neuropeptides 2018; 68:15-21. [PMID: 29128104 DOI: 10.1016/j.npep.2017.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/30/2017] [Accepted: 10/31/2017] [Indexed: 12/23/2022]
Abstract
OBJECTIVE This study aimed to evaluate the effects of intermedin (IMD) on autophagy in cerebral ischemia/reperfusion (I/R) injury (CIRI). METHODS Sixty rats were randomly averaged into four groups: sham, ischemia/reperfusion (I/R), IMD, and 3-methyladenine (3-MA). In the sham group, the right common carotid artery, external carotid artery, and internal carotid artery were detached, and no monofilament was inserted. In the other groups, two hours after cerebral ischemia, the rats were injected through the lateral ventricle with normal saline for I/R group, IMD for the IMD group, and 3-MA for the 3-MA group for 24h. The cerebral injury was assessed by evaluation of neurological function, hematoxylin and eosin (H&E) staining, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The expressions of autophagy associated proteins, such as microtubule-associated protein 1 light chain 3 (LC3), Beclin1, and sequestosome 1 (P62) were analyzed using immunohistochemistry staining and western blot. Meanwhile, transmission electron microscopy was used to investigate the ultrastructure of the brains. RESULTS IMD could reduce neuron cell damage and infarction formation and has a protective effect against CIRI as 3-MA. The levels of LC3II/LC3I and Beclin1 were significantly decreased and the P62 level was significantly higher in the IMD group compared with I/R group, which is similar to the effect of 3-MA on CIRI. CONCLUSIONS IMD has a similar effect as 3-MA, can reduce pathological neuronal injury and protect the brain against CIRI in rats by attenuating the effects of autophagy. Our findings provide evidence for IMD's protective effects in relation to ischemic cerebrovascular diseases.
Collapse
Affiliation(s)
- Meng Wang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei Province, China
| | - Jing Wang
- Department of Clinical Medicine, Tangshan Vocational and Technical College, Tangshan 063000, Hebei Province, China; Department of Internal Medicine, Tangshan Union Medical College Hospital, Tangshan 063000, Hebei Province, China
| | - Zhengang Liu
- Department of Neurosurgery, The Second People's Hospital of Liaocheng, Liaocheng 252600, Shandong province, China
| | - Xin Guo
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei Province, China
| | - Ning Wang
- College of Psychology, North China University of Science and Technology, Tangshan 063000, Hebei Province, China
| | - Nana Jia
- College of Psychology, North China University of Science and Technology, Tangshan 063000, Hebei Province, China
| | - Yanshu Zhang
- School of Public Health, North China University of Science and Technology, Tangshan 063000, Hebei Province, China
| | - Jie Yuan
- Institute of Mental Health, North China University of Science and Technology, Tangshan 063000, Hebei Province, China.
| |
Collapse
|
31
|
Ranjbar K, Zarrinkalam E, Salehi I, Komaki A, Fayazi B. Cardioprotective effect of resistance training and Crataegus oxyacantha extract on ischemia reperfusion–induced oxidative stress in diabetic rats. Biomed Pharmacother 2018; 100:455-460. [DOI: 10.1016/j.biopha.2018.02.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 01/03/2023] Open
|
32
|
Wu D, Shi L, Li P, Ni X, Zhang J, Zhu Q, Qi Y, Wang B. Intermedin 1-53 Protects Cardiac Fibroblasts by Inhibiting NLRP3 Inflammasome Activation During Sepsis. Inflammation 2018; 41:505-514. [PMID: 29192367 DOI: 10.1007/s10753-017-0706-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Sepsis is a disease that occurs as a result of systemic inflammatory response syndrome (SIRS) in response to an infection, contributing to multiple organ dysfunction and a high mortality rate. Interleukin-lβ (IL-1β) is a cytokine that plays critical roles in inflammation and cardiac dysfunction during severe sepsis. Intermedin1-53 (IMD1-53) has been recently discovered to possess potential endogenous anti-inflammatory and strong cardiovascular protective effects. To investigate whether IMD1-53 can inhibit the NLRP3/caspase-1/IL-1β pathway to alleviate cardiac injury and rescue heart function, sepsis was induced in vivo by caecal ligation and puncture (CLP) surgery, and lipopolysaccharides were used as septic stressors for cardiac fibroblasts (CFs) in vitro. The expressions of IMD1-53 receptors in sepsis rat heart were increased. After IMD1-53 treatment, inflammation caused by sepsis in vivo was greatly reduced, as shown by the downregulation of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), nucleotide-binding domain and leucine-rich repeat containing family, pyrin containing 3 (NLRP3), pro-IL-1β, caspase 1, and nuclear translocation of nuclear factor-κB (NF-kB) protein levels. In addition, cardiac function was significantly improved and mean arterial blood pressure (MABP) increased by 34.8% (P < 0.05) which almost back to normal. Surprisingly, IMD1-53 inhibited cell apoptosis, as caspase 3 activity and Bax expression was significantly reduced in the heart upon IMD1-53 treatment. IMD1-53 abolished the upregulation of ASC, NLRP3, and caspase 1 protein levels in CFs induced by lipopolysaccharide (LPS). IMD1-53 increased cell survival rates and inhibited IL-1β production in the cell culture medium. IMD1-53 can protect against inflammation and heart injury during sepsis via attenuating the NLRP3/caspase-1/IL-1β pathway.
Collapse
Affiliation(s)
- Di Wu
- The Peking University Aerospace School of Clinical Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Lin Shi
- The Peking University People's Hospital, Beijing, 100191, China
| | - Pengyang Li
- Texas Heart Institute, Houston, TX, 77030, USA
| | - Xianqiang Ni
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Jinsheng Zhang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Qing Zhu
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Yongfen Qi
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
| | - Bin Wang
- The Peking University Aerospace School of Clinical Medicine, Peking University Health Science Center, Beijing, 100191, China.
- The First Affiliated Hospital of Shantou University Medical College, Guangdong, 515041, China.
| |
Collapse
|
33
|
Beneficial Effect of Intermedin 1-53 in Septic Shock Rats: Contributions of Rho Kinase and BKCA Pathway-Mediated Improvement in Cardiac Function. Shock 2018; 46:557-565. [PMID: 27355401 DOI: 10.1097/shk.0000000000000639] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Intermedin (IMD) is a calcitonin gene-related peptide shown to have a protective effect on myocardial function in ischemia-reperfusion injury. Whether IMD has beneficial effect in severe sepsis and septic shock (and its underlying mechanisms) is not known. METHODS We induced septic shock using cecal ligation and puncture (CLP). We focused on the potential beneficial effect of IMD1-53 on cardiac papillary muscle and cardiomyocytes against septic shock and its relationship with the protection of cardiac function. RESULTS Early (immediately after CLP) and late (12 h after CLP) administration of IMD1-53 (0.5 μg/kg) improved animal survival significantly, increased cardiac contractility and function, and improved tissue perfusion and oxygen delivery. The effect of early administration of IMD1-53 was better than that of late administration. The Rho kinase/TnI and BKCa pathways participated in the protective effect of IMD1-53 on cardiac function in septic rats. An inhibitor of Rho kinase (Y-27632) or a BKCa opener (NS1619) abolished the protective effect of IMD1-53 on cardiac function. IMD1-53 increased expression of Rho kinase in cardiac muscle and inhibited TnI phosphorylation. IMD1-53 inhibited currents in BKCa channels and intracellular calcium concentration in cardiomyocytes. CONCLUSIONS IMD1-53 is beneficial against severe sepsis/septic shock. IMD1-53 can improve cardiac contractility and cardiac function significantly, and then improve tissue perfusion and oxygen delivery. Rho kinase and the BKCa pathways have important roles in these effects. These findings provide a new treatment strategy for severe sepsis with cardiac dysfunction.
Collapse
|
34
|
Telli G, Tel BC, Yersal N, Korkusuz P, Gumusel B. Effect of intermedin/adrenomedullin2 on the pulmonary vascular bed in hypoxia-induced pulmonary hypertensive rats. Life Sci 2018; 192:62-67. [DOI: 10.1016/j.lfs.2017.11.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 10/18/2022]
|
35
|
Fu W, Xu H, Yu X, Lyu C, Tian Y, Guo M, Sun J, Sui D. 20(S)-Ginsenoside Rg2 attenuates myocardial ischemia/reperfusion injury by reducing oxidative stress and inflammation: role of SIRT1. RSC Adv 2018; 8:23947-23962. [PMID: 35540288 PMCID: PMC9081734 DOI: 10.1039/c8ra02316f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/11/2018] [Indexed: 11/21/2022] Open
Abstract
Previously we demonstrated that 20(S)-ginsenoside Rg2 protects cardiomyocytes from H2O2-induced injury by inhibiting reactive oxygen species (ROS) production, increasing intracellular levels of antioxidants and attenuating apoptosis. We explored the protective effect of 20(S)-ginsenoside Rg2 on myocardial ischemia/reperfusion (MI/R) injury and to clarify its potential mechanism of action. Rats were exposed to 20(S)-ginsenoside Rg2 in the presence/absence of the silent information regulator SIRT(1) inhibitor EX527 and then subjected to MI/R. 20(S)-Ginsenoside Rg2 conferred a cardioprotective effect by improving post-ischemic cardiac function, decreasing infarct size, reducing the apoptotic index, diminishing expression of creatine kinase-MB, aspartate aminotransferase and lactate dehydrogenase in serum, upregulating expression of SIRT1, B-cell lymphoma-2, procaspase-3 and procaspase-9, and downregulating expression of Bax and acetyl (Ac)-p53. Pretreatment with 20(S)-ginsenoside Rg2 also resulted in reduced myocardial superoxide generation, gp91phox expression, malondialdehyde content, cardiac pro-inflammatory markers and increased myocardial activities of superoxide dismutase, catalase and glutathione peroxidase. These results suggested that MI/R-induced oxidative stress and inflammation were attenuated significantly by 20(S)-ginsenoside Rg2. However, these protective effects were blocked by EX527, indicating that SIRT1 signaling may be involved in the pharmacological action of 20(S)-ginsenoside Rg2. Our results demonstrated that 20(S)-ginsenoside Rg2 attenuates MI/R injury by reducing oxidative stress and inflammatory responses via SIRT1 signaling. 20(S)-Ginsenoside Rg2 confers a protective effect against MI/R injury via SIRT1 signaling, by alleviating oxidative stress and reducing myocardium inflammation.![]()
Collapse
Affiliation(s)
- Wenwen Fu
- Department of Pharmacology
- School of Pharmaceutical Sciences
- Jilin University
- Changchun 130021
- China
| | - Huali Xu
- Department of Pharmacology
- School of Pharmaceutical Sciences
- Jilin University
- Changchun 130021
- China
| | - Xiaofeng Yu
- Department of Pharmacology
- School of Pharmaceutical Sciences
- Jilin University
- Changchun 130021
- China
| | - Chen Lyu
- Department of Pharmacology
- School of Pharmaceutical Sciences
- Jilin University
- Changchun 130021
- China
| | - Yuan Tian
- Department of Pharmacology
- School of Pharmaceutical Sciences
- Jilin University
- Changchun 130021
- China
| | - Minyu Guo
- Department of Pharmacology
- School of Pharmaceutical Sciences
- Jilin University
- Changchun 130021
- China
| | - Jiao Sun
- School of Nursing
- Jilin University
- Changchun
- China
| | - Dayun Sui
- Department of Pharmacology
- School of Pharmaceutical Sciences
- Jilin University
- Changchun 130021
- China
| |
Collapse
|
36
|
Li H, Xiao CS, Bian YF, Bai R, Gao F. Intermedin attenuates high-glucose exacerbated simulated hypoxia/reoxygenation injury in H9c2 cardiomyocytes via ERK1/2 signaling. EUR J INFLAMM 2017. [DOI: 10.1177/1721727x17744096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: This study investigated whether and how intermedin (IMD) exerted a protective effect against simulated hypoxia/reoxygenation (H/R) injury in high-glucose-treated H9c2 cells. Methods: Cellular viability was assessed via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. Oxidative stress was determined by malondialdehyde and superoxide dismutase content in the culture medium supernatant. Flow cytometry with Annexin V/propidium iodide staining was used to detect the cardiomyocyte apoptosis rate. The protein expression of Bax, Bcl-2, caspase-3, and ERK1/2 was determined by western blot. Results: IMD administration to H9c2 cells during H/R injury decreased oxidative stress product generation and inhibited apoptosis ( P < 0.05 or P < 0.01) while these effects were blocked by the ERK1/2 inhibitor ( P < 0.05 or P < 0.01). Through the application of a specific ERK1/2 inhibitor, it was demonstrated that IMD mitigates high-glucose-induced oxidative stress and apoptosis via ERK1/2 signaling. Conclusion: Intermedin may be a novel therapeutic agent for mitigating diabetic cardiovascular injury in the clinical setting.
Collapse
Affiliation(s)
- Hong Li
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chuan-Shi Xiao
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yun-Fei Bian
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Rui Bai
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Fen Gao
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
37
|
Adrenomedullin protects Leydig cells against lipopolysaccharide-induced oxidative stress and inflammatory reaction via MAPK/NF-κB signalling pathways. Sci Rep 2017; 7:16479. [PMID: 29184072 PMCID: PMC5705677 DOI: 10.1038/s41598-017-16008-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/06/2017] [Indexed: 12/14/2022] Open
Abstract
This study aimed to explore the possible benefits of adrenomedullin (ADM) in preventing oxidative stress and inflammation by using an in vitro primary culture model of rat Leydig cells exposed to lipopolysaccharide (LPS). Cell proliferation was detected through CCK-8 and BrdU incorporation assays. ROS were determined with a DCFDA kit, and cytokine concentrations were measured with ELISA assay kits. Protein production was examined by immunohistochemical staining and Western blot, and gene expression was observed through RT-qPCR. Results revealed that ADM significantly reduced LPS-induced cytotoxicity, and pretreatment with ADM significantly suppressed ROS overproduction and decreased 4-HNE and 8-OHdG expression levels and concentrations. ADM pretreatment also significantly attenuated the overactivation of enzymatic antioxidants, namely, superoxide dismutase, catalase, thioredoxin reductase, glutathione peroxidase, glutathione reductase and glutathione-S-transferase. ADM supplementation reversed the significantly increased gene expression levels and concentrations of TNF-α, IL-1β, TGF-β1, MCP-1 and MIF. ADM pretreatment significantly inhibited the gene expression and protein production of TLR-2 and 4. Furthermore, ADM pretreatment markedly reduced the phosphorylation of JNK, ERK 1/2 and p38, phosphorylation and degradation of IκBα and nuclear translocation of p65. Our findings demonstrated that ADM protects Leydig cells from LPS-induced oxidative stress and inflammation, which might be associated with MAPK/NF-κB signalling pathways.
Collapse
|
38
|
Ahmed N, Linardi D, Muhammad N, Chiamulera C, Fumagalli G, Biagio LS, Gebrie MA, Aslam M, Luciani GB, Faggian G, Rungatscher A. Sphingosine 1-Phosphate Receptor Modulator Fingolimod (FTY720) Attenuates Myocardial Fibrosis in Post-heterotopic Heart Transplantation. Front Pharmacol 2017; 8:645. [PMID: 28966593 PMCID: PMC5605636 DOI: 10.3389/fphar.2017.00645] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022] Open
Abstract
Background and Objective: Sphingosine 1-phosphate (S1P), and S1P receptor modulator fingolimod have been suggested to play important cardioprotective role in animal models of myocardial ischemia/reperfusion injuries. To understand the cardioprotective function of S1P and its mechanism in vivo, we analyzed apoptotic, inflammatory biomarkers, and myocardial fibrosis in an in vivo heterotopic rat heart transplantation model. Methods: Heterotopic heart transplantation is performed in 60 Sprague–Dawley (SD) rats (350–400 g). The heart transplant recipients (n = 60) are categorized into Group A (control) and Group B (fingolimod treated 1 mg/kg intravenous). At baseline with 24 h after heart transplantation, blood and myocardial tissue are collected for analysis of myocardial biomarkers, apoptosis, inflammatory markers, oxidative stress, and phosphorylation of Akt/Erk/STAT-3 signaling pathways. Myocardial fibrosis was investigated using Masson’s trichrome staining and L-hydroxyline. Results: Fingolimod treatment activates both Reperfusion Injury Salvage Kinase (RISK) and Survivor Activating Factor Enhancement (SAFE) pathways as evident from activation of anti-apoptotic and anti-inflammatory pathways. Fingolimod treatment caused a reduction in myocardial oxidative stress and hence cardiomyocyte apoptosis resulting in a decrease in myocardial reperfusion injury. Moreover, a significant (p < 0.001) reduction in collagen staining and hydroxyproline content was observed in fingolimod treated animals 30 days after transplantation demonstrating a reduction in cardiac fibrosis. Conclusion: S1P receptor activation with fingolimod activates anti-apoptotic and anti-inflammatory pathways, leading to improved myocardial salvage causing a reduction in cardiac fibrosis.
