1
|
Fu Y, Wang Q, Wang D, Li Y. Dexmedetomidine Inhibits Ferroptosis to Alleviate Hypoxia/Reoxygenation-Induced Cardiomyocyte Injury by Regulating the HDAC2/FPN Pathway. Cardiovasc Drugs Ther 2025:10.1007/s10557-024-07664-z. [PMID: 39747742 DOI: 10.1007/s10557-024-07664-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE Myocardial ischemia/reperfusion injury (MIRI) is closely associated with ferroptosis. Dexmedetomidine (Dex) has good therapeutic effects on MIRI. This study investigates whether dexmedetomidine (Dex) regulates ferroptosis during MIRI by affecting ferroportin1 (FPN) levels and elucidates the underlying mechanisms. METHODS A murine MIRI model was established using male C57BL/6 J mice subjected to 30 min of left anterior descending coronary artery ligation followed by 48 h of reperfusion. In vitro, cardiomyocyte hypoxia/reoxygenation (H/R) models were created with 16 h of hypoxia and 8 h of reoxygenation. Triphenyltetrazolium chloride (TTC) staining was employed to determine infarct size. The pathological changes in myocardial tissues were assessed using hematoxylin-eosin (HE) staining. Lipid reactive oxygen species (ROS) level was detected using BODIPY™ 581/591 C11, and ferrous iron (Fe2+) and malondialdehyde (MDA) levels were measured using the kits. Cardiomyocyte viability was examined using cell counting kit-8 (CCK8) assay. The histone H3 lysine 27 acetylation (H3K27Ac) level in the FPN promoter region was determined using DNA pulldown assay. Chromatin immunoprecipitation (ChIP) assay was used to investigate the relationship between histone deacetylase 2 (HDAC2) and FPN promoter. RESULTS Dex alleviated ferroptosis in cardiomyocytes by upregulating FPN levels, which mitigated H/R-induced oxidative damage. FPN knockdown abolished the protective effects of Dex, confirming its dependence on FPN expression. Additionally, HDAC2 knockdown alleviated I/R-induced myocardial injury and ferroptosis in mice. Moreover, H/R-induced HDAC2 upregulation transcriptionally inhibited FPN expression by reducing the H3K27Ac level in the FPN promoter region, but Dex therapy restored this impact via inhibition of HDAC2. As expected, HDAC2 overexpression partially reversed the inhibitory effect of Dex on H/R-mediated cardiomyocyte ferroptosis. CONCLUSION Dex alleviated H/R-mediated cardiomyocyte ferroptosis through regulating the HDAC2/FPN axis. Our findings lend theoretical support to the use of Dex in MIRI therapy.
Collapse
Affiliation(s)
- Yueqi Fu
- Jiamusi University, Jiamusi, 154002, Heilongjiang Province, China
| | - QingDong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Jiamusi University, No.348 Dexiang Street, Xiangyang District, Jiamusi, 154002, Heilongjiang Province, China
| | - DongWei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Jiamusi University, No.348 Dexiang Street, Xiangyang District, Jiamusi, 154002, Heilongjiang Province, China.
| | - Yicong Li
- Department of Anesthesiology, Hainan Hosiptal of Chinese PLA General Hospital, No.80 Jianglin Street, Haitang District, Sanya City, Hainan Province, China.
| |
Collapse
|
2
|
Yang FF, Liu JJ, Xu XL, Hu T, Liu JQ, He ZX, Zhao GY, Wei B, Ma LY. Discovery of Novel Imidazo[1,2- a]pyridine-Based HDAC6 Inhibitors as an Anticarcinogen with a Cardioprotective Effect. J Med Chem 2024; 67:14345-14369. [PMID: 39102466 DOI: 10.1021/acs.jmedchem.4c01168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Cardiotoxicity associated with chemotherapy has gradually become the major cause of death in cancer patients. The development of bifunctional drugs with both cardioprotective and antitumor effects has become the future direction. HDAC6 plays important roles in the progression, treatment, and prognosis of cancer and cardiovascular diseases, but bifunctional inhibitors have not been reported. Herein, structure-activity relationship studies driven by pharmacophore-based remodification and fragment-based design were performed to yield highly potent HDAC6 inhibitor I-c4 containing imidazo[1,2-a]pyridine. Importantly, I-c4 effectively suppressed the growth of MGC-803 xenografts in vitro and in vivo by inhibiting the deacetylation pathway without causing myocardial damage after long-term administration. Meanwhile, I-c4 could mitigate severe myocardial damage against H2O2 or myocardial ischemia/reperfusion in vitro and in vivo. Further studies revealed that the cardioprotective effect of I-c4 was associated with reduction of inflammatory cytokines. Taken together, I-c4 may represent a novel lead compound for further development of an anticarcinogen with a cardioprotective effect.
Collapse
Affiliation(s)
- Fei-Fei Yang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Jing-Jing Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Xue-Li Xu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Ting Hu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Jian-Quan Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Zhang-Xu He
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Guang-Yuan Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Bo Wei
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Li-Ying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
- China Meheco Topfond Pharmaceutical Co.; Key Laboratory of Cardio-cerebrovascular Drug, Zhumadian 463000, China
| |
Collapse
|
3
|
Li X, Cheng S, Yu C, Li Y, Cao X, Wang Y, Zhang Z, Huang J. Co-delivery of retinoic acid and miRNA by functional Au nanoparticles for improved survival and CT imaging tracking of MSCs in pulmonary fibrosis therapy. Asian J Pharm Sci 2024; 19:100944. [PMID: 39660166 PMCID: PMC11630633 DOI: 10.1016/j.ajps.2024.100944] [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: 12/29/2023] [Revised: 05/09/2024] [Accepted: 05/18/2024] [Indexed: 12/12/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as promising candidates for idiopathic pulmonary fibrosis (IPF) therapy. Increasing the MSC survival rate and deepening the understanding of the behavior of transplanted MSCs are of great significance for improving the efficacy of MSC-based IPF treatment. Therefore, dual-functional Au-based nanoparticles (Au@PEG@PEI@TAT NPs, AuPPT) were fabricated by sequential modification of cationic polymer polyetherimide (PEI), polyethylene glycol (PEG), and transactivator of transcription (TAT) penetration peptide on AuNPs, to co-deliver retinoic acid (RA) and microRNA (miRNA) for simultaneously enhancing MSC survive and real-time imaging tracking of MSCs during IPF treatment. AuPPT NPs, with good drug loading and cellular uptake abilities, could efficiently deliver miRNA and RA to protect MSCs from reactive oxygen species and reduce their expression of apoptosis executive protein Caspase 3, thus prolonging the survival time of MSC after transplantation. In the meantime, the intracellular accumulation of AuPPT NPs enhanced the computed tomography imaging contrast of transplanted MSCs, allowing them to be visually tracked in vivo. This study establishes an Au-based dual-functional platform for drug delivery and cell imaging tracking, which provides a new strategy for MSC-related IPF therapy.
Collapse
Affiliation(s)
- Xiaodi Li
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Shengnan Cheng
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Chenggong Yu
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yuxuan Li
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Xiaoling Cao
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Yuhan Wang
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhijun Zhang
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jie Huang
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
4
|
Li R, Zhang K, Xu Z, Yu Y, Wang D, Li K, Liu W, Pan J. Liraglutide ameliorates TAC-induced cardiac hypertrophy and heart failure by upregulating expression level of ANP expression. Heliyon 2024; 10:e32229. [PMID: 38868006 PMCID: PMC11168427 DOI: 10.1016/j.heliyon.2024.e32229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 06/14/2024] Open
Abstract
Recent studies have underscored the cardioprotective properties of liraglutide. This research explores its impact on cardiac hypertrophy and heart failure following transverse aortic constriction (TAC). We found that liraglutide administration markedly ameliorated cardiac hypertrophy, fibrosis, and function. These benefits correlated with increased ANP expression and reduced activity in the calcineurin A/NFATc3 signaling pathway. Moreover, liraglutide mitigated ER stress and cardiomyocyte apoptosis, and enhanced autophagy. Notably, the positive effects of liraglutide diminished when co-administered with A71915, an ANP inhibitor, suggesting that ANP upregulation is critical to its cardioprotective mechanism.
Collapse
Affiliation(s)
- Ruisha Li
- Institute of Cardiothoracic Vascular Disease, Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Keyin Zhang
- Institute of Cardiothoracic Vascular Disease, Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhenjun Xu
- Institute of Cardiothoracic Vascular Disease, Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yanrong Yu
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Dongjin Wang
- Institute of Cardiothoracic Vascular Disease, Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Kai Li
- Institute of Cardiothoracic Vascular Disease, Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenxue Liu
- Institute of Cardiothoracic Vascular Disease, Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jun Pan
- Institute of Cardiothoracic Vascular Disease, Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Wang Z, Lin D, Cui B, Zhang D, Wu J, Ma J. Melatonin protects against myocardial ischemia-reperfusion injury by inhibiting excessive mitophagy through the Apelin/SIRT3 signaling axis. Eur J Pharmacol 2024; 963:176292. [PMID: 38128867 DOI: 10.1016/j.ejphar.2023.176292] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Excessive or uncontrolled mitophagy may result in a drastic shortage of healthy mitochondrial for ATP supply after reperfusion, leading to irreversible myocardial damage. Melatonin, a hormone produced by the pineal gland, has been proven to ameliorate myocardial ischemia-reperfusion (I/R) injury via regulating mitophagy. However, its underlying mechanism has not been fully elucidated. The present study focused on the role of mitophagy in the cardioprotective effects of melatonin by using the myocardial I/R rat model. The rats were pretreated with or without the apelin inhibitor ML221, the sirtuin 3 (SIRT3) inhibitor 3-TYP and then subjected to I/R injury, with melatonin administrated 10 min before reperfusion. The effects of melatonin on myocardial infarct size, biomarkers of myocardial injury, oxidative stress, and mitochondrial function were detected, and the expression of apelin, SIRT3, and mitophagy-related proteins were also measured. Excessive mitophagy was activated after I/R injury and was correlated with oxidative stress and mitochondrial dysfunction. Melatonin pretreatment ameliorated myocardial injury by decreasing oxidative stress, restoring mitochondrial function, and inhibiting excessive mitophagy. However, ML221 or 3-TYP disrupted these beneficial effects of melatonin on I/R injury. Taken together, these results suggest that melatonin pretreatment ameliorates myocardial I/R injury through regulating the apelin/SIRT3 pathway to inhibit excessive mitophagy.
Collapse
Affiliation(s)
- Zhaoqi Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Duomao Lin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Boqun Cui
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Dongni Zhang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jinjing Wu
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jun Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
6
|
Naseroleslami M, Sharifi M, Rakhshan K, Mokhtari B, Aboutaleb N. Nesfatin-1 attenuates injury in a rat model of myocardial infarction by targeting autophagy, inflammation, and apoptosis. Arch Physiol Biochem 2023; 129:122-130. [PMID: 32762481 DOI: 10.1080/13813455.2020.1802486] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nesfatin-1 plays an important role in the modulation of heart performance. However, it remains unclear how nesfatin-1 contributes to cell survival in acute myocardial infarction (MI). A rat model of MI was established via ligation of left anterior descending coronary artery (LAD) for 30 min and 20 µg/kg concentration of nesfatin-1 was intraperitoneally infused prior to reperfusion. At 24 h after reperfusion, oxidative stress markers, the expression of caspase3, beclin-1, pro-inflammatory cytokines, and the mRNA levels of Bax and Bcl-2 were evaluated. Results showed that nesfatin-1 markedly restored GSH content and SOD activity as well as reduced MDA levels compared to only the MI group (p < .05). Likewise, nesfatin-1 contributed to cell survival by inhibiting autophagy and apoptosis markers such as caspase3 and Bax (p < .05). Collectively, these findings support the idea that nasfatin-1 can be used as a good candidate to treat MI by targeting oxidative stress, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Maryam Naseroleslami
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Masuomeh Sharifi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kamran Rakhshan
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnaz Mokhtari
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Aboutaleb
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Xu C, Liu Y, Yang J, Zhai M, Fan Z, Qiao R, Jin P, Yang L. Effects of berbamine against myocardial ischemia/reperfusion injury: Activation of the 5' adenosine monophosphate-activated protein kinase/nuclear factor erythroid 2-related factor pathway and changes in the mitochondrial state. Biofactors 2022; 48:651-664. [PMID: 35129229 PMCID: PMC9305777 DOI: 10.1002/biof.1820] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/10/2021] [Indexed: 12/13/2022]
Abstract
This study was designed to investigate whether berbamine (BA)-induced cardioprotective effects were related to 5' adenosine monophosphate-activated protein kinase (AMPK)/nuclear factor erythroid 2-related factor (Nrf2) signaling and changes in the mitochondria in myocardial ischemia/reperfusion (I/R) injury. C57/BL6 mice were exposed to BA (10 mg/kg/d), with or without administration of the AMPK specific inhibitor compound C (5 mg/kg/d) or the Nrf2 specific inhibitor ML-385 (30 mg/kg/d), and then subjected to a myocardial I/R operation. As expected, BA significantly improved post-ischemic cardiac function, reduced infarct size and apoptotic cell death, decreased oxidative stress, and improved the mitochondrial state. Furthermore, BA markedly increased AMPK activation, Nrf2 nuclear translocation, and the levels of NAD(P)H quinone dehydrogenase and heme oxygenase-1. Nevertheless, these BA-induced changes were abrogated by compound C. In addition, ML-385 also canceled the cardioprotective effects of BA but had little effect on AMPK activation. Our results demonstrate that BA alleviates myocardial I/R injury and the mitochondrial state by inhibiting apoptosis and oxidative stress via the AMPK/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Chennian Xu
- Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangChina
- Department of Cardiovascular SurgeryXijing Hospital, Air Force Medical UniversityXi'anChina
| | - Yang Liu
- Department of Cardiovascular SurgeryXijing Hospital, Air Force Medical UniversityXi'anChina
| | - Jian Yang
- Department of Cardiovascular SurgeryXijing Hospital, Air Force Medical UniversityXi'anChina
| | - Mengen Zhai
- Department of Cardiovascular SurgeryXijing Hospital, Air Force Medical UniversityXi'anChina
| | - Zhenge Fan
- Department of Cardiovascular SurgeryXijing Hospital, Air Force Medical UniversityXi'anChina
| | - Rui Qiao
- Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangChina
| | - Ping Jin
- Department of Cardiovascular SurgeryXijing Hospital, Air Force Medical UniversityXi'anChina
| | - Lifang Yang
- Department of AnesthesiologyXi'an Children's HospitalXi'anChina
| |
Collapse
|
8
|
Cox-2 Antagonizes the Protective Effect of Sevoflurane on Hypoxia/Reoxygenation-Induced Cardiomyocyte Apoptosis through Inhibiting the Akt Pathway. DISEASE MARKERS 2021; 2021:4114593. [PMID: 34917200 PMCID: PMC8670977 DOI: 10.1155/2021/4114593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/16/2021] [Indexed: 11/23/2022]
Abstract
Objective To uncover the protective role of sevoflurane on hypoxia/reoxygenation-induced cardiomyocyte apoptosis through the protein kinase B (Akt) pathway. Methods An in vitro hypoxia/reoxygenation (H/R) model was established in cardiomyocyte cell line H9c2. Sevoflurane (SEV) was administrated in H9c2 cells during the reoxygenation period. Viability, layered double hydroxide (LDH) release, and apoptosis in H9c2 cells were determined to assess H/R-induced cell damage. Relative levels of apoptosis-associated genes were examined. Moreover, phosphorylation of Akt was determined. Results H/R injury declined viability and enhanced LDH release and apoptotic rate in H9c2 cells. Cyclooxygenase-2 (Cox-2) was upregulated following H/R injury, which was partially reversed by SEV treatment. In addition, SEV treatment reversed changes in viability and LDH release owing to H/R injury in H9c2 cells, which were further aggravated by overexpression of Cox-2. The Akt pathway was inhibited in H9c2 cells overexpressing Cox-2. Conclusions Sevoflurane protects cardiomyocyte damage following H/R via the Akt pathway, and its protective effect was abolished by overexpression of Cox-2.
Collapse
|
9
|
Chen X, Xie Q, Zhu Y, Xu J, Lin G, Liu S, Su Z, Lai X, Li Q, Xie J, Yang X. Cardio-protective effect of tetrahydrocurcumin, the primary hydrogenated metabolite of curcumin in vivo and in vitro: Induction of apoptosis and autophagy via PI3K/AKT/mTOR pathways. Eur J Pharmacol 2021; 911:174495. [PMID: 34555398 DOI: 10.1016/j.ejphar.2021.174495] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/14/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022]
Abstract
Tetrahydrocurcumin (THC) is an essential metabolite of curcumin, a major active component of the Curcuma species, which have been used traditionally for the treatment of cardiovascular diseases. The PI3K/AKT/mTOR signaling pathways serve a vital role during myocardial ischemia-reperfusion (MI/R) injury. The aim of the present study was to investigate the cardioprotective potential and mechanism of THC. In the in vivo study, an animal model of MI/R was induced by coronary occlusion. Results indicated that THC (50 mg/kg/day) protected the rat hearts from MI/R-induced heart failure by increasing ejection fraction (EF) and fractional shortening (FS) and decreasing left ventricular end systolic diameter (LVESD) and left ventricular end systolic volume (LVESV). THC also reduced myocardial infarct size and apoptosis. Furthermore, H9c2 cells were incubated with THC (20 μM) to explore its potential effect following exposure to hypoxia and reoxygenation (H/R). THC post-treatment significantly augmented cell viability and prevented lactate dehydrogenase (LDH) release after H/R exposure. THC effectively improved antioxidant activity by increasing SOD and CAT activities and decreasing MDA level. THC also enhanced mitochondrial membrane potential, inhibited apoptotic cell death, diminished the Bax/Bcl-2 ratio and cleaved caspase-3 level relative to the H/R model. In addition, THC effectively decreased Beclin1 expression and LC3 II/LC3 I ratio, but increased p62 expression, compared with the H/R model group, and decreased the formation of H/R-induced autophagosomes and autolysosomes. Furthermore, THC promoted the phosphorylation of PI3K/AKT/mTOR and induced the expression of hypoxia-inducible factor 1α (HIF-1α) after H/R. However, these effects on H9c2 cells were notably abolished by the PI3K inhibitor LY294002 and mTOR inhibitor rapamycin. In conclusion, THC effectively inhibited H/R-induced autophagy and apoptosis via, at least partially, activating the PI3K/AKT/mTOR pathways. THC might have the potential to be further developed into a potential candidate for the treatment of MI/R injury.
Collapse
Affiliation(s)
- Xiaoying Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Qingfeng Xie
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Ying Zhu
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, PR China
| | - Jiamin Xu
- The Second School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Guoshu Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Shujun Liu
- The Second School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China; Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, PR China
| | - Xiaoping Lai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China; Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, PR China.
| | - Qian Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China
| | - Xiaobo Yang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China.
| |
Collapse
|
10
|
Han RH, Huang HM, Han H, Chen H, Zeng F, Xie X, Liu DY, Cai Y, Zhang LQ, Liu X, Xia ZY, Tang J. Propofol postconditioning ameliorates hypoxia/reoxygenation induced H9c2 cell apoptosis and autophagy via upregulating forkhead transcription factors under hyperglycemia. Mil Med Res 2021; 8:58. [PMID: 34753510 PMCID: PMC8579603 DOI: 10.1186/s40779-021-00353-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 10/26/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Administration of propofol, an intravenous anesthetic with antioxidant property, immediately at the onset of post-ischemic reperfusion (propofol postconditioning, P-PostC) has been shown to confer cardioprotection against ischemia-reperfusion injury, while the underlying mechanism remains incompletely understood. The FoxO transcription factors are reported to play critical roles in activating cardiomyocyte survival signaling throughout the process of cellular injuries induced by oxidative stress and are also involved in hypoxic postconditioning mediated neuroprotection, however, the role of FoxO in postconditioning mediated protection in the heart and in particular in high glucose condition is unknown. METHODS Rat heart-derived H9c2 cells were exposed to high glucose (HG) for 48 h (h), then subjected to hypoxia/reoxygenation (H/R, composed of 8 h of hypoxia followed by 12 h of reoxygenation) in the absence or presence of postconditioning with various concentrations of propofol (P-PostC) at the onset of reoxygenation. After having identified the optical concentration of propofol, H9c2 cells were subjected to H/R and P-PostC in the absence or presence of FoxO1 or FoxO3a gene silencing to explore their roles in P-PostC mediated protection against apoptotic and autophagic cell deaths under hyperglycemia. RESULTS The results showed that HG with or without H/R decreased cell viability, increased lactate dehydrogenase (LDH) leakage and the production of reactive oxygen species (ROS) in H9c2 cells, all of which were significantly reversed by propofol (P-PostC), especially at the concentration of 25 µmol/L (P25) (all P < 0.05, NC vs. HG; HG vs. HG + HR; HG + HR + P12.5 or HG + HR + P25 or HG + HR + P50 vs. HG + HR). Moreover, we found that propofol (P25) decreased H9c2 cells apoptosis and autophagy that were concomitant with increased FoxO1 and FoxO3a expression (all P < 0.05, HG + HR + P25 vs. HG + HR). The protective effects of propofol (P25) against H/R injury were reversed by silencing FoxO1 or FoxO3a (all P < 0.05, HG + HR + P25 vs. HG + HR + P25 + siRNA-1 or HG + HR + P25 + siRNA-5). CONCLUSION It is concluded that propofol postconditioning attenuated H9c2 cardiac cells apoptosis and autophagy induced by H/R injury through upregulating FoxO1 and FoxO3a under hyperglycemia.
Collapse
Affiliation(s)
- Rong-Hui Han
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - He-Meng Huang
- Department of Emergency, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Hong Han
- Department of Anesthesiology, the Eighth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 518000, China
| | - Hao Chen
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - Fei Zeng
- Department of Anesthesiology, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, 510000, China
| | - Xiang Xie
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Dan-Yong Liu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - Yin Cai
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China.,Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, China
| | - Liang-Qing Zhang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - Xin Liu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - Zheng-Yuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China. .,State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Pok Fu Lam, 999077, Hong Kong SAR, China.
| | - Jing Tang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China.
| |
Collapse
|
11
|
Ren J, Wang X, Zhang Y. Editorial: New Drug Targets for Proteotoxicity in Cardiometabolic Diseases. Front Physiol 2021; 12:745296. [PMID: 34603089 PMCID: PMC8481817 DOI: 10.3389/fphys.2021.745296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/20/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Xin Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Yingmei Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Xie M, Cho GW, Kong Y, Li DL, Altamirano F, Luo X, Morales CR, Jiang N, Schiattarella GG, May HI, Medina J, Shelton J, Ferdous A, Gillette TG, Hill JA. Activation of Autophagic Flux Blunts Cardiac Ischemia/Reperfusion Injury. Circ Res 2021; 129:435-450. [PMID: 34111934 PMCID: PMC8317428 DOI: 10.1161/circresaha.120.318601] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/09/2021] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Min Xie
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Geoffrey W. Cho
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Yongli Kong
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Dan L. Li
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Francisco Altamirano
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Xiang Luo
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Cyndi R. Morales
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Nan Jiang
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Gabriele G. Schiattarella
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Herman I. May
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Jessica Medina
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - John Shelton
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Anwarul Ferdous
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Thomas G. Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Joseph A. Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| |
Collapse
|
13
|
Selective striatal cell loss is ameliorated by regulated autophagy of the cortex. Life Sci 2021; 282:119822. [PMID: 34271058 DOI: 10.1016/j.lfs.2021.119822] [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: 12/16/2020] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 11/24/2022]
Abstract
AIMS The harmful cellular environment leads to brain damage, and each brain subregion exhibits a differential vulnerability to its effects. This study investigated the causes of selectively striatal cell loss in systemic 3-nitropropionic acid (3-NP) infused mice. MAIN METHODS This study was performed in the neuronal cell line, primary neuron, cultured mouse brain, and mice brain tissues. The 3-NP solution was delivered using an osmotic mini-pump system for 7 days. ROS in brain tissue were detected and evaluated with the signals of CM-H2DCFDA for total cellular ROS and MitoSOX Red for mitochondrial ROS. Cellular ROS and the functional status of mitochondria were assessed with a detection kit and analyzed using flow cytometry. To quantify oxidative damaged DNA, apurinic/apyrimidinic (AP) site numbers in DNA were measured. The protein expression level was assessed using Western blotting, and immunohistochemistry was performed. Cleaved caspase-3 activities were measured by using an enzyme-linked immunosorbent assay (ELISA) kit. KEY FINDINGS By 3-NP, mitochondrial dysfunction was higher in the striatum than in the cortex, and mitochondria-derived ROS levels were higher in the striatum than in the cortex. However, autophagy that may restore the energy depletion resulting from mitochondrial dysfunction occurred comparably less in the striatum than in the cortex. Inhibition of ASK1 by NQDI1 regulates MAPK signaling, apoptosis, and autophagy. Regulated autophagy of the cortex improved non-cell autonomously striatal damaged condition. SIGNIFICANCE This study illustrated that the different vulnerabilities of the brain subregions, striatum or cortex, against 3-NP are rooted in different mitochondria-derived ROS amounts and autophagic capacity.
Collapse
|
14
|
Xu C, Wang J, Fan Z, Zhang S, Qiao R, Liu Y, Yang J, Yang L, Wang H. Cardioprotective effects of melatonin against myocardial ischaemia/reperfusion injury: Activation of AMPK/Nrf2 pathway. J Cell Mol Med 2021; 25:6455-6459. [PMID: 34128312 PMCID: PMC8406481 DOI: 10.1111/jcmm.16691] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 01/03/2023] Open
Abstract
Although reperfusion is the most effective therapy for patients with acute myocardial infarction, reperfusion injury limits the therapeutic effects of early reperfusion. Oxidative stress plays a crucial role in myocardial ischaemia/reperfusion (I/R) injury. Melatonin, a circulating hormone, is well-known as an antioxidant in cardiovascular diseases. In this short communication, we show that melatonin significantly improves post-ischaemic cardiac function, reduces infarct size and decreases oxidative stress. Furthermore, melatonin markedly increases AMPK activation and Nrf2 nuclear translocation. Nevertheless, these melatonin-induced changes are abrogated by compound C. In addition, ML-385, an Nrf2 inhibitor, also withdraws the antioxidative effects of melatonin but has little effect on AMPK activation. In conclusion, our results demonstrate that melatonin alleviates myocardial I/R injury by inhibiting oxidative stress via the AMPK/Nrf2 signalling pathway.
Collapse
Affiliation(s)
- Chennian Xu
- Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangChina
- Department of UltrasoundChinese PLA 985 HospitalTaiyuanChina
| | - Jian Wang
- Department of UltrasoundChinese PLA 985 HospitalTaiyuanChina
| | - Zhenge Fan
- Department of Cardiovascular SurgeryXijing HospitalAir Force Medical UniversityXi'anChina
| | - Shuang Zhang
- Department of AnesthesiologyXi'an Children's HospitalXi'anChina
| | - Rui Qiao
- Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangChina
| | - Yu Liu
- Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangChina
| | - Jian Yang
- Department of Cardiovascular SurgeryXijing HospitalAir Force Medical UniversityXi'anChina
| | - Lifang Yang
- Department of AnesthesiologyXi'an Children's HospitalXi'anChina
| | - Huishan Wang
- Department of Cardiovascular SurgeryGeneral Hospital of Northern Theater CommandShenyangChina
| |
Collapse
|
15
|
Tian H, Liu S, Ren J, Lee JKW, Wang R, Chen P. Role of Histone Deacetylases in Skeletal Muscle Physiology and Systemic Energy Homeostasis: Implications for Metabolic Diseases and Therapy. Front Physiol 2020; 11:949. [PMID: 32848876 PMCID: PMC7431662 DOI: 10.3389/fphys.2020.00949] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle is the largest metabolic organ in the human body and is able to rapidly adapt to drastic changes during exercise. Histone acetyltransferases (HATs) and histone deacetylases (HDACs), which target histone and non-histone proteins, are two major enzyme families that control the biological process of histone acetylation and deacetylation. Balance between these two enzymes serves as an essential element for gene expression and metabolic and physiological function. Genetic KO/TG murine models reveal that HDACs possess pivotal roles in maintaining skeletal muscles' metabolic homeostasis, regulating skeletal muscles motor adaptation and exercise capacity. HDACs may be involved in mitochondrial remodeling, insulin sensitivity regulation, turn on/off of metabolic fuel switching and orchestrating physiological homeostasis of skeletal muscles from the process of myogenesis. Moreover, many myogenic factors and metabolic factors are modulated by HDACs. HDACs are considered as therapeutic targets in clinical research for treatment of cancer, inflammation, and neurological and metabolic-related diseases. This review will focus on physiological function of HDACs in skeletal muscles and provide new ideas for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Haili Tian
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Sujuan Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jason Kai Wei Lee
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Global Asia Institute, National University of Singapore, Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore, Singapore
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
16
|
Zhang J, Zhang X. Ischaemic preconditioning-induced serum exosomes protect against myocardial ischaemia/reperfusion injury in rats by activating the PI3K/AKT signalling pathway. Cell Biochem Funct 2020; 39:287-295. [PMID: 32767595 DOI: 10.1002/cbf.3578] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/04/2020] [Accepted: 06/22/2020] [Indexed: 12/27/2022]
Abstract
Ischaemia/reperfusion (I/R) injury can lead to severe arrhythmia and aggravate myocardial damage. Exosomes are small-membrane vesicles that play a protective role in myocardial I/R injury. This study aimed to explore the protective effects of ischaemic preconditioning (IPC)-induced serum exosomes (IPC-Exo) on myocardial I/R injury in rats and its underlying mechanism. Serum exosomes were extracted from IPC rats and quantified using a bicinchoninic acid assay kit. IPC-Exo (50 μg) was injected into the infarcted myocardium immediately after ligation. Rats were randomly divided into Sham, I/R, IPC-Exo + I/R, I/R + LY294002, and I/R + IPC-Exo + LY294002 groups. Haemodynamic parameters were measured by physiological recording. Transthoracic echocardiography was used to detect cardiac function. The serum levels of creatine kinase isomer-MB, lactate dehydrogenase, aspartate transaminase, tumour necrosis factor-alpha, interleukin (IL)-1β, and IL-10 were detected by enzyme-linked immunosorbent assay. Triphenyl tetrazolium chloride staining was used to measure the myocardial infarct size. Apoptosis in myocardial tissues was detected by TUNEL staining. Western blotting was used to detect the levels of PI3K/AKT and apoptosis-related proteins. Our results showed that treatment with IPC-Exo ameliorated cardiac function and reduced inflammatory factor production, cardiomyocyte apoptosis, and myocardial infarct size. Moreover, IPC-Exo treatment promoted the protein expression of Bcl-2, p-PI3K, and p-AKT but inhibited that of caspase-3 and Bax. However, treatment with LY294002 significantly reversed that IPC-Exo-induced increase in p-PI3K and p-AKT levels, improvement of haemodynamics, and decrease of inflammatory factor production and apoptosis in the I/R + IPC-Exo group. Taken together, our results suggest that IPC-Exo may alleviate I/R injury via activating the PI3K/AKT signalling pathway.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Internal Medicine II, Shandong Police Officer General Hospital, Jinan City, China
| | - Xijiang Zhang
- Department of Emergency, Zhangqiu District People's Hospital, Jinan City, China
| |
Collapse
|
17
|
Yang M, Zhang Y, Ren J. Acetylation in cardiovascular diseases: Molecular mechanisms and clinical implications. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165836. [PMID: 32413386 DOI: 10.1016/j.bbadis.2020.165836] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023]
Abstract
Acetylation belongs to a class of post-translational modification (PTM) processes that epigenetically regulate gene expression and gene transcriptional activity. Reversible histone acetylation on lysine residues governs the interactions between DNA and histones to mediate chromatin remodeling and gene transcription. Non-histone protein acetylation complicates cellular function whereas acetylation of key mitochondrial enzymes regulates bioenergetic metabolism. Acetylation and deacetylation of functional proteins are essential to the delicated homeostatic regulation of embryonic development, postnatal maturation, cardiomyocyte differentiation, cardiac remodeling and onset of various cardiovascular diseases including obesity, diabetes mellitus, cardiometabolic diseases, ischemia-reperfusion injury, cardiac remodeling, hypertension, and arrhythmias. Histone acetyltransferase (HATs) and histone deacetylases (HDACs) are essential enzymes mainly responsible for the regulation of lysine acetylation levels, thus providing possible drugable targets for therapeutic interventions in the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Mingjie Yang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 210032, China
| | - Yingmei Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 210032, China.
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 210032, China.
| |
Collapse
|
18
|
Wu NN, Tian H, Chen P, Wang D, Ren J, Zhang Y. Physical Exercise and Selective Autophagy: Benefit and Risk on Cardiovascular Health. Cells 2019; 8:cells8111436. [PMID: 31739509 PMCID: PMC6912418 DOI: 10.3390/cells8111436] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022] Open
Abstract
Physical exercise promotes cardiorespiratory fitness, and is considered the mainstream of non-pharmacological therapies along with lifestyle modification for various chronic diseases, in particular cardiovascular diseases. Physical exercise may positively affect various cardiovascular risk factors including body weight, blood pressure, insulin sensitivity, lipid and glucose metabolism, heart function, endothelial function, and body fat composition. With the ever-rising prevalence of obesity and other types of metabolic diseases, as well as sedentary lifestyle, regular exercise of moderate intensity has been indicated to benefit cardiovascular health and reduce overall disease mortality. Exercise offers a wide cadre of favorable responses in the cardiovascular system such as improved dynamics of the cardiovascular system, reduced prevalence of coronary heart diseases and cardiomyopathies, enhanced cardiac reserve capacity, and autonomic regulation. Ample clinical and experimental evidence has indicated an emerging role for autophagy, a conservative catabolism process to degrade and recycle cellular organelles and nutrients, in exercise training-offered cardiovascular benefits. Regular physical exercise as a unique form of physiological stress is capable of triggering adaptation while autophagy in particular selective autophagy seems to be permissive to such cardiovascular adaptation. Here in this mini-review, we will summarize the role for autophagy in particular mitochondrial selective autophagy namely mitophagy in the benefit versus risk of physical exercise on cardiovascular function.
Collapse
Affiliation(s)
- Ne N. Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Haili Tian
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; (H.T.); (P.C.)
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; (H.T.); (P.C.)
| | - Dan Wang
- School of Physical Education and Sport Training, Shanghai University of Sport, Shanghai 200438, China;
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Correspondence: (J.R.); (Y.Z.)
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Correspondence: (J.R.); (Y.Z.)
| |
Collapse
|
19
|
Duan J, Chen Z, Wu Y, Zhu B, Yang L, Yang C. Metabolic remodeling induced by mitokines in heart failure. Aging (Albany NY) 2019; 11:7307-7327. [PMID: 31498116 PMCID: PMC6756899 DOI: 10.18632/aging.102247] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 08/22/2019] [Indexed: 04/11/2023]
Abstract
The prevalence rates of heart failure (HF) are greater than 10% in individuals aged >75 years, indicating an intrinsic link between aging and HF. It has been recognized that mitochondrial dysfunction contributes to the pathology of HF. Mitokines are a type of cytokines, peptides, or signaling pathways produced or activated by the nucleus or the mitochondria through cell non-autonomous responses during cellular stress. In addition to promoting the communication between the mitochondria and the nucleus, mitokines also exert a systemic regulatory effect by circulating to distant tissues. It is noteworthy that increasing evidence has demonstrated that mitokines are capable of reducing the metabolic-related HF risk factors and are associated with HF severity. Consequently, mitokines might represent a potential therapy target for HF.
Collapse
Affiliation(s)
- Jiahao Duan
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Zijun Chen
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Yeshun Wu
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Chun Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
20
|
Chen X, Wang L, Deng Y, Li X, Li G, Zhou J, Cheng D, Yang Y, Yang Q, Chen G, Wang G. Inhibition of Autophagy Prolongs Recipient Survival Through Promoting CD8 + T Cell Apoptosis in a Rat Liver Transplantation Model. Front Immunol 2019; 10:1356. [PMID: 31258533 PMCID: PMC6587890 DOI: 10.3389/fimmu.2019.01356] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
In liver transplantation (LT), although various immunosuppressants have been used in clinical practice, acute rejection remains a common complication that significantly shortens recipient survival. In recent years, manipulating immune tolerance has been regarded as one of the promising solutions to rejection. Autophagy, an evolutionarily conserved protein degradation system, has been reported to be involved in immune rejection and may be a target to establish immune tolerance. However, the role of autophagy in acute rejection reaction after LT has not been elucidated. Here, we showed that the autophagy of CD8+ T cells was strongly enhanced in patients with graft rejection and that the autophagy level was positively correlated with the severity of rejection. Similar findings were observed in a rat acute hepatic rejection model. Furthermore, administration of the autophagy inhibitor 3-methyladenine (3-MA) largely decreased the viability and function of CD8+ T cells through inhibiting autophagy, which significantly prolonged graft survival in rats. In addition, inhibiting the autophagy of activated CD8+ T cells in vitro considerably suppressed mitochondria mediated survival and downregulated T cell function. Conclusions: We first showed that the inhibition of autophagy significantly prolongs liver allograft survival by promoting the apoptosis of CD8+ T cells, which may provide a novel strategy for immune tolerance induction.
Collapse
Affiliation(s)
- Xiaolong Chen
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Li Wang
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yinan Deng
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuejiao Li
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guolin Li
- Department of Biliary and Pancreatic Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Zhou
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Daorou Cheng
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Yang
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qing Yang
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guihua Chen
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Genshu Wang
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
21
|
Li Y, Zhao K, Zong P, Fu H, Zheng Y, Bao D, Yin Y, Chen Q, Lu L, Dai Y, Hou D, Kong X. CD47 deficiency protects cardiomyocytes against hypoxia/reoxygenation injury by rescuing autophagic clearance. Mol Med Rep 2019; 19:5453-5463. [PMID: 31059044 DOI: 10.3892/mmr.2019.10199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 04/04/2019] [Indexed: 11/06/2022] Open
Abstract
To assess the effect of cluster of differentiation (CD47) downregulation on autophagy in hypoxia/reoxygenation (H/R)‑treated H9c2 cardiomyocytes. H9c2 cells were maintained in normoxic conditions (95% air, 5% CO2, 37˚C) without CD47 antibodies, Si‑CD47 or chloroquine (CQ) treatment; H9c2 cells in the H/R group were subjected to 24 h of hypoxia (1% O2, 94% N2, 5% CO2, 37˚C) followed by 12 h of reoxygenation (95% air, 5% CO2, 37˚C). All assays were controlled, triplicated and repeated on three separately initiated cultures. The biochemical parameters in the medium supernatant were measured to evaluate the oxidative stress in cardiomyocytes. The Annexin V‑fluorescein isothiocyanate assay was used to detect the apoptotic rate in the H9c2 cells. Transmission electron microscope, immunofluorescent staining and western blot analysis were performed to detect the effect of the CD47 antibody on autophagic flux in H/R‑treated H9c2 cardiomyocytes. The cardiomyocytic oxidative stress and apoptotic rate decreased and autophagic clearance increased after CD47 downregulation. H/R triggered cell autophagy, autophagosome accumulation and apoptosis in H9c2 cell lines. However, these effects can be attenuated by CD47 downregulation. This study demonstrates its clinical implications in ischemia/reperfusion injury treatment.
Collapse
Affiliation(s)
- Yong Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Kun Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Pengyu Zong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Heling Fu
- Key Laboratory of The Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yuan Zheng
- Key Laboratory of The Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Dan Bao
- Key Laboratory of The Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yuan Yin
- Key Laboratory of The Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qin Chen
- Key Laboratory of The Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lu Lu
- Key Laboratory of The Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Youjin Dai
- Key Laboratory of The Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Daorong Hou
- Key Laboratory of The Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
22
|
Gambogenic acid triggers apoptosis in human nasopharyngeal carcinoma CNE-2Z cells by activating volume-sensitive outwardly rectifying chloride channel. Fitoterapia 2019; 133:150-158. [DOI: 10.1016/j.fitote.2019.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/06/2019] [Accepted: 01/11/2019] [Indexed: 02/08/2023]
|
23
|
Effect of eNOS on Ischemic Postconditioning-Induced Autophagy against Ischemia/Reperfusion Injury in Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5201014. [PMID: 30881990 PMCID: PMC6387714 DOI: 10.1155/2019/5201014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/15/2019] [Indexed: 12/21/2022]
Abstract
Autophagy is involved in the development of numerous illnesses, including ischemia/reperfusion (I/R). Endothelial nitric oxide synthase (eNOS) participates in the protective effects of ischemic postconditioning (IPostC). However, it remains unclear whether eNOS-mediated autophagy serves as a critical role in IPostC in the hearts of mice, in protecting against I/R injury. In the present study, the hearts of mice with left anterior descending coronary artery ligation were studied as I/R models. H9c2 cells underwent exposure to hypoxia/reoxygenation (H/R) and were examined as in vitro model. IPostC reduced mice myocardial infarct size and improved the structure of the heart. IPostC increased the formation of autophagosomes and increased the phosphorylation of eNOS and adenosine monophosphate-activated protein kinase (AMPK). Autophagy and eNOS inhibition suppressed the cardioprotective effects of IPostC. AMPK or eNOS inhibition abolished the improvement effect of IPostC on autophagy. AMPK inhibition decreased eNOS phosphorylation in the heart. Additionally, H9c2 cells suffering hypoxia were used as in vitro model. Autophagy or eNOS inhibition abolished the protective effects of hypoxic postconditioning (HPostC) against H/R injury. AMPK and eNOS inhibition/knockout decreased autophagic activity in the HPostC group. These results indicated that IPostC protects the heart against I/R injury, partially via promoting AMPK/eNOS-mediated autophagy.
Collapse
|
24
|
Ren Z, Xiao W, Zeng Y, Liu MH, Li GH, Tang ZH, Qu SL, Hao YM, Yuan HQ, Jiang ZS. Fibroblast growth factor-21 alleviates hypoxia/reoxygenation injury in H9c2 cardiomyocytes by promoting autophagic flux. Int J Mol Med 2019; 43:1321-1330. [PMID: 30664197 PMCID: PMC6365083 DOI: 10.3892/ijmm.2019.4071] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 01/17/2019] [Indexed: 12/16/2022] Open
Abstract
Fibroblast growth factor (FGF)-21, a member of the family of FGFs, exhibits protective effects against myocardial ischemia and ischemia/reperfusion injury; it is also an enhancer of autophagy. However, the mechanisms underlying the protective role of FGF-21 against cardiomyocyte hypoxia/reoxygenation (H/R) injury remain unclear. The present study aimed to investigate the effect of FGF-21 on H9c2 cardiomyocyte injury induced by H/R and the mechanism associated with changes in autophagy. Cultured H9c2 cardiomyocytes subjected to hypoxia were treated with a vehicle or FGF-21 during reoxygenation. The viability of H9c2 rat cardiomyocytes was measured using Cell Counting Kit-8 and trypan blue exclusion assays. The contents of creatine kinase (CK) and creatine kinase isoenzymes (CK-MB), cardiac troponin I (cTnT), cardiac troponin T (cTnI) and lactate dehydrogenase (LDH) in culture medium were detected with a CK, CK-MB, cTnT, cTnI and LDH assay kits. The protein levels were examined by western blot analysis. Autophagic flux was detected by Ad-mCherry-GFP-LC3B autophagy fluorescent adenovirus reagent. The results indicated that FGF-21 alleviated H/R-induced H9c2 myocardial cell injury and enhanced autophagic flux during H/R, and that this effect was antagonized by co-treatment with 3-methyladenine, an autophagy inhibitor. Furthermore, FGF-21 increased the expression levels of Beclin-1 and Vps34 proteins, but not of mechanistic target of rapamycin. These data indicate that FGF-21 treatment limited H/R injury in H9c2 cardiomyocytes by promoting autophagic flux through upregulation of the expression levels of Beclin-1 and Vps34 proteins.
Collapse
Affiliation(s)
- Zhong Ren
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Weijin Xiao
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yun Zeng
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Mi-Hua Liu
- Department of Clinical Laboratory, Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Guo-Hua Li
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhi-Han Tang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shun-Lin Qu
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Ya-Meng Hao
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hou-Qin Yuan
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhi-Sheng Jiang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
25
|
Wang R, Lin J, Bagchi RA. Novel molecular therapeutic targets in cardiac fibrosis: a brief overview 1. Can J Physiol Pharmacol 2018; 97:246-256. [PMID: 30388374 DOI: 10.1139/cjpp-2018-0430] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiac fibrosis, characterized by excessive accumulation of extracellular matrix, abolishes cardiac contractility, impairs cardiac function, and ultimately leads to heart failure. In recent years, significant evidence has emerged that supports the highly dynamic and responsive nature of the cardiac extracellular matrix. Although our knowledge of cardiac fibrosis has advanced tremendously over the past decade, there is still a lack of specific therapies owing to an incomplete understanding of the disease etiology and process. In this review, we attempt to highlight some of the recently investigated molecular determinants of ischemic and non-ischemic fibrotic remodeling of the myocardium that present as promising avenues for development of anti-fibrotic therapies.
Collapse
Affiliation(s)
- Ryan Wang
- a Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Justin Lin
- b Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Rushita A Bagchi
- c Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
26
|
Chen P, Shang A, Yang J, Wang W. microRNA‐874 inhibition targeting STAT3 protects the heart from ischemia–reperfusion injury by attenuating cardiomyocyte apoptosis in a mouse model. J Cell Physiol 2018; 234:6182-6193. [PMID: 30370578 DOI: 10.1002/jcp.27398] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/16/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Pei‐Jun Chen
- Department of Anesthesiology The First Affiliated Hospital of Soochow University Suzhou China
- Department of Anesthesiology The Sixth People’s Hospital of Yancheng City Yancheng China
| | - An‐Quan Shang
- Department of Laboratory Medicine School of Medicine, Tongji Hospital of Tongji University Shanghai China
| | - Jian‐Ping Yang
- Department of Anesthesiology The First Affiliated Hospital of Soochow University Suzhou China
| | - Wei‐Wei Wang
- Department of Pathology The Sixth People’s Hospital of Yancheng City Yancheng China
| |
Collapse
|
27
|
Chang CJ, Li SJ, Chen YC, Huang SY, Chen SA, Chen YJ. Histone deacetylase inhibition attenuates atrial arrhythmogenesis in sterile pericarditis. Transl Res 2018; 200:54-64. [PMID: 30670155 DOI: 10.1016/j.trsl.2018.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 06/06/2018] [Accepted: 06/06/2018] [Indexed: 10/14/2022]
Abstract
Cardiac surgery is complicated with atrial fibrillation (AF). Histone deacetylase (HDAC) inhibition reduces AF occurrence. In pericarditis, HDAC inhibition may modulate AF trigger and substrate. We recorded electrocardiograms in control and pericardiotomic (op) rabbits without and with an intraperitoneal injection of MPT0E014 (HDAC inhibitor). Conventional microelectrodes recorded action potentials (APs) in pulmonary veins (PVs), the right and left atrium (LA). Masson's trichrome was used to identify collagen fibers in PVs and the LA. Electrocardiograms showed frequent atrial premature contractions in op rabbits, but not in the other 3 groups. The beating rates in PVs and opPVs were decreased by MPT0E014 treatment. Spontaneous burst firings occurred in opPVs (36.4%), but not in control PVs. H2O2 induced greater burst firings in opPVs (72.7%) than in control PVs (11.1%), MPT0E014-treated PVs (16.7%), and MPT0E014-treated opPVs (12.5%). The AP duration at a repolarization extent of 90% (APD90) was shorter in the opLA than that in the control LA. In the presence of isoproterenol (1 μM), rapid atrial pacing (RAP, 20 Hz) induced a higher incidence of burst firings in the opLA (90%) than in the other groups. In contrast, acetylcholine (5 mM) and RAP induced a lower incidence of burst firing in the MPT0E014-treated LA (33.3%) than in the other groups. Fibrosis prevailed in opPVs and the opLA compared to the respective control PVs and LA, which was attenuated in those that received MPT0E014. In conclusion, a pericardiotomy increased fibrosis and arrhythmogenesis in PVs and the LA, which were prevented by HDAC inhibition.
Collapse
Affiliation(s)
- Chien-Jung Chang
- Division of Cardiology, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Shao-Jung Li
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering and Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Yu Huang
- Division of Cardiology, Department of Internal Medicine, Cathay General Hospital, Taipei, Taiwan
| | - Shih-Ann Chen
- National Yang-Ming University, School of Medicine, Taipei, Taiwan; Division of Cardiology and Cardiovascular Research Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
28
|
Mst1 knockout enhances cardiomyocyte autophagic flux to alleviate angiotensin II-induced cardiac injury independent of angiotensin II receptors. J Mol Cell Cardiol 2018; 125:117-128. [PMID: 30193956 DOI: 10.1016/j.yjmcc.2018.08.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/19/2018] [Accepted: 08/29/2018] [Indexed: 11/22/2022]
Abstract
AIMS Angiotension II (Ang II) plays a central role in the pathogenesis of renin-angiotensin-aldosterone system (RAAS)-induced heart failure. Mst1 exerts its function in cardiomyocytes subjected to pathological stimuli via inhibiting autophagy and aggravating apoptosis, but its role in RAAS-mediated cardiac injury is still unknown. Here, we aimed to determine whether cardiomyocyte-specific Mst1 knockout can alleviate Ang II-induced cardiac injury by improving cardiomyocyte autophagy and whether these functions depend on Ang II receptors. RESULTS Mst1 knockout alleviated Ang II-induced heart failure, without affecting blood pressure and compensatory concentric hypertrophy. Mst1 specific knockout improved the effects of Ang II on cardiomyocyte autophagy, as evidenced by further increased LC3-II expression and decreased P62 expression. More typical autophagosomes accompanied by less damaged mitochondria were also observed by electron microscopy in Ang II-treated Mst1Δ/Δ mice. In vitro, Mst1 knockdown promoted cardiomyocyte autophagic flux, as demonstrated by more GFP-mRFP-LC3 puncta per cell. Increased LC3-II and decreased P62 expression both in the presence and absence of chloroquine were observed in Mst1 knockdown cardiomyocytes administered with Ang II. Treatment with 3-MA, an inhibitor of autophagy, abolished the beneficial effects of Mst1 knockout against Ang II-induced cardiac dysfunction. The compensatory effects of Ang II on upregulated autophagy were associated with Mst1 inhibition. Interestingly, the knockdown or antagonization of AT1R inhibited cardiomyocyte autophagy, which may represent a threat to cardiac function. Importantly, Mst1 knockout consistently enhanced cardiomyocyte autophagy following the knockdown or blocking of AT1R and AT2R. CONCLUSION Cardiomyocyte-specific Mst1 knockout alleviates Ang II-induced cardiac injury by enhancing cardiomyocyte autophagy. Mst1 inhibition may counteract the undesirable effects of Ang II receptors blockage on cardiomyocyte autophagy and represent a promising complementary treatment strategy against Ang II-induced cardiac injury.
Collapse
|
29
|
The Impact of Moderate Chronic Hypoxia and Hyperoxia on the Level of Apoptotic and Autophagic Proteins in Myocardial Tissue. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5786742. [PMID: 30186545 PMCID: PMC6116398 DOI: 10.1155/2018/5786742] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/08/2018] [Accepted: 06/21/2018] [Indexed: 02/07/2023]
Abstract
The redox imbalance and the consequent oxidative stress have been implicated in many pathological conditions, including cardiovascular diseases. The lack or the excess of O2 supply can alter the redox balance. The aim of the present study was to understand the heart responses to prolonged hypoxia or hyperoxia and how such situations may activate survival mechanisms or trigger cell death. Seven-week-old Foxn1 mice were exposed to hypoxia (10% O2), normoxia (21% O2), or hyperoxia (30% O2) for 28 days, then the heart tissue was excised and analyzed. The alterations in redox balance, housekeeping protein levels, and autophagic and apoptotic process regulation were studied. The D-ROM test demonstrated an increased oxidative stress in the hypoxic group compared to the hyperoxic group. The level of hypoxia inducible factor-1 (HIF-1α) was increased by hypoxia while HIF-2α was not affected by treatments. Chronic hypoxia activated the biochemical markers of autophagy, and we observed elevated levels of Beclin-1 while LC3B-II and p62 were constant. Nevertheless, we measured significantly enhanced number of TUNEL-positive cells and higher Bax/Bcl2 ratio in hyperoxia with respect to hypoxia. Surprisingly, our results revealed alterations in the level of housekeeping proteins. The expression of α-tubulin, total-actin, and GAPDH was increased in the hypoxic group while decreased in the hyperoxic group. These findings suggest that autophagy is induced in the heart under hypoxia, which may serve as a protective mechanism in response to enhanced oxidative stress. While prolonged hypoxia-induced autophagy leads to reduced heart apoptosis, low autophagic level in hyperoxia failed to prevent the excessive DNA fragmentation.
Collapse
|
30
|
Liu YY, Sun C, Xue FS, Yang GZ, Li HX, Liu Q, Liao X. Effect of Autophagy Inhibition on the Protection of Ischemia Preconditioning against Myocardial Ischemia/Reperfusion Injury in Diabetic Rats. Chin Med J (Engl) 2018; 131:1702-1709. [PMID: 29998890 PMCID: PMC6048915 DOI: 10.4103/0366-6999.235867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Ischemia preconditioning (IPC) remains the most powerful intervention of protection against myocardial ischemia/reperfusion injury (IRI), but diabetes can weaken or eliminate its cardioprotective effect and detailed mechanisms remain unclear. In this study, we aimed to explore whether changes of autophagy in the diabetic condition are attributable to the decreased cardioprotective effect of IPC. METHODS Sixty diabetic male Sprague-Dawley rats were randomly divided into the control (C), IRI, rapamycin (R), wortmannin (W), rapamycin + IPC (R + IPC), and wortmannin + IPC (W + IPC) groups. The in vivo rat model of myocardial IRI was established by ligaturing and opening the left anterior descending coronary artery via the left thoracotomy. Durations of ischemia and reperfusion are 30 min and 120 min, respectively. Blood samples were taken at 120 min of reperfusion for measuring serum concentrations of troponin I (TnI) and creatine kinase isoenzyme MB (CK-MB) using the enzyme-linked immunosorbent assay. The infarct size was assessed by Evans blue and triphenyltetrazolium chloride staining. The expressions of LC3-II, beclin-1, phosphoinositide 3-kinase (PI3K), mammalian target of rapamycin (mTOR), and P-Akt/Akt ratio in the ischemic myocardium were assessed by Western blotting. RESULTS Compared to the IRI group, infarct size (56.1% ± 6.1% vs. 75.4 ± 7.1%, P < 0.05), serum cTnI (0.61 ± 0.21 vs. 0.95 ± 0.26 ng/ml, P < 0.05), and CK-MB levels (6.70 ± 1.25 vs. 11.51 ± 2.35 ng/ml, P < 0.05) obviously decreased in the W + IPC group. Compared with the C group, myocardial expressions of LC3-II (0.46 ± 0.04 and 0.56 ± 0.04 vs. 0.36 ± 0.04, P < 0.05) and beclin-1 (0.34 ± 0.08 and 0.38 ± 0.07 vs. 0.24 ± 0.03, P < 0.05) evidently increased, and myocardial expressions of mTOR (0.26 ± 0.08 and 0.25 ± 0.07 vs. 0.38 ± 0.06, P < 0.05), PI3K (0.29 ± 0.04 and 0.30 ± 0.03 vs. 0.38 ± 0.02, P < 0.05), and P-Akt/Akt ratio (0.49 ± 0.10 and 0.48 ± 0.06 vs. 0.72 ± 0.07, P < 0.05) markedly decreased in the IRI and R groups, indicating an increased autophagy. Compared with the IRI group, myocardial expression of beclin-1 (0.26 ± 0.03 vs. 0.34 ± 0.08, P < 0.05) significantly decreased, and myocardial expressions of mTOR (0.36 ± 0.04 vs. 0.26 ± 0.08, P < 0.05), PI3K (0.37 ± 0.03 vs. 0.29 ± 0.04, P < 0.05), and P-Akt/Akt ratio (0.68 ± 0.05 vs. 0.49 ± 0.10, P < 0.05) increased obviously in the W + IPC group, indicating a decreased autophagy. CONCLUSIONS Increased autophagy in the diabetic myocardium is attributable to decreased cardioprotection of IPC, and autophagy inhibited by activating the PI3K-Akt-mTOR signaling pathway can result in an improved protection of IPC against diabetic myocardial IRI.
Collapse
Affiliation(s)
- Ya-Yang Liu
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100041, China
| | - Chao Sun
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100041, China
| | - Fu-Shan Xue
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100041, China
| | - Gui-Zhen Yang
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100041, China
| | - Hui-Xian Li
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100041, China
| | - Qing Liu
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100041, China
| | - Xu Liao
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100041, China
| |
Collapse
|
31
|
Wright LH, Herr DJ, Brown SS, Kasiganesan H, Menick DR. Angiokine Wisp-1 is increased in myocardial infarction and regulates cardiac endothelial signaling. JCI Insight 2018; 3:95824. [PMID: 29467324 DOI: 10.1172/jci.insight.95824] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 01/10/2018] [Indexed: 12/17/2022] Open
Abstract
Myocardial infarctions (MIs) cause the loss of myocytes due to lack of sufficient oxygenation and latent revascularization. Although the administration of histone deacetylase (HDAC) inhibitors reduces the size of infarctions and improves cardiac physiology in small-animal models of MI injury, the cellular targets of the HDACs, which the drugs inhibit, are largely unspecified. Here, we show that WNT-inducible secreted protein-1 (Wisp-1), a matricellular protein that promotes angiogenesis in cancers as well as cell survival in isolated cardiac myocytes and neurons, is a target of HDACs. Further, Wisp-1 transcription is regulated by HDACs and can be modified by the HDAC inhibitor, suberanilohydroxamic acid (SAHA/vorinostat), after MI injury. We observe that, at 7 days after MI, Wisp-1 is elevated 3-fold greater in the border zone of infarction in mice that experience an MI injury and are injected daily with SAHA, relative to MI alone. Additionally, human coronary artery endothelial cells (HCAECs) produce WISP-1 and are responsive to autocrine WISP-1-mediated signaling, which functionally promotes their proangiogenic behavior. Altering endogenous expression of WISP-1 in HCAECs directly impacts their network density in vitro. Therapeutic interventions after a heart attack define the extent of infarct injury, cell survival, and overall prognosis. Our studies shown here identify a potentially novel cardiac angiokine, Wisp-1, that may contribute to beneficial post-MI treatment modalities.
Collapse
Affiliation(s)
| | | | - Symone S Brown
- College of Graduate Studies, Summer Undergraduate Research Program, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Donald R Menick
- Division of Cardiology, and.,Ralph H. Johnson VA Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
32
|
Guo W, Liu X, Li J, Shen Y, Zhou Z, Wang M, Xie Y, Feng X, Wang L, Wu X. Prdx1 alleviates cardiomyocyte apoptosis through ROS-activated MAPK pathway during myocardial ischemia/reperfusion injury. Int J Biol Macromol 2018; 112:608-615. [PMID: 29410271 DOI: 10.1016/j.ijbiomac.2018.02.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 01/06/2018] [Accepted: 02/02/2018] [Indexed: 11/17/2022]
Abstract
Apoptosis induced by oxidative stress blocks the recovery of heart function in myocardial ischemia reperfusion injury (MIRI). Peroxiredoxin 1 (Prdx1) inhibits oxidative stress. However, the expression and function of Prdx1 in MIRI are unclear. In present study, Prdx1 protein level increased in rat MIRI model, associated with cardiomyocyte apoptosis. Cultured rat embryonic ventricular myocardial H9c2 cells with hypoxia/reoxygenation (H/R) treatment was utilized to mimic MIRI in vitro, showing that H/R treatment increased the ratio of p-p38/p38, p-JNK/JNK and apoptosis index. But Prdx1 ameliorate the up-regulation of p-p38/p38 ratio and p-JNK/JNK ratio, as well as decreased H9c2 cell apoptosis. SB203580 (p38 inhibitor) and SP600125 (JNK inhibitor) inhibited H9c2 cell apoptosis, and at the same time Prdx1 down-regulated the activation of p38 MAPK and JNK during H/R treatment. In addition, a ROS scavenger N-acetyl-l-cysteine (NAC) down-regulated the protein level of p-p38, p-JNK and Prdx1, and H9c2 cell apoptosis. In summary, these findings indicated that Prdx1 inhibited MAPK pathway induced cells apoptosis, and ROS is the upstream regulator of H/R induced apoptosis.
Collapse
Affiliation(s)
- Wanwan Guo
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Jingjing Li
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Yimin Shen
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Zijian Zhou
- Department of Geriatric Medicine, the Second People's Hospital, Nantong 226001, Jiangsu, China
| | - Mingming Wang
- Department of Geriatric Medicine, the Second People's Hospital, Nantong 226001, Jiangsu, China
| | - Yuyi Xie
- Department of Geriatric Medicine, the Second People's Hospital, Nantong 226001, Jiangsu, China
| | - Xuemei Feng
- Department of Geriatric Medicine, the Second People's Hospital, Nantong 226001, Jiangsu, China
| | - Liyang Wang
- Department of Geriatric Medicine, the Second People's Hospital, Nantong 226001, Jiangsu, China
| | - Xiang Wu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
33
|
Xia Y, Liu Y, Xia T, Li X, Huo C, Jia X, Wang L, Xu R, Wang N, Zhang M, Li H, Wang X. Activation of volume-sensitive Cl- channel mediates autophagy-related cell death in myocardial ischaemia/reperfusion injury. Oncotarget 2018; 7:39345-39362. [PMID: 27322431 PMCID: PMC5129937 DOI: 10.18632/oncotarget.10050] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/03/2016] [Indexed: 12/19/2022] Open
Abstract
Excessive reactive oxygen species (ROS) plays an important role in myocardial ischemia/reperfusion (I/R) injury, which triggers not only myocardial cellular apoptosis but also autophagy-related cell death, in which volume-sensitive outwardly rectifying (VSOR) Cl− channel-activated by ROS contributes to cell apoptotic volume decrease, playing an incipient incident of cellular apoptosis. However, whether VSOR Cl− channel concurrently participates in autophagy-related cell death regulation remains unclear. To illuminate the issue, studies underwent in myocardial vitro and vivo I/R model. Rats were performed to ischemia 30 minutes and subsequent reperfusion 24-96 hours, ROS scavenger (NAC), VSOR Cl− channel blocker (DCPIB) and autophagy inhibitor (3MA) were administered respectively. Results showed that oxidative stress, LC3-II stain and inflammation in myocardial tissue were markedly increased, lysosome associated membrane protein-2 (LAMP2) were significantly reduced with I/R group as compared with sham group, reperfusion significantly led to damage in myocardial tissue and heart function, whereas the disorder could be rescued through these agents. Moreover, primary neonatal rat cardiomyocytes hypoxia/reoxygenation model were administered, results showed that VSOR Cl− channel-activated by reoxygenation could cause both cell volume decrease and intracellular acidification, which further increased LC3 and depleted of LAMP2, resulting in autophagy-related cell death. Interestingly, VSOR Cl− channel-blocked by DCPIB could stably maintain the cell volume, intracellular pH, abundant LAMP2 and autophagic intensity regardless of ROS intension derived from reoxygenation injury or adding H2O2. These results first demonstrate that VSOR Cl− channel-activated is a pivotal event to trigger autophagy-related death, which reveals a novel therapeutic target to decrease myocardial I/R injury.
Collapse
Affiliation(s)
- Yuesheng Xia
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yan Liu
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tong Xia
- Department of Preventive Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xing Li
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Cong Huo
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin Jia
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lin Wang
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Rong Xu
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ning Wang
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Mingming Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hong Li
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaoming Wang
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
34
|
Jiang XX, Liu GY, Lei H, Li ZL, Feng QP, Huang W. Activation of transient receptor potential vanilloid 1 protects the heart against apoptosis in ischemia/reperfusion injury through upregulating the PI3K/Akt signaling pathway. Int J Mol Med 2017; 41:1724-1730. [PMID: 29286076 DOI: 10.3892/ijmm.2017.3338] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 12/13/2017] [Indexed: 11/06/2022] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is a nonselective cation channel and a molecular integrator of noxious stimuli. TRPV1 activation confers cardiac protection against ischemia/reperfusion (I/R) injury. The present study aimed to investigate whether the cardioprotective effects of TRPV1 were associated with the inhibition of apoptosis via the phosphatidylinositol 3‑kinase (PI3K)/protein kinase B (Akt) and extracellular signal‑regulated protein kinase 1/2 (ERK1/2) signaling pathways. Briefly, the hearts of TRPV1 knockout (TRPV1‑/‑) or wild‑type (WT) mice were isolated and subjected to 30 min of ischemia followed by 60 min of reperfusion in a Langendorff apparatus in the presence or absence of the PI3K inhibitor, LY294002. At the end of reperfusion, infarct size was measured using 2,3,5‑triphenyltetrazolium chloride staining and myocardial apoptosis was assessed by terminal deoxynucleotidyl transferase‑mediated dUTP nick‑end labeling (TUNEL) staining. The expression levels of B‑cell lymphoma 2 (Bcl‑2), Bcl‑2‑associated X protein (Bax), and phosphorylated Akt and ERK1/2 were determined by western blot analysis. There was a significant increase in the extent of infarction and the percentage of TUNEL‑positive cells, and a decrease in the Bcl‑2/Bax ratio, and Akt and ERK1/2 phosphorylation in TRPV1‑/‑ hearts. In addition, treatment with LY294002 increased infarct size and the percentage of TUNEL‑positive cells, and reduced Bcl‑2/Bax expression and Akt phosphorylation in WT hearts, but not in TRPV1‑/‑ hearts, following I/R. Taken together, these data suggested that TRPV1 serves a protective role against myocardial apoptosis during I/R via the PI3K/Akt signaling pathway. In conclusion, activating TRPV1 may be considered a potential approach to protect the heart against I/R injury.
Collapse
Affiliation(s)
- Xiao-Xue Jiang
- Cardiovascular Laboratory, Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Guan-Yu Liu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Han Lei
- Cardiovascular Laboratory, Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zuo-Ling Li
- Cardiovascular Laboratory, Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qing-Ping Feng
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON N6A‑5C1, Canada
| | - Wei Huang
- Cardiovascular Laboratory, Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
35
|
Li S, Liu C, Gu L, Wang L, Shang Y, Liu Q, Wan J, Shi J, Wang F, Xu Z, Ji G, Li W. Autophagy protects cardiomyocytes from the myocardial ischaemia-reperfusion injury through the clearance of CLP36. Open Biol 2017; 6:rsob.160177. [PMID: 27512143 PMCID: PMC5008017 DOI: 10.1098/rsob.160177] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/08/2016] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of the death worldwide. An increasing number of studies have found that autophagy is involved in the progression or prevention of CVD. However, the precise mechanism of autophagy in CVD, especially the myocardial ischaemia-reperfusion injury (MI/R injury), is unclear and controversial. Here, we show that the cardiomyocyte-specific disruption of autophagy by conditional knockout of Atg7 leads to severe contractile dysfunction, myofibrillar disarray and vacuolar cardiomyocytes. A negative cytoskeleton organization regulator, CLP36, was found to be accumulated in Atg7-deficient cardiomyocytes. The cardiomyocyte-specific knockout of Atg7 aggravates the MI/R injury with cardiac hypertrophy, contractile dysfunction, myofibrillar disarray and severe cardiac fibrosis, most probably due to CLP36 accumulation in cardiomyocytes. Altogether, this work reveals autophagy may protect cardiomyocytes from the MI/R injury through the clearance of CLP36, and these findings define a novel relationship between autophagy and the regulation of stress fibre in heart.
Collapse
Affiliation(s)
- Shiguo Li
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Lei Gu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Lina Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yongliang Shang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Qiong Liu
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Junyi Wan
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Jian Shi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Zhiliang Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Guangju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
36
|
Rasekhi M, Soleimani M, Bakhshandeh B, Sadeghizadeh M. A novel protocol to provide a suitable cardiac model from induced pluripotent stem cells. Biologicals 2017; 50:42-48. [DOI: 10.1016/j.biologicals.2017.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 09/06/2017] [Accepted: 09/11/2017] [Indexed: 12/11/2022] Open
|
37
|
Xue Q, Pei H, Liu Q, Zhao M, Sun J, Gao E, Ma X, Tao L. MICU1 protects against myocardial ischemia/reperfusion injury and its control by the importer receptor Tom70. Cell Death Dis 2017; 8:e2923. [PMID: 28703803 PMCID: PMC5550843 DOI: 10.1038/cddis.2017.280] [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: 03/04/2017] [Revised: 04/21/2017] [Accepted: 05/15/2017] [Indexed: 01/26/2023]
Abstract
Mitochondrial Ca2+ overload is a main contributor to mitochondrial damage hence cardiomyocyte death in myocardial ischemia/reperfusion (MI/R) injury. MICU1 has been recently identified as an important regulator of mitochondrial Ca2+ homeostasis. Here we try to identify the role of MICU1 in MI/R, and to investigate whether the mitochondrial importer receptor Tom70 possesses critical roles in the mitochondrial translocation of MICU1 and MI/R. Specific small interfering RNA (20 μg) against MICU1 and Tom70, and lentivirus vectors carrying the Tom70a sequences (3.3 × 107 TU) were delivered through intramyocardial injection. Seventy-two hours after injection, mice were subjected to 30 min of MI followed by 3 h (for cell apoptosis and mitochondrial damage assessment) or 24 h (for cardiac function and infarct size determination) of reperfusion. MI/R had no significant effect on total MICU1 expression, but caused significant reduction of MICU1 in mitochondria. Knockdown of MICU1 significantly aggravated MI/R injury, as evidenced by enlarged infarct size, depressed cardiac function and increased myocardial apoptosis. Moreover, MICU1 deficiency resulted in markedly aggravated mitochondrial Ca2+ overload, consequently destructed mitochondrial morphology and suppressed mitochondrial function (evidenced by decreased ATP production). Interestingly, mitochondrial Tom70 was also decreased in MI/R. Genetic loss-function study revealed that mitochondrial MICU1 expression was depressed by Tom70 ablation. Furthermore, Tom70 deficiency significantly aggravated MI/R injury and worsened mitochondrial Ca2+ overload. However, supplementation of Tom70 significantly attenuated MI/R injury, preserved mitochondrial morphology and function, and inhibited mitochondrial Ca2+ overload, all of which were abolished by MICU1 suppression. Mitochondrial Tom70/MICU1 pathway protects against MI/R injury, in which mitochondrial localization of MICU1 is governed by Tom70, and MICU1 serves as an indispensable factor in Tom70’s cardioprotection.
Collapse
Affiliation(s)
- Qiang Xue
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Haifeng Pei
- Department of Cardiology, Chengdu Military General Hospital, Chengdu 610083, China
| | - Qinshe Liu
- Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Mingjun Zhao
- Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Jing Sun
- Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Erhe Gao
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
38
|
Remote Postconditioning Induced by Trauma Protects the Mouse Heart against Ischemia Reperfusion Injury. Involvement of the Neural Pathway and Molecular Mechanisms. Cardiovasc Drugs Ther 2017; 30:271-80. [PMID: 27067902 DOI: 10.1007/s10557-016-6661-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE Abdominal superficial surgical incision elicits cardioprotection against myocardial ischemia reperfusion (I/R) injury in mice. This cardioprotective phenomenon, termed remote preconditioning of trauma (RPCT), results in an 80 to 85 % reduction in cardiac infarct size. We evaluated cardioprotection and the molecular mechanisms of remote postconditioning of trauma (RPostCT) in a murine I/R injury model. METHODS Mice were analyzed using a previously established I/R injury model. An abdominal superficial surgical incision was made 45 min after myocardial ischemia at the end of coronary occlusion, and infarct size was determined 24 h after reperfusion. RESULTS The results indicated that a strong cardioprotective effect occurred during RPostCT (56.94 ± 2.71 % sham vs. 15.58 ± 2.16 % RPostCT; the mean area of the infarct divided by the mean area of the region at risk; p ≤ 0.05; n = 10). Furthermore, pharmacological intervention revealed neurogenic signaling involvement in the beneficial effects of RPostCT via sensory and sympathetic thoracic nerves. Pharmacological experiments in transgenic mice demonstrated that bradykinin receptors, β-adrenergic receptors (AR), and protein kinase C were implicated in the cardioprotective effects of RPostCT. CONCLUSIONS RPostCT significantly decreased myocardial infarction size via neurogenic transmission and various signaling pathways. This study describes a new cardiac I/R injury prevention method that might lead to the development of therapies that are more clinically relevant for myocardial I/R injury.
Collapse
|
39
|
Potential signaling pathways of acute endurance exercise-induced cardiac autophagy and mitophagy and its possible role in cardioprotection. J Physiol Sci 2017; 67:639-654. [PMID: 28685325 PMCID: PMC5684252 DOI: 10.1007/s12576-017-0555-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/20/2017] [Indexed: 02/06/2023]
Abstract
Cardiac myocytes are terminally differentiated cells and possess extremely limited regenerative capacity; therefore, preservation of mature cardiac myocytes throughout the individual's entire life span contributes substantially to healthy living. Autophagy, a lysosome-dependent cellular catabolic process, is essential for normal cardiac function and mitochondria maintenance. Therefore, it may be reasonable to hypothesize that if endurance exercise promotes cardiac autophagy and mitochondrial autophagy or mitophagy, exercise-induced cardiac autophagy (EICA) or exercise-induced cardiac mitophagy (EICM) may confer propitious cellular environment and thus protect the heart against detrimental stresses, such as an ischemia-reperfusion (I/R) injury. However, although the body of evidence supporting EICA and EICM is growing, the molecular mechanisms of EICA and EICM and their possible roles in cardioprotection against an I/R injury are poorly understood. Here, we introduce the general mechanisms of autophagy in an attempt to integrate potential molecular pathways of EICA and EICM and also highlight a potential insight into EICA and EICM in cardioprotection against an I/R insult.
Collapse
|
40
|
Shao H, Yang L, Wang L, Tang B, Wang J, Li Q. MicroRNA-34a protects myocardial cells against ischemia-reperfusion injury through inhibiting autophagy via regulating TNFα expression. Biochem Cell Biol 2017; 96:349-354. [PMID: 28544853 DOI: 10.1139/bcb-2016-0158] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND ischemia-reperfusion (I/R) is a consequence of restored blood supply after myocardial infarction. Myocardial I/R injury can be alleviated by reducing autophagy in heart tissue. MicroRNA-34a (miR-34a) has been shown to regulate autophagy in a renal model of I/R, but it is not known whether it can protect cardiac tissues from I/R injury. This study investigated how miR-34a protects myocardial cells from I/R injury by inhibiting autophagy via regulation of tumor necrosis factor α (TNFα). METHODS we constructed an I/R model in vivo using Langendorff perfusion, and we constructed an in vivo model by treating neonatal rat cardiomyocytes (NRCMs) with hypoxia-reoxygenation (H/R method). Transfected adenoviral-overexpressed miR-34a mimics and controlled NRCMs after H/R. We analyzed cell viability using the MTT assay and a cell counting kit-8 (CCK-8) assay. Changes in the rate of apoptosis were detected by flow cytometry. We investigated the effect mechanisms of miR-34a with Western blot and luciferase assays. RESULTS miR-34a expression decreased after in vivo reperfusion of the myocardial cells and heart tissues of neonatal rats. MiR-34a reduced apoptosis of the NRCMs and autophagy levels, simultaneously, after H/R injury. Further, miR-34a decreased the expression of Lc3-II and p62, indicating that miR-34a reduces myocardial I/R injury by decreasing TNFα expression. CONCLUSION miR-34a can inhibit autophagy levels after I/R by targeting TNFα, thereby reducing myocardial injury.
Collapse
Affiliation(s)
- Haifeng Shao
- Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China.,Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Lili Yang
- Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China.,Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Li Wang
- Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China.,Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Bozan Tang
- Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China.,Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Jian Wang
- Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China.,Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| | - Qiang Li
- Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China.,Department of Cardiology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, China
| |
Collapse
|
41
|
He S, Wang X, Zhong Y, Tang L, Zhang Y, Ling Y, Tan Z, Yang P, Chen A. Hesperetin post-treatment prevents rat cardiomyocytes from hypoxia/reoxygenation injury in vitro via activating PI3K/Akt signaling pathway. Biomed Pharmacother 2017; 91:1106-1112. [PMID: 28531921 DOI: 10.1016/j.biopha.2017.05.003] [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: 01/31/2017] [Revised: 04/11/2017] [Accepted: 05/01/2017] [Indexed: 01/04/2023] Open
Abstract
Hesperidin (HES), a citrus fruit extract, has beneficial effects on various ischemia/reperfusion (I/R) models. Here, we investigated the possible positive effect of hesperetin (HPT), an active metabolite of HES, and identified the potential molecular mechanisms involved in cardiomyocytes H/R-induced injury. To construct the cardiomyocyte model of hypoxia/reoxygenation (H/R) injury, cultured neonatal rat cardiomyocytes were subjected to 3h of hypoxia followed by 3h of reoxygenation. Cell viability and apoptosis were detected. The levels of Apoptosis-related proteins and PI3K/Akt proteins were detected by western blot. Our results showed that HPT post-treatment significantly inhibited apoptosis by elevating the expression of Bcl-2, decreasing the expression of Bax and cleaved caspase-3, and diminished the apoptotic cardiomyocytes ratio. Mechanism studies demonstrated that HPT post-treatment up-regulated the expression levels of p-PI3K, and p-Akt. Co-treatment of the cardiomyocytes with the PI3K/Akt-specific inhibitor LY294002 blocked the HPT-induced cardioprotective effects. Taken together, these data suggested that HPT post-treatment prevented cardiomyocytes from H/R injury in vitro most likely through the activation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Shangfei He
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, No. 253 Gongye Road, Guangzhou 510280, China
| | - Xianbao Wang
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, No. 253 Gongye Road, Guangzhou 510280, China
| | - Yongkang Zhong
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, No. 253 Gongye Road, Guangzhou 510280, China
| | - Lu Tang
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, No. 253 Gongye Road, Guangzhou 510280, China
| | - Ya Zhang
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, No. 253 Gongye Road, Guangzhou 510280, China
| | - Yuanna Ling
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, No. 253 Gongye Road, Guangzhou 510280, China
| | - Zhipeng Tan
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, No. 253 Gongye Road, Guangzhou 510280, China
| | - Pingzhen Yang
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, No. 253 Gongye Road, Guangzhou 510280, China
| | - Aihua Chen
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, No. 253 Gongye Road, Guangzhou 510280, China.
| |
Collapse
|
42
|
Pohjoismäki JL, Goffart S. The role of mitochondria in cardiac development and protection. Free Radic Biol Med 2017; 106:345-354. [PMID: 28216385 DOI: 10.1016/j.freeradbiomed.2017.02.032] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/27/2017] [Accepted: 02/14/2017] [Indexed: 12/31/2022]
Abstract
Mitochondria are essential for the development as well as maintenance of the myocardium, the most energy consuming tissue in the human body. Mitochondria are not only a source of ATP energy but also generators of reactive oxygen species (ROS), that cause oxidative damage, but also regulate physiological processes such as the switch from hyperplastic to hypertrophic growth after birth. As excess ROS production and oxidative damage are associated with cardiac pathology, it is not surprising that much of the research focused on the deleterious aspects of free radicals. However, cardiomyocytes are naturally highly adapted against repeating oxidative insults, with evidence suggesting that moderate and acute ROS exposure has beneficial consequences for mitochondrial maintenance and cardiac health. Antioxidant defenses, mitochondrial quality control, mtDNA maintenance mechanisms as well as mitochondrial fusion and fission improve mitochondrial function and cardiomyocyte survival under stress conditions. As these adaptive processes can be induced, promoting mitohormesis or mitochondrial biogenesis using controlled ROS exposure could provide a promising strategy to increase cardiomyocyte survival and prevent pathological remodeling of the myocardium.
Collapse
Affiliation(s)
- Jaakko L Pohjoismäki
- University of Eastern Finland, Department of Environmental and Biological Sciences, P.O. Box 111, 80101 Joensuu, Finland.
| | - Steffi Goffart
- University of Eastern Finland, Department of Environmental and Biological Sciences, P.O. Box 111, 80101 Joensuu, Finland
| |
Collapse
|
43
|
Yu J, Wu J, Xie P, Maimaitili Y, Wang J, Xia Z, Gao F, Zhang X, Zheng H. Sevoflurane postconditioning attenuates cardiomyocyte hypoxia/reoxygenation injury via restoring mitochondrial morphology. PeerJ 2016; 4:e2659. [PMID: 27833818 PMCID: PMC5101611 DOI: 10.7717/peerj.2659] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/05/2016] [Indexed: 12/25/2022] Open
Abstract
Background Anesthetic postconditioning is a cellular protective approach whereby exposure to a volatile anesthetic renders a tissue more resistant to subsequent ischemic/reperfusion event. Sevoflurane postconditioning (SPostC) has been shown to exert cardioprotection against ischemia/reperfusion injury, but the underlying mechanism is unclear. We hypothesized that SPostC protects cardiomyocytes against hypoxia/reoxygenation (H/R) injury by maintaining/restoring mitochondrial morphological integrity, a critical determinant of cell fate. Methods Primary cultures of neonatal rat cardiomyocytes (NCMs) were subjected to H/R injury (3 h of hypoxia followed by 3 h reoxygenation). Intervention with SPostC (2.4% sevoflurane) was administered for 15 min upon the onset of reoxygenation. Cell viability, Lactate dehydrogenase (LDH) level, cell death, mitochondrial morphology, mitochondrial membrane potential and mitochondrial permeability transition pore (mPTP) opening were assessed after intervention. Mitochondrial fusion and fission regulating proteins (Drp1, Fis1, Mfