1
|
Gao F, Zhang Y, Jin L, Gong H, Zhang X, Zheng Q, Chen Z, Qin X. Motor protein KIF5B inhibition as a novel strategy of controlled reperfusion against myocardial ischemia/reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167785. [PMID: 40057204 DOI: 10.1016/j.bbadis.2025.167785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/13/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025]
Abstract
Metabolic dysregulation triggered by nutrient influx at reperfusion onset induces reactive oxygen species (ROS) burst and cellular injury, contributing to the detrimental effects observed in ischemia/reperfusion (I/R) injury. Thus, implementing controlled reperfusion emerges as a superior cardioprotective strategy to alleviate reperfusion injury. Kinesin KIF5B transports GLUT4- and CD36-containing vesicles to the plasma membrane, facilizing the import of glucose and fatty acids into cells, suggesting a role in controlled reperfusion. Herein, we aim to investigate its specific role in myocardial I/R injury. By genetic and pharmacological modulation of KIF5B, we investigated its role in myocardial I/R injury both in vivo and in vitro. During reperfusion, a coordinated inhibition of metabolism-related genes and KIF5B expression occurred, probably mitigating the metabolic stress encountered as a compensatory mechanism. Genetic inhibition of KIF5B using AAV9-shRNA attenuated myocardial I/R injury, as evidenced by reduced infarct size, decreased cardiac biomarkers, and reduced cell apoptosis. Additionally, KIF5B inhibition mitigated post-reperfusion oxidative stress and arrhythmias. Mechanistically, concurrent decrease in CD36 membrane translocation following KIF5B ablation post-reperfusion was confirmed by immunofluorescence double staining, and siRNA knockdown of Kif5b inhibited fatty acids uptake in isolated primary neonatal rat cardiomyocytes. Intraperitoneal administration of rose bengal lactone (RBL, 1 mg/kg), a selective inhibitor of KIF5B, was shown to confer cardioprotective effects against myocardial I/R injury. Our findings demonstrate that the inhibition of KIF5B, as a novel strategy of controlled reperfusion, provides cardioprotection against myocardial I/R injury, and highlights the clinical potential of its inhibitor, RBL, to ameliorate reperfusion injury.
Collapse
Affiliation(s)
- Feng Gao
- Xi'an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Yudi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Lingyan Jin
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Haoyu Gong
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xing Zhang
- Department of Aerospace Medicine, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Ziwei Chen
- Research Center for Prevention and Treatment of Respiratory Disease, School of Clinical Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, China.
| | - Xinghua Qin
- Xi'an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
2
|
Bai J, Yang SY, Yu SM, Cao Y, Ma CH, Hu XY, Chen X, Song YN, Chen HJ. IGFBP5 mediates the therapeutic effect of isoliquiritigenin in myocardial ischemia-reperfusion injury via AKT/GLUT4 regulated insulin resistance. Front Pharmacol 2025; 16:1544869. [PMID: 40365313 PMCID: PMC12069378 DOI: 10.3389/fphar.2025.1544869] [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: 12/13/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Background Myocardial ischemia/reperfusion injury (MIRI) is a critical problem in cardiovascular medicine, often occurring after coronary revascularization procedures or cardiopulmonary bypass. The characters of MIRI are both energy metabolism disturbances and severe myocardium insulin resistance (IR), which exacerbated myocardial damage and cell death. Isoliquiritigenin (ISL), a flavonoid derived from licorice roots (Glycyrrhiza spp.), has demonstrated protective effects on MIRI. However, the potential cardio-protective effects and mechanism of ISL in MIRI remain unclear. Propose In this study, we aimed to investigate ISL's therapeutic effects on MIRI. Moreover, we elucidate the underlying mechanisms of ISL regulated myocardium insulin resistance in vivo and in vitro. Methods In vivo, SD rats underwent left anterior descending coronary artery ligation/reperfusion to induce MIRI. Chest echocardiography was performed to monitor cardiac function post-reperfusion, followed by measurement of myocardial injury and IR markers. In vitro, H9C2 cardiomyocytes subjected to oxygen-glucose deprivation/reperfusion (OGD/R). Markers associated with myocardial injury and IR were assessed. Then, we identified potential therapeutic targets IGFBP5 for MIRI by network pharmacology and molecular docking analysis. Finally, lentivirus were used to silence or over-express IGFBP5 to elucidate the role of IGFBP5 in regulating the therapeutic effects of ISL on IR in MIRI. Results In the present study, In vivo experiments demonstrated that ISL attenuated myocardial infarct size, decreased serum markers of myocardial injury, improved left ventricular systolic function, and enhanced insulin sensitivity. In vitro data revealed that ISL ameliorated glucose uptake and cell survival rate. Furthermore, ISL increased AKT phosphorylation and upregulated membrane-bound GLUT4 (M-GLUT4) protein expression levels. These effects of ISL are mediated by the induction of IGFBP5, as demonstrated using gene-specific shRNA or overexpression for IGFBP5. Conclusion Our results reveal that ISL protects against myocardial damage caused by MIRI through the regulation of IR via the IGFBP5/AKT/GLUT4 pathway.
Collapse
Affiliation(s)
- Jue Bai
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Si-Yuan Yang
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Shao-Mei Yu
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Ying Cao
- Department of Anesthesiology, The Affliated JinYang Hospital of Guizhou Medical University, The Second People’s Hospital of Guiyang, Guiyang, Guizhou, China
| | - Chang-Han Ma
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xuan-Yi Hu
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiong Chen
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying-Nan Song
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Hong-Jin Chen
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
3
|
Wu H, Hui Y, Qian X, Wang X, Xu J, Wang F, Pan S, Chen K, Liu Z, Gao W, Bai J, Liang G. Exosomes derived from mesenchymal stem cells ameliorate impaired glucose metabolism in myocardial Ischemia/reperfusion injury through miR-132-3p/PTEN/AKT pathway. Cell Cycle 2025:1-20. [PMID: 40181235 DOI: 10.1080/15384101.2025.2485834] [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: 03/12/2023] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 04/05/2025] Open
Abstract
Exosomes secreted by mesenchymal stem cells (MSCs) have been considered as a novel biological therapy for myocardial ischemia/reperfusion injury (MIRI). However, the underlying mechanism of exosomes has not been completely established, especially in the early stage of MIRI. In this study, we primarily investigated the protective effect of exosomes on MIRI from both in vitro and ex vivo perspectives. Bioinformatic analysis was conducted to identify exosomal miRNA associated with myocardial protection, Genes and proteins related to functional studies and myocardial energy metabolism were analyzed and evaluated using techniques such as Polymerase Chain Re-action (PCR), Western blotting, double luciferase biochemical techniques, flow cytometry assay, etc. It was discovered that exosomes ameliorated cardiomyocyte injury t by delivery of miR-132-3p.This process reduced the expression of Phosphatase and tensin homolog (PTEN) mRNA and protein, enhanced the expression of phosphorylated protein kinase (pAKT), regulated the insulin signaling pathway, facilitated intracellular Glucose transporter 4 (GLUT4) protein membrane translocation, and enhanced glucose uptake and Adenosine Triphosphate (ATP) production. This study confirmed, for the first time, that MSC-EXO can provide myocardial protection in the early stages of MIRI through miR-132/PTEN/AKT pathway. This research establishes a theoretical and experimental foundation for the clinical application of MSC-derived exosomes.
Collapse
Affiliation(s)
- Hongkun Wu
- School of basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Cardiac Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yongpeng Hui
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xingkai Qian
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Cardiac Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xueting Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, University Town, Guiyang, China
| | - Jianwei Xu
- National Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Center for Tissue Engineering and Stem Cell Research, Guizhou Province Key Laboratory of Regenerative Medicine, Guizhou Medical University, Guiyang, China
| | - Feng Wang
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Sisi Pan
- Department of Cardiac Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Kaiyuan Chen
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhou Liu
- School of basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Weilong Gao
- School of basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jue Bai
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Cardiac Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Guiyou Liang
- Department of Cardiac Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
4
|
Chen KY, Liu Z, Yi J, Hui YP, Song YN, Lu JH, Chen HJ, Yang SY, Hu XY, Zhang DS, Liang GY. PDHA1 Alleviates Myocardial Ischemia-Reperfusion Injury by Improving Myocardial Insulin Resistance During Cardiopulmonary Bypass Surgery in Rats. Cardiovasc Drugs Ther 2025; 39:17-31. [PMID: 37610688 DOI: 10.1007/s10557-023-07501-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 08/24/2023]
Abstract
OBJECTIVE Cardiopulmonary bypass (CPB) is a requisite technique for thoracotomy in advanced cardiovascular surgery. However, the consequent myocardial ischemia-reperfusion injury (MIRI) is the primary culprit behind cardiac dysfunction and fatal consequences post-operation. Prior research has posited that myocardial insulin resistance (IR) plays a vital role in exacerbating the progression of MIRI. Nonetheless, the exact mechanisms underlying this phenomenon remain obscure. METHODS We constructed pyruvate dehydrogenase E1 α subunit (PDHA1) interference and overexpression rats and used ascending aorta occlusion in an in vivo model of CPB-MIRI. We devised an in vivo model of CPB-MIRI by constructing rat models with both pyruvate dehydrogenase E1α subunit (PDHA1) interference and overexpression through ascending aorta occlusion. We analyzed myocardial glucose metabolism and the degree of myocardial injury using functional monitoring, biochemical assays, and histological analysis. RESULTS We discovered a clear downregulation of glucose transporter 4 (GLUT4) protein content expression in the CPB I/R model. In particular, cardiac-specific PDHA1 interference resulted in exacerbated cardiac dysfunction, significantly increased myocardial infarction area, more pronounced myocardial edema, and markedly increased cardiomyocyte apoptosis. Notably, the opposite effect was observed with PDHA1 overexpression, leading to a mitigated cardiac dysfunction and decreased incidence of myocardial infarction post-global ischemia. Mechanistically, PDHA1 plays a crucial role in regulating the protein content expression of GLUT4 on cardiomyocytes, thereby controlling the uptake and utilization of myocardial glucose, influencing the development of myocardial insulin resistance, and ultimately modulating MIRI. CONCLUSION Overall, our study sheds new light on the pivotal role of PDHA1 in glucose metabolism and the development of myocardial insulin resistance. Our findings hold promising therapeutic potential for addressing the deleterious effects of MIRI in patients.
Collapse
Affiliation(s)
- Kai-Yuan Chen
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Zhou Liu
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Jing Yi
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou Province, China
| | - Yong-Peng Hui
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Ying-Nan Song
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Jun-Hou Lu
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Hong-Jin Chen
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China
| | - Si-Yuan Yang
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
| | - Xuan-Yi Hu
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China
| | - Deng-Shen Zhang
- Department of Cardiovascular Surgery, the Affiliated Hospital of Zunyi Medical University, Zunyi, 563009, Guizhou Province, China
| | - Gui-You Liang
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang, 550001, Guizhou Province, China.
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, Guizhou Province, China.
| |
Collapse
|
5
|
Moran TB, Birnbaum Y. Enhancing Glucose Uptake as a Means to Protect the Heart During Cardiopulmonary Bypass or Ischemia-Reperfusion Injury. Cardiovasc Drugs Ther 2025; 39:15-16. [PMID: 39535626 DOI: 10.1007/s10557-024-07648-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Tyler B Moran
- The Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Yochai Birnbaum
- The Section of Cardiology, Baylor College of Medicine, Houston, TX, USA.
- John S. Dunn Chair in Cardiology Research and Education, The Department of Medicine, Section of Cardiology, Baylor College of Medicine, 7200 Cambridge Street, MS BCM620, Houston, TX, 77030, USA.
| |
Collapse
|
6
|
Tian H, Zhao X, Zhang Y, Xia Z. Abnormalities of glucose and lipid metabolism in myocardial ischemia-reperfusion injury. Biomed Pharmacother 2023; 163:114827. [PMID: 37141734 DOI: 10.1016/j.biopha.2023.114827] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/23/2023] [Accepted: 04/30/2023] [Indexed: 05/06/2023] Open
Abstract
Myocardial ischemia-reperfusion injury is a common condition in cardiovascular diseases, and the mechanism of its occurrence involves multiple complex metabolic pathways and signaling pathways. Among these pathways, glucose metabolism and lipid metabolism play important roles in regulating myocardial energy metabolism. Therefore, this article focuses on the roles of glucose metabolism and lipid metabolism in myocardial ischemia-reperfusion injury, including glycolysis, glucose uptake and transport, glycogen metabolism and the pentose phosphate pathway; and triglyceride metabolism, fatty acid uptake and transport, phospholipid metabolism, lipoprotein metabolism, and cholesterol metabolism. Finally, due to the different alterations and development of glucose metabolism and lipid metabolism in myocardial ischemia-reperfusion, there are also complex interregulatory relationships between them. In the future, modulating the equilibrium between glucose metabolism and lipid metabolism in cardiomyocytes and ameliorating aberrations in myocardial energy metabolism represent highly promising novel strategies for addressing myocardial ischemia-reperfusion injury. Therefore, a comprehensive exploration of glycolipid metabolism can offer novel theoretical and clinical insights into the prevention and treatment of myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Hao Tian
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xiaoshuai Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yuxi Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
7
|
Liu XS, Zeng J, Yang YX, Qi CL, Xiong T, Wu GZ, Zeng CY, Wang DX. DRD4 Mitigates Myocardial Ischemia/Reperfusion Injury in Association With PI3K/AKT Mediated Glucose Metabolism. Front Pharmacol 2021; 11:619426. [PMID: 33584304 PMCID: PMC7873565 DOI: 10.3389/fphar.2020.619426] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
Ischemia-reperfusion (I/R) could cause heart irreversible damage, which is tightly combined with glucose metabolism disorder. It is demonstrated that GLUT4 (glucose transporter 4) translocation is critical for glucose metabolism in the cardiomyocytes under I/R injury. Moreover, DRD4 (dopamine receptor D4) modulate glucose metabolism, and protect neurocytes from anoxia/reoxygenation (A/R) injury. Thus, DRD4 might regulate myocardial I/R injury in association with GLUT4-mediated glucose metabolism. However, the effects and mechanisms are largely unknown. In the present study, the effect of DRD4 in heart I/R injury were studied ex vivo and in vitro. For I/R injury ex vivo, DRD4 agonist (PD168077) was perfused by Langendorff system in the isolated rat heart. DRD4 activated by PD168077 improved cardiac function in the I/R-injured heart as determined by the left ventricular developed pressure (LVDP), +dp/dt, and left ventricular end diastolic pressure (LVEDP), and reduced heart damage evidenced by infarct size, the release of troponin T (TNT) and lactate dehydrogenase (LDH). DRD4 activation diminished I/R injury induced apoptosis and enhanced cell viability impaired by I/R injury in cardiomyocyte, showed by TUNEL staining, flow cytometer and CCK8 assay. Furthermore, DRD4 activation did not change total GULT4 protein expression level but increased the membrane GULT4 localization determined by western blot. In terms of mechanism, DRD4 activation increased pPI3K/p-AKT but not the total PI3K/AKT during anoxia/reoxygenation (A/R) injury in vitro. Interestingly, PI3K inhibitor, Wortmannin, blocked PI3K/AKT pathway and depleted the membrane GULT4, and further promoted apoptosis showed by TUNEL staining, flow cytometer, western blot of cleaved caspase 3, BAX and BCL2 expression. Thus, DRD4 activation exerted a protective effect against I/R injury by promoting GLUT4 translocation depended on PI3K/AKT pathway, which enhanced the ability of glucose uptake, and ultimately reduced the apoptosis in cardiomyocytes.
Collapse
Affiliation(s)
- Xue-Song Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jing Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Yu-Xue Yang
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Chun-Lei Qi
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ting Xiong
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Geng-Ze Wu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Chun-Yu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Da-Xin Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou people's Hospital), Taizhou, China
| |
Collapse
|
8
|
Zhang DS, Liang GY, Liu DX, Yu J, Wang F. Role of Phosphorylated AMP-Activated Protein Kinase (AMPK) in Myocardial Insulin Resistance After Myocardial Ischemia-Reperfusion During Cardiopulmonary Bypass Surgery in Dogs. Med Sci Monit 2019; 25:4149-4158. [PMID: 31160548 PMCID: PMC6561136 DOI: 10.12659/msm.916517] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background The aim of this study was to determine the role of AMP-activated protein kinase (AMPK) in myocardial insulin resistance after myocardial ischemia-reperfusion during cardiopulmonary bypass surgery in dogs. Material/Methods Twenty-four mongrel dogs were randomly assigned to 4 groups. The control group did not undergo aortic cross-clamping; the model group underwent 60 mins of aortic cross-clamping with 150 ml cardioplegic solution. The treatment group, the inhibition group respectively with 0.11mg/kg AICAR (AMPK agonist) in 150 ml cardioplegic solution and 0.11 mg/kg Compound C (AMPK inhibitor) in 150 ml cardioplegic solution. The blood flow was determined and left ventricular myocardial tissue were taken at pre-bypass, 15, 60, and 90 min after aorta declamping, respectively. Expression of AMPK mRNA, p-AMPK and GLUT-4 proteins was determined by RT-PCR, IHC and WB. Results Compared with the control group, receiving 60 min ischemia at 15 min after reperfusion, Myocardial Glucose Extraction Ratio were significantly decreased in the other 3 groups, it was significantly decreased from 20.0% to 1.2% at 60 min of reperfusion, and recovered to 6.1% after 90 min reperfusion in model group, while recovered to 4.1%, 12.0% after 90 min reperfusion respectively exposed to Compound C and AICAR. The expressions of p-AMPK, GLUT-4 protein and AMPK mRNA in myocardium were decreased in different experiment groups, but these changes occurred to a lesser extent in the treatment group. Conclusions The inability of GLUT-4 expression induced by the decreases in p-AMPK protein expression that may be one of the reasons for myocardial insulin resistance.
Collapse
Affiliation(s)
- Deng-Shen Zhang
- Department of Thoracic and Cardiovascular Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China (mainland)
| | - Gui-You Liang
- Department of Thoracic and Cardiovascular Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China (mainland)
| | - Da-Xing Liu
- Department of Thoracic and Cardiovascular Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China (mainland)
| | - Jie Yu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China (mainland)
| | - Feng Wang
- Department of Thoracic and Cardiovascular Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China (mainland)
| |
Collapse
|
9
|
Li T, Xu J, Qin X, Hou Z, Guo Y, Liu Z, Wu J, Zheng H, Zhang X, Gao F. Glucose oxidation positively regulates glucose uptake and improves cardiac function recovery after myocardial reperfusion. Am J Physiol Endocrinol Metab 2017; 313:E577-E585. [PMID: 28325730 DOI: 10.1152/ajpendo.00014.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/10/2017] [Accepted: 03/15/2017] [Indexed: 01/31/2023]
Abstract
Myocardial reperfusion decreases glucose oxidation and uncouples glucose oxidation from glycolysis. Therapies that increase glucose oxidation lessen myocardial ischemia-reperfusion (I/R) injury. However, the regulation of glucose uptake during reperfusion remains poorly understood. We found that glucose uptake was remarkably diminished in the myocardium following reperfusion in Sprague-Dawley rats as detected by 18F-labeled and fluorescent-labeled glucose analogs, even though GLUT1 was upregulated by threefold and GLUT4 translocation remained unchanged compared with those of sham-treated rats. The decreased glucose uptake was accompanied by suppressed glucose oxidation. Interestingly, stimulating glucose oxidation by inhibition of pyruvate dehydrogenase kinase 4 (PDK4), a rate-limiting enzyme for glucose oxidation, increased glucose uptake and alleviated I/R injury. In vitro data in neonatal myocytes showed that PDK4 overexpression decreased glucose uptake, whereas its knockdown increased glucose uptake, suggesting that PDK4 has a role in regulating glucose uptake. Moreover, inhibition of PDK4 increased myocardial glucose uptake with concomitant enhancement of cardiac insulin sensitivity following myocardial I/R. These results showed that the suppressed glucose oxidation mediated by PDK4 contributes to the reduced glucose uptake in the myocardium following reperfusion, and enhancement of glucose uptake exerts cardioprotection. The findings suggest that stimulating glucose oxidation via PDK4 could be an efficient approach to improve recovery from myocardial I/R injury.
Collapse
Affiliation(s)
- Tingting Li
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China; and
| | - Jie Xu
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China; and
| | - Xinghua Qin
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China; and
| | - Zuoxu Hou
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China; and
| | - Yongzheng Guo
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China; and
| | - Zhenhua Liu
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China; and
| | - Jianjiang Wu
- Department of Anesthesiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hong Zheng
- Department of Anesthesiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xing Zhang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China; and
| | - Feng Gao
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China; and
| |
Collapse
|
10
|
Zhong CB, Chen X, Zhou XY, Wang XB. The Role of Peroxisome Proliferator-Activated Receptor γ in Mediating Cardioprotection Against Ischemia/Reperfusion Injury. J Cardiovasc Pharmacol Ther 2017; 23:46-56. [PMID: 28466688 DOI: 10.1177/1074248417707049] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Myocardial infarction (MI) is a serious cardiovascular disease resulting in high rates of morbidity and mortality. Although advances have been made in restoring myocardial perfusion in ischemic areas, decreases in cardiomyocyte death and infarct size are still limited, attributing to myocardial ischemia/reperfusion (I/R) injury. It is necessary to develop therapies to restrict myocardial I/R injury and protect cardiomyocytes against further damage after MI. Many studies have suggested that peroxisome proliferator-activated receptor γ (PPARγ), a ligand-inducible nuclear receptor that predominantly regulates glucose and lipid metabolism, is a promising therapeutic target for ameliorating myocardial I/R injury. Thus, this review focuses on the role of PPARγ in cardioprotection during myocardial I/R. The cardioprotective effects of PPARγ, including attenuating oxidative stress, inhibiting inflammatory responses, improving glucose and lipid metabolism, and antagonizing apoptosis, are described. Additionally, the underlying mechanisms of cardioprotective effects of PPARγ, such as regulating the expression of target genes, influencing other transcription factors, and modulating kinase signaling pathways, are further discussed.
Collapse
Affiliation(s)
- Chong-Bin Zhong
- 1 The Second Clinical Institute of Southern Medical University, Guangzhou, China
| | - Xi Chen
- 1 The Second Clinical Institute of Southern Medical University, Guangzhou, China
| | - Xu-Yue Zhou
- 1 The Second Clinical Institute of Southern Medical University, Guangzhou, China
| | - Xian-Bao Wang
- 2 Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Qiang Y, Liang G, Yu L. Human amniotic mesenchymal stem cells alleviate lung injury induced by ischemia and reperfusion after cardiopulmonary bypass in dogs. J Transl Med 2016; 96:537-46. [PMID: 26927516 DOI: 10.1038/labinvest.2016.37] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 01/06/2016] [Accepted: 01/18/2016] [Indexed: 12/20/2022] Open
Abstract
Transplantation of mesenchymal stem cells may inhibit pathological immune processes contributing to ischemia/reperfusion (I/R) injury. This study aimed to assess the capacity of human amniotic MSC (hAMSCs) to ameliorate I/R injury in a dog model of cardiopulmonary bypass (CPB). Dissociated hAMSCs were cultured ex vivo, and their immunophenotypes were assessed by flow cytometry and immunohistochemistry. A dog model of CPB was established by surgical blockage of the aorta for 1 h. Dogs either underwent mock surgery (Sham group), CPB (model group), or CPB, followed by femoral injection of 2 × 10(7) hAMSCs (n=6). Anti-human nuclei staining revealed hAMSCs in the lungs 3 h after surgery. Oxygen index (OI) and respiratory index (RI) of arterial blood were measured using a biochemical analyzer. Venous blood TNF-α, IL-8, MMP-9, and IL-10 concentrations were measured by ELISA. Pathological changes in the lung were assessed by light microscopy. Third-generation-cultured hAMSCs expressed high levels of CD29, CD44, CD49D, CD73, and CD166 levels, but low CD34 or CD45 amounts and their cytoplasm contained Vimentin. In CPB model animals, OI was elevated and RI reduced; TNF-α, IL-8, and MMP-9 levels were elevated, and IL-10 levels reduced within 3h (P<0.05), but hAMSC transplantation significantly ameliorated these changes (P<0.05). Pathological changes observed in the hAMSC group were significantly less severe than those in the CPB group. In conclusion, hAMSC transplantation can downregulate proinflammatory factors and reduce MMP-9 levels, whereas upregulating the anti-inflammatory molecule IL-10, thus reducing I/R lung injury in a dog model of CPB.
Collapse
Affiliation(s)
- Yong Qiang
- Department of Cardiothoracic Surgery, Nanjing General Hospital of Nanjing Military Command, Nanjing, Jiangsu Province, China
| | - Guiyou Liang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Limei Yu
- Key Laboratory of Cell Engineering in Guizhou Province, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| |
Collapse
|
12
|
Intervention of rosiglitazone on myocardium Glut-4 mRNA expression during ischemia–reperfusion injury in cardio-pulmonary bypass in dogs. Mol Cell Biochem 2012; 373:279-84. [DOI: 10.1007/s11010-012-1501-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 10/25/2012] [Indexed: 11/25/2022]
|
13
|
Vyas AK, Yang KC, Woo D, Tzekov A, Kovacs A, Jay PY, Hruz PW. Exenatide improves glucose homeostasis and prolongs survival in a murine model of dilated cardiomyopathy. PLoS One 2011; 6:e17178. [PMID: 21359201 PMCID: PMC3040766 DOI: 10.1371/journal.pone.0017178] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 01/24/2011] [Indexed: 12/21/2022] Open
Abstract
Background There is growing awareness of secondary insulin resistance and alterations in myocardial glucose utilization in congestive heart failure. Whether therapies that directly target these changes would be beneficial is unclear. We previously demonstrated that acute blockade of the insulin responsive facilitative glucose transporter GLUT4 precipitates acute decompensated heart failure in mice with advanced dilated cardiomyopathy. Our current objective was to determine whether pharmacologic enhancement of insulin sensitivity and myocardial glucose uptake preserves cardiac function and survival in the setting of primary heart failure. Methodology/Principal Findings The GLP-1 agonist exenatide was administered twice daily to a murine model of dilated cardiomyopathy (TG9) starting at 56 days of life. TG9 mice develop congestive heart failure and secondary insulin resistance in a highly predictable manner with death by 12 weeks of age. Glucose homeostasis was assessed by measuring glucose tolerance at 8 and 10 weeks and tissue 2-deoxyglucose uptake at 75 days. Exenatide treatment improved glucose tolerance, myocardial GLUT4 expression and 2-deoxyglucose uptake, cardiac contractility, and survival over control vehicle-treated TG9 mice. Phosphorylation of AMP kinase and AKT was also increased in exenatide-treated animals. Total myocardial GLUT1 levels were not different between groups. Exenatide also abrogated the detrimental effect of the GLUT4 antagonist ritonavir on survival in TG9 mice. Conclusion/Significance In heart failure secondary insulin resistance is maladaptive and myocardial glucose uptake is suboptimal. An incretin-based therapy, which addresses these changes, appears beneficial.
Collapse
Affiliation(s)
- Arpita Kalla Vyas
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kai-Chien Yang
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Dennis Woo
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Anatoly Tzekov
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Attila Kovacs
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Patrick Y. Jay
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Paul W. Hruz
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
14
|
Liu KX, Li C, Li YS, Yuan BL, Xu M, Xia Z, Huang WQ. Proteomic analysis of intestinal ischemia/reperfusion injury and ischemic preconditioning in rats reveals the protective role of aldose reductase. Proteomics 2010; 10:4463-75. [PMID: 21136599 DOI: 10.1002/pmic.201000078] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a critical condition associated with high morbidity and mortality. Studies show that ischemic preconditioning (IPC) can protect the intestine from I/R injury. However, the underlying molecular mechanisms of this event have not been fully elucidated. In the present study, 2-DE combined with MALDI-MS was employed to analyze intestinal mucosa proteomes of rat subjected to I/R injury in the absence or presence of IPC pretreatment. The protein content of 16 proteins in the intestinal mucosa changed more than 1.5-fold following intestinal I/R. These proteins were, respectively, involved in the cellular processes of energy metabolism, anti-oxidation and anti-apoptosis. One of these proteins, aldose reductase (AR), removes reactive oxygen species. In support of the 2-DE results, the mRNA and protein expressions of AR were significantly downregulated upon I/R injury and enhanced by IPC as confirmed by RT-PCR and western blot analysis. Further study showed that AR-selective inhibitor epalrestat totally turned over the protective effect of IPC, indicating that IPC confers protection against intestinal I/R injury primarily by increasing intestinal AR expression. The finding that AR may play a key in intestinal ischemic protection might offer evidences to foster the development of new therapies against intestinal I/R injury.
Collapse
Affiliation(s)
- Ke-Xuan Liu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China.
| | | | | | | | | | | | | |
Collapse
|
15
|
Li YS, Wang ZX, Li C, Xu M, Li Y, Huang WQ, Xia Z, Liu KX. Proteomics of ischemia/reperfusion injury in rat intestine with and without ischemic postconditioning. J Surg Res 2009; 164:e173-80. [PMID: 20189595 DOI: 10.1016/j.jss.2009.10.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 09/11/2009] [Accepted: 10/01/2009] [Indexed: 12/11/2022]
Abstract
BACKGROUND Intestinal ischemia/reperfusion (I/R) injury is a critical condition associated with high morbidity and mortality. Our previous study showed that ischemic postconditioning (IPo) protects the intestinal mucosa from I/R injury. However, the precise molecular mechanisms of this event remain poorly elucidated. The aim of this study was to investigate the differentially expressed proteins of intestinal mucosa after intestinal I/R with or without IPo, and to explore the potential mechanisms of intestinal I/R injury and the protective effect of IPo in relation to the differential proteins. MATERIALS AND METHODS Intestinal I/R injury was established by occluding the superior mesenteric artery (SMA) for 60 min followed by 60 min reperfusion. The rats were randomly allocated into one of three groups based upon the intervention (n = 8); sham : sham surgical preparation including isolation of the SMA without occlusion was performed; injury: there was no intervention either before or after SMA occlusion; IPo: three cycles of 30 s reperfusion-30 s reocclusion were imposed immediately upon reperfusion. A comparative proteomics approach with two-dimensional gel electrophoresis was used to isolate proteins in intestinal mucosa, the expression of which were regulated by I/R injury post-treated with or without IPo. The differentially displayed proteins were identified through matrix-assisted laser desorption/ionization-time of flight-mass spectrometry (MALDI-TOF-MS). RESULTS Image analysis revealed that an average of 1300 protein spots were detected on each gel; 16 and 9 proteins showing more than 1.5-fold difference were identified between the Sham versus Injury group and injury group versus IPo group, respectively. The identified proteins were functionally involved in the cellular processes of energy metabolism, anti-oxidation, and anti-apoptosis. CONCLUSIONS This study provided new clues for understanding the mechanisms of IPo against intestinal I/R injury.
Collapse
Affiliation(s)
- Yun-Sheng Li
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Xu C, Zhang X, Yu C, Lu G, Chen S, Xu L, Ding W, Shi Q, Li Y. Proteomic analysis of hepatic ischemia/reperfusion injury and ischemic preconditioning in mice revealed the protective role of ATP5beta. Proteomics 2009; 9:409-419. [PMID: 19142948 DOI: 10.1002/pmic.200800393] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Indexed: 12/13/2022]
Abstract
Hepatic ischemia/reperfusion (I/R) injury is an inevitable consequence during liver surgery. Ischemic preconditioning (IPC) has been shown to protect the livers from I/R injury, partially mediated by preservation of hepatic ATP contents. However, the precise molecular mechanisms of these events remain poorly elucidated. In this study, liver proteomes of the mice subjected to I/R injury pretreated with or without IPC were analyzed using 2-DE combined with MALDI-TOF/TOF mass analysis. Twenty proteins showing more than 1.5-fold difference were identified in the livers upon I/R injury. Among these proteins, four proteins were further regulated by IPC when compared with nonpretreated controls. One of these proteins, ATP synthase beta subunit (ATP5beta) catalyzes the rate-limiting step of ATP formation. The expression level of ATP5beta, which was further validated by Western blot analysis, was significantly decreased upon I/R injury while turned over by IPC pretreatment. Change pattern of hepatic ATP corresponded with that of ATP5beta expression, indicating that increasing hepatic ATP5beta expression might be a reason for ATP-preserving effect of IPC. In summary, this study provided new clues for understanding the mechanisms of IPC against I/R injury. The protective role of ATP5beta might give evidences for developing new therapeutic approaches against hepatic I/R injury.
Collapse
Affiliation(s)
- Chengfu Xu
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|