Collapse
Affiliation(s)
- Naseer Ahmed
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy.,Faculty of Health Sciences, University of PunjabLahore, Pakistan.,Research Unit, Faculty of Allied Health Sciences, University of LahoreLahore, Pakistan.,Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Daniele Linardi
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Nazeer Muhammad
- COMSATS Institute of Information TechnologyWah Cantt, Pakistan
| | - Cristiano Chiamulera
- Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Guido Fumagalli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Livio San Biagio
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Mebratu A Gebrie
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy.,Department of Anatomy, Università di Addis AbebaAddis Ababa, Ethiopia
| | - Muhammad Aslam
- Department of Internal Medicine, Cardiology and Angiology, University Hospital, Justus Liebig UniversityGiessen, Germany
| | - Giovanni Battista Luciani
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Giuseppe Faggian
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Alessio Rungatscher
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| |
Collapse
|
39
|
NLRP3 Inflammasome Activation-Mediated Pyroptosis Aggravates Myocardial Ischemia/Reperfusion Injury in Diabetic Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9743280. [PMID: 29062465 PMCID: PMC5618779 DOI: 10.1155/2017/9743280] [Citation(s) in RCA: 272] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/22/2017] [Accepted: 08/17/2017] [Indexed: 02/07/2023]
Abstract
The reactive oxygen species- (ROS-) induced nod-like receptor protein-3 (NLRP3) inflammasome triggers sterile inflammatory responses and pyroptosis, which is a proinflammatory form of programmed cell death initiated by the activation of inflammatory caspases. NLRP3 inflammasome activation plays an important role in myocardial ischemia/reperfusion (MI/R) injury. Our present study investigated whether diabetes aggravated MI/R injury through NLRP3 inflammasome-mediated pyroptosis. Type 1 diabetic rat model was established by intraperitoneal injection of streptozotocin (60 mg/kg). MI/R was induced by ligating the left anterior descending artery (LAD) for 30 minutes followed by 2 h reperfusion. H9C2 cardiomyocytes were exposed to high glucose (HG, 30 mM) conditions and hypoxia/reoxygenation (H/R) stimulation. The myocardial infarct size, CK-MB, and LDH release in the diabetic rats subjected to MI/R were significantly higher than those in the nondiabetic rats, accompanied with increased NLRP3 inflammasome activation and increased pyroptosis. Inhibition of inflammasome activation with BAY11-7082 significantly decreased the MI/R injury. In vitro studies showed similar effects, as BAY11-7082 or the ROS scavenger N-acetylcysteine, attenuated HG and H/R-induced H9C2 cell injury. In conclusion, hyperglycaemia-induced NLRP3 inflammasome activation may be a ROS-dependent process in pyroptotic cell death, and NLRP3 inflammasome-induced pyroptosis aggravates MI/R injury in diabetic rats.
Collapse
|
40
|
Bayrami G, Karimi P, Agha-Hosseini F, Feyzizadeh S, Badalzadeh R. Effect of Ischemic Postconditioning on Myocardial Function and Infarct Size Following Reperfusion Injury in Diabetic Rats Pretreated With Vildagliptin. J Cardiovasc Pharmacol Ther 2017; 23:174-183. [PMID: 28901167 DOI: 10.1177/1074248417729881] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Cardioprotective actions of ischemic postconditioning (IPostC) against ischemia/reperfusion (I/R) injury are abolished in diabetic hearts. This study has investigated the combined effects of IPostC and vildagliptin (Vilda) on myocardial function and infarct size (IS) against I/R injury in diabetic myocardium. METHODS Diabetes was induced by a high-fat diet/low dose of streptozotocin (35 mg/kg; intraperitoneally) in Wistar rats (200-250 g) and lasted for 12 weeks. Vilda (6 mg/kg/d) was orally administered for 5 weeks in diabetic groups after seventh week of diabetes. At the end of the 12-week period, the hearts of rats were removed and subjected to 35-minute regional ischemia (through left anterior descending ligation) followed by 60-minute reperfusion, on Langendorff apparatus. Ischemic postconditioning was induced by 6 repetitive cycles of 10-second ischemia and 10-second reperfusion, immediately at the onset of the reperfusion. Myocardial hemodynamic was measured throughout the experiment. The IS was assessed by triphenyltetrazolium chloride staining method. The myocardial contents of troponin-I (cTnI), interleukin-6 (IL-6), and 8-isoprostane were measured in the homogenate from ischemic zone of left ventricles by enzyme-linked immunosorbent assay kit. RESULTS Pretreatment of the diabetic rats with Vilda significantly recovered the diabetes-induced reduction in left ventricular developed pressures and contractility at the baseline ( P < .05 to P < .01). After I/R injury, IPostC could not significantly improve the myocardial function, cTnI content, and IS of the diabetic hearts. However, in Vilda-treated hearts, concomitant application of IPostC significantly recovered the heart functions, returned cTnI content as well as myocardial IL-6 and 8-isoprostane levels back to the control values ( P < .01 to P < .001), and reduced IS more effectively (by 45%) in comparison to the diabetic group ( P < .001). CONCLUSION Besides its glycemic and lipid profile controlling effects, Vilda has a protective effect on heart function and tends to restore cardioprotective effects of IPostC on diabetic hearts.
Collapse
Affiliation(s)
- Goltaj Bayrami
- 1 Physiology Laboratory, Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouran Karimi
- 2 Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Agha-Hosseini
- 1 Physiology Laboratory, Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeid Feyzizadeh
- 3 Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Badalzadeh
- 1 Physiology Laboratory, Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
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
|
42
|
Wu Y, Xia ZY, Zhao B, Leng Y, Dou J, Meng QT, Lei SQ, Chen ZZ, Zhu J. (-)-Epigallocatechin-3-gallate attenuates myocardial injury induced by ischemia/reperfusion in diabetic rats and in H9c2 cells under hyperglycemic conditions. Int J Mol Med 2017; 40:389-399. [PMID: 28714516 PMCID: PMC5504977 DOI: 10.3892/ijmm.2017.3014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 05/23/2017] [Indexed: 11/27/2022] Open
Abstract
(−)-Epigallocatechin gallate (EGCG) exerts multiple beneficial effects on cardiovascular performance. In this study, we aimed to examine the effects of EGCG on diabetic cardiomyopathy during myocardial ischemia/reperfusion (I/R) injury. EGCG (100 mg/kg/day) was administered at week 6 for 2 weeks to diabetic rats following the induction of type 1 diabetes by streptozotocin (STZ). At the end of week 8, the animals were subjected to myocardial I/R injury. The EGCG-elicited structural and functional effects were analyzed. Additionally, EGCG (20 μM) was administered for 24 h to cultured cardiac H9c2 cells under hyperglycemic conditions (30 mM glucose) prior to hypoxia/reoxygenation (H/R) challenge, and its effects on oxidative stress were compared to H9c2 cells transfecteed with silent information regulator 1 (SIRT1) small interfering RNA (siRNA). In rats with STZ-induced diabetes, EGCG treatment ameliorated post-ischemic cardiac dysfunction, decreased the myocardial infarct size, apoptosis and cardiac fibrosis, and reduced the elevated lactate dehydrogenase (LDH) and malonaldehyde (MDA) levels, and attenuated oxidative stress. Furthermore, EGCG significantly reduced H/R injury in cardiac H9c2 cells exposed to high glucose as evidenced by reduced apoptotic cell death and oxidative stress. The protein expression levels of SIRT1 and manganese superoxide dismutase (MnSOD) were reduced in the diabetic rats and the H9c2 cells under hyperglycemic conditions, compared with the control rats following I/R injury and H9c2 cells under normal glucose conditions. EGCG pre-treatment significantly upregulated the levels of htese proteins in vitro and in vivo. However, treatment with EX527 and SIRT1 siRNA blocked the EGCG-mediated cardioprotective effects. Taken together, our data indicate that SIRT1 plays a critical role in the EGCG-mediated amelioration of I/R injury in diabetic rats, which suggests that EGCG may be a promising dietary supplement for the prevention of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan Leng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Juan Dou
- Department of Cardiac Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing-Tao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shao-Qing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhi-Ze Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jie Zhu
- Department of Clinical Nutrition, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
43
|
Wu D, Wang J, Wang H, Ji A, Li Y. Protective roles of bioactive peptides during ischemia-reperfusion injury: From bench to bedside. Life Sci 2017; 180:83-92. [PMID: 28527782 DOI: 10.1016/j.lfs.2017.05.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/14/2022]
Abstract
Ischemia-reperfusion (I/R) is a well-known pathological condition which may lead to disability and mortality. I/R injury remains an unresolved and complicated situation in a number of clinical conditions, such as cardiac arrest with successful reanimation, as well as ischemic events in brain and heart. Peptides have many attractive advantages which make them suitable candidate drugs in treating I/R injury, such as low toxicity and immunogenicity, good solubility property, distinct tissue distribution pattern, and favorable pharmacokinetic profile. An increasing number of studies indicate that peptides could protect against I/R injury in many different organs and tissues. Peptides also face several therapeutic challenges that limit their clinical application. In this review, we present the mechanisms of action of peptides in reducing I/R injury, as well as further discuss modification strategies to improve the functional properties of bioactive peptides.
Collapse
Affiliation(s)
- Dongdong Wu
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Jun Wang
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Honggang Wang
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Ailing Ji
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China.
| | - Yanzhang Li
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
44
|
Yayi H, Yeda X, Huaxin W, Yang W, Qian S, Zhongyuan X. Toll-like receptor 7 involves the injury in acute kidney ischemia/reperfusion of STZ-induced diabetic rats. Acta Cir Bras 2017; 31:448-55. [PMID: 27487279 DOI: 10.1590/s0102-865020160070000004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/24/2016] [Indexed: 12/31/2022] Open
Abstract
PURPOSE To determine whether Toll-like receptor 7 (TLR7) is the potential targets of prevention or progression in the renal ischemia/reperfusion (I/R) injury of STZ-induced diabetic rats. METHODS Thirty six Sprague-Dawley rats were randomly arranged to the nondiabetic (ND) or diabetic group (DM), with each group further divided into sham (no I/R injury), I/R (ischemia-reperfusion) and CD (given by Chloroquine) group. Preoperatively, Chloroquine (40 mg/kg, intraperitoneal injection.) was administrated 6 days for treatment group. I/R animals were subjected to 25 min of bilateral renal ischemia. Renal function, histology, apoptosis, cytokines, expression of TLR7, MyD88 and NF-κB were detected. RESULTS The serum levels of blood urea nitrogen, creatinine, IL-6 and TNF-α, apoptotic tubular epithelial cells, expression of TLR7, MyD88 and NF-κB were significantly increased in DM+I/R group, compared with ND+I/R group (p<0.05). All these changes were further improved by TLR7 inhibition Chloroquine except Paller scores (p<0.05). CONCLUSION Toll-like receptor 7 inhibition attenuates the acute renal ischemia/reperfusion injury of STZ-induced diabetic in SD rats.
Collapse
Affiliation(s)
- Huang Yayi
- Master, Department of Anesthesia, Renmin Hospital, Wuhan University, China. Conception and design of the study, acquisition and interpretation of data, manuscript writing
| | - Xiao Yeda
- Master, Department of Anesthesia, Renmin Hospital, Wuhan University, China. Acquisition of data, critical revision
| | - Wang Huaxin
- PhD, Department of Anesthesia, Renmin Hospital, Wuhan University, China. Acquisition of data
| | - Wu Yang
- PhD, Department of Anesthesia, Renmin Hospital, Wuhan University, China. Acquisition of data
| | - Sun Qian
- PhD, Department of Anesthesia, Renmin Hospital, Wuhan University, China. Acquisition of data
| | - Xia Zhongyuan
- PhD, Full Professor, Department of Anesthesia, Renmin Hospital, Wuhan University, China. Design and supervised all phases of the study, critical revision
| |
Collapse
|
45
|
Ding M, Dong Q, Liu Z, Liu Z, Qu Y, Li X, Huo C, Jia X, Fu F, Wang X. Inhibition of dynamin-related protein 1 protects against myocardial ischemia-reperfusion injury in diabetic mice. Cardiovasc Diabetol 2017; 16:19. [PMID: 28173848 PMCID: PMC5297196 DOI: 10.1186/s12933-017-0501-2] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/31/2017] [Indexed: 01/05/2023] Open
Abstract
Background Many cardioprotective pharmacological agents failed to exert their protective effects in diabetic hearts subjected to myocardial ischemia/reperfusion (MI/R). Identify the molecular basis linking diabetes with MI/R injury is scientifically important and may provide effective therapeutic approaches. Dynamin-related protein 1 (Drp1)-mediated mitochondrial fission plays an important role in MI/R injury under non-diabetic conditions. Importantly, recent studies indicated that Drp1-mediated mitochondrial fission is enhanced in the myocardium of diabetic mice. The above evidences suggested that Drp1 may be one critical molecule linking diabetes with MI/R injury. We hypothesized that inhibition of Drp1 may be effective to reduce MI/R injury in diabetic hearts. Methods High-fat diet and streptozotocin-induced diabetic mice were subjected to MI/R or sham operation. Mdivi-1 (1.2 mg/kg), a small molecule inhibitor of Drp1 or vehicle was administrated 15 min before the onset of reperfusion. Outcome measures included mitochondrial morphology, mitochondrial function, myocardial injury, cardiac function and oxidative stress. Results Mitochondrial fission was significantly increased following MI/R as evidenced by enhanced translocation of Drp1 to mitochondria and decreased mitochondrial size. Delivery of Mdivi-1 into diabetic mice markedly inhibited Drp1 translocation to the mitochondria and reduced mitochondrial fission following MI/R. Inhibition of Drp1 in diabetic hearts improved mitochondrial function and cardiac function following MI/R. Moreover, inhibition of Drp1 reduced myocardial infarct size and serum cardiac troponin I and lactate dehydrogenase activities. These cardioprotective effects were associated with decreased cardiomyocyte apoptosis and malondialdehyde production and increased activities of antioxidant enzyme manganese superoxide dismutase. Conclusions Pharmacological inhibition of Drp1 prevents mitochondrial fission and reduces MI/R injury in diabetic mice. The findings suggest Drp1 may be a potential novel therapeutic target for diabetic cardiac complications.
Collapse
Affiliation(s)
- Mingge Ding
- Department of Geriatrics, Xi'an Central Hospital, Xi'an, 710003, China.,Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, 15 Changlexi Road, Xi'an, 710032, China
| | - Qianqian Dong
- Department of Natural Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhenghua Liu
- Department of Physiology, Fourth Military Medical University, 169 Changlexi Road, Xi'an, 710032, China
| | - Zheng Liu
- Department of Physiology, Fourth Military Medical University, 169 Changlexi Road, Xi'an, 710032, China
| | - Yinxian Qu
- Department of Geriatrics, Xi'an Central Hospital, Xi'an, 710003, China
| | - Xing Li
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, 15 Changlexi Road, Xi'an, 710032, China
| | - Cong Huo
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, 15 Changlexi Road, Xi'an, 710032, China
| | - Xin Jia
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, 15 Changlexi Road, Xi'an, 710032, China
| | - Feng Fu
- Department of Physiology, Fourth Military Medical University, 169 Changlexi Road, Xi'an, 710032, China.
| | - Xiaoming Wang
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, 15 Changlexi Road, Xi'an, 710032, China.
| |
Collapse
|
46
|
Lei J, Zhu F, Zhang Y, Duan L, Lei H, Huang W. Transient Receptor Potential Vanilloid Subtype 1 Inhibits Inflammation and Apoptosis via the Release of Calcitonin Gene-Related Peptide in the Heart after Myocardial Infarction. Cardiology 2016; 134:436-43. [PMID: 27144592 DOI: 10.1159/000444439] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/04/2016] [Indexed: 11/19/2022]
Abstract
OBJECTIVE A high mortality rate occurs with silent myocardial infarction (MI), particularly in aging and diabetic populations due to defects in the transient receptor potential vanilloid (TRPV1)-positive sensory nerve function. We have previously shown that TRPV1 deficiency markedly enhances post-MI inflammation and remodeling. However, the mechanisms remain unknown. The objective of this study was to clarify whether calcitonin gene-related peptide (CGRP) release was associated with the protective role of TRPV1 against postmyocardial inflammation and apoptosis. METHODS TRPV1 gene knockout (TRPV1KO) and wild-type (WT) mice were subjected to left anterior descending ligation or sham operation. The concentration of CGRP in the myocardium was measured at 30 min, 1, 6 and 24 h post-MI. Mice received saline vehicle, CGRP or the CGRP antagonist CGRP8-37 before ligation. Inflammation was evaluated by ELISA assay and histological staining. Apoptosis was assessed by Western blot and TUNEL assay. RESULTS Post-MI, both TRPV1KO and WT mice displayed elevated CGRP levels in myocardium when compared to sham controls. However, the levels of CGRP were significantly lower in TRPV1KO mice than in WT mice at 30 min after MI. Exogenous CGRP downregulated the levels of tumor necrosis factor-α and interleukin-6 expression in TRPV1KO mice post-MI. Moreover, exogenous CGRP decreased the neutrophil infiltration in TRPV1KO mice, whereas inhibition of CGRP by CGRP8-37 increased the neutrophil infiltration in WT mice. Western blotting data indicated that CGRP attenuated caspase-3 and caspase-9 expression, and enhanced Bcl-2 expression in TRPV1KO mice post-MI. CGRP8-37 upregulated caspase-3 and caspase-9 expression and downregulated Bcl-2 expression in WT mice. CONCLUSION Our data suggest a protective role of TRPV1 activation against inflammation and apoptosis in mice post-MI, possibly through CGRP release. These findings elucidate a neurogenic mechanism in mice post-MI, which may participate in sensory neurotransmitter-mediated protection in TRPV1 activation.
Collapse
Affiliation(s)
- Jiayan Lei
- Department of Cardiology, First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
47
|
Pang Y, Li Y, Lv Y, Sun L, Zhang S, Li Y, Wang Y, Liu G, Xu MJ, Wang X, Jiang C. Intermedin Restores Hyperhomocysteinemia-induced Macrophage Polarization and Improves Insulin Resistance in Mice. J Biol Chem 2016; 291:12336-45. [PMID: 27080257 DOI: 10.1074/jbc.m115.702654] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Indexed: 12/18/2022] Open
Abstract
Hyperhomocysteinemia (HHcy) is a condition characterized by an abnormally high level of homocysteine, an inflammatory factor. This condition has been suggested to promote insulin resistance. To date, the underlying molecular mechanism remains largely unknown, and identifying novel therapeutic targets for HHcy-induced insulin resistance is of high priority. It is well known that intermedin (IMD), a calcitonin family peptide, exerts potent anti-inflammatory effects. In this study, the effects of IMD on HHcy-induced insulin resistance were investigated. Glucose tolerance and insulin tolerance tests were performed on mice treated with IMD by minipump implantation (318 ng/kg/h for 4 weeks) or adipocyte-specific IMD overexpression mice (Adipo-IMD transgenic mice). The expression of genes and proteins related to M1/M2 macrophages and endoplasmic reticulum stress (ERS) was evaluated in adipose tissues or cells. The expression of IMD was identified to be lower in the plasma and adipose tissues of HHcy mice. In both IMD treatment by minipump implantation and Adipo-IMD transgenic mice, IMD reversed HHcy-induced insulin resistance, as revealed by glucose tolerance and insulin tolerance tests. Further mechanistic study revealed that IMD reversed the Hcy-elevated ratio of M1/M2 macrophages by inhibiting AMP-activated protein kinase activity. Adipo-IMD transgenic mice displayed reduced ERS and lower inflammation in adipose tissues with HHcy. Soluble factors from Hcy-treated macrophages induced adipocyte ERS, which was reversed by IMD treatment. These findings revealed that IMD treatment restores the M1/M2 balance, inhibits chronic inflammation in adipose tissues, and improves systemic insulin sensitivity of HHcy mice.
Collapse
Affiliation(s)
- Yanli Pang
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Center for Reproductive Medicine of Third Hospital, Peking University, Beijing 100191, China
| | - Yang Li
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and
| | - Ying Lv
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and
| | - Lulu Sun
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and
| | - Songyang Zhang
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and
| | - Yin Li
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and
| | - Yuhui Wang
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and
| | - George Liu
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and
| | - Ming-Jiang Xu
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and
| | - Xian Wang
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and
| | - Changtao Jiang
- From the Department of Physiology and Pathophysiology, Basic Medical College, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and
| |
Collapse
|
48
|
Zhang JS, Hou YL, Lu WW, Ni XQ, Lin F, Yu YR, Tang CS, Qi YF. Intermedin 1-53 Protects Against Myocardial Fibrosis by Inhibiting Endoplasmic Reticulum Stress and Inflammation Induced by Homocysteine in Apolipoprotein E-Deficient Mice. J Atheroscler Thromb 2016; 23:1294-1306. [PMID: 27052784 PMCID: PMC5113747 DOI: 10.5551/jat.34082] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIM Endoplasmic reticulum stress (ERS) and inflammation participate in cardiac fibrosis. Importantly, a novel paracrine/autocrine peptide intermedin1-53 (IMD1-53) in the heart inhibits myocardial fibrosis in rats. However, the mechanisms are yet to be fully elucidated. METHODS Myocardial fibrosis in apolipoprotein E-deficient (ApoE -/-) mice and neonatal rat cardiac fibroblasts (CFs) were induced using homocysteine (Hcy). RESULTS IMD1-53 inhibited myocardial fibrosis in vivo and in vitro. Picrosirius red staining showed that IMD1-53 reduced myocardial interstitial collagen deposition in ApoE-/- mice treated with Hcy and decreased the expression of myocardial collagen I and III, which was further verified in rat CFs. IMD1-53 attenuated myocardial hypertrophy, as shown by cardiomyocyte cross-sectional area, ratio of heart weight to body weight, and mRNA levels of atrial natriuretic peptide and brain natriuretic peptide. IMD1-53 inhibited the upregulation of ERS hallmarkers such as glucose-regulated protein 78 (GRP78), GRP94, activating transcription factor 6 (ATF6), ATF4, inositol-requiring enzyme 1α, spliced-X-box-binding protein-1, protein kinase receptor-like ER kinase, and eukaryotic translation initiation factor 2α in mouse myocardium and rat CFs treated with Hcy. In addition, IMD1-53 decreased the production of inflammatory factors such as tumor necrosis factor-α, monocyte chemotactic protein-1, interleukin-6 (IL-6), and IL-1β in the mouse myocardium and rat CFs treated with Hcy. Concurrently, IMD1-53 ameliorated the expression of nuclear factor-κB, transforming growth factor-β1, and c-Jun N-terminal kinase in the mouse myocardium and rat CFs treated with Hcy. CONCLUSIONS IMD potentially protects against myocardial fibrosis induced by Hcy in ApoE-/- mice, possibly via attenuating myocardial ERS and inflammation.
Collapse
Affiliation(s)
- Jin-Sheng Zhang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Wang Y, Tian J, Qiao X, Su X, Mi Y, Zhang R, Li R. Intermedin protects against renal ischemia-reperfusion injury by inhibiting endoplasmic reticulum stress. BMC Nephrol 2015; 16:169. [PMID: 26498843 PMCID: PMC4619099 DOI: 10.1186/s12882-015-0157-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/02/2015] [Indexed: 11/15/2022] Open
Abstract
Background Intermedin (IMD) is a novel member of the calcitonin/calcitonin gene-related peptide family. Endoplasmic reticulum stress (ERS) has been implicated in the pathology of renal ischemia/reperfusion (IRI). In the present study, we investigated whether IMD could reduce ERS damage after renal ischemia. Methods The kidneys of SD rats were subjected to 45 min of warm ischemia followed by 24 h of reperfusion. The hypoxia/reoxygenation(H/R) model in NRK-52E cells consisted of hypoxia for 1 h and reoxygenation for 2 h. IMD was over-expressed in vivo and in vitro using the vector pcDNA3.1-IMD. The serum creatinine concentration and lactate dehydrogenase (LDH) activity in the plasma were determined. Histologic examinations of renal tissues were performed with PAS staining. Real-time PCR and Western blotting were used to determine the mRNA and protein levels, respectively. Additionally, ER staining was used to detect the ERS response. Results In the rat renal IRI model, we found that IMD gene transfer markedly improved renal function and pathology and decreased LDH activity and cell apoptosis compared with the kidneys that were transfected with the control plasmid. IMD significantly attenuated the ERS stress parameters compared with IRI group. Indeed, IMD down-regulated glucose-regulated protein 78 (GRP78), C/EBP homologous protein(CHOP), and caspase 12 protein and mRNA levels. Moreover, in the NRK-52E cell H/R model, IMD overexpression prevented the apoptosis induced by H/R. Furthermore, IMD ameliorated the ER structural changes and concomitantly decreased the levels of GRP78, CHOP and caspase-12. Conclusion This study revealed that IMD protects against renal IRI by suppressing ERS and ERS-related apoptosis.
Collapse
Affiliation(s)
- Yanhong Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Jihua Tian
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Xi Qiao
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Xiaole Su
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Yang Mi
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Ruijing Zhang
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Rongshan Li
- Department of Nephrology, the Affiliated People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, No. 29 Shuang Ta East Street, Taiyuan, 030012, , Shanxi, P. R. China.
| |
Collapse
|
50
|
SIRT1 protects against myocardial ischemia-reperfusion injury via activating eNOS in diabetic rats. Cardiovasc Diabetol 2015; 14:143. [PMID: 26489513 PMCID: PMC4618275 DOI: 10.1186/s12933-015-0299-8] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 09/30/2015] [Indexed: 12/20/2022] Open
Abstract
Background Diabetic patients are more sensitive to myocardial ischemic injury than non-diabetic patients. Silent information regulator 1 (SIRT1) is a nicotinamide adenine dinucleotide-dependent histone deacetylase making the heart more resistant to ischemic injury. As SIRT1 expression is considered to be reduced in diabetic heart, we therefore hypothesized that up-regulation of SIRT1 in the diabetic heart may overcome its increased susceptibility to ischemic injury. Methods Male Sprague–Dawley rats were fed with high-fat diet and injected with streptozotocin once to induce diabetes. Diabetic rats received injections of adenoviral vectors encoding SIRT1 (Ad-SIRT1) at five myocardial sites. Four days after adenoviral injection, the rats were subjected to myocardial ischemia and reperfusion (MI/R). Outcome measures included left ventricular function, infarct size, cellular death and oxidative stress. Results Delivery of Ad-SIRT1 into the hearts of diabetic rats markedly increased SIRT1 expression. Up-regulation of SIRT1 in diabetic hearts improved cardiac function and reduced infarct size to the extent as in non-diabetic animals following MI/R, which was associated with reduced serum creatine kinase-MB, lactate dehydrogenase activities and cardiomyocyte apoptosis. Moreover, Ad-SIRT1 reduced the increase in the superoxide generation and malonaldialdehyde content and simultaneously increased the antioxidant capability. Furthermore, Ad-SIRT1 increased eNOS phosphorylation and reduced eNOS acetylation in diabetic hearts. NOS inhibitor L-NAME inhibited SIRT1-enhanced eNOS phosphorylation, and blunted SIRT1-mediated anti-apoptotic and anti-oxidative effects and cardioprotection. Conclusions Overexpression of SIRT1 reduces diabetes-exacerbated MI/R injury and oxidative stress via activating eNOS in diabetic rats. The findings suggest SIRT1 may be a promising novel therapeutic target for diabetic cardiac complications.
Collapse
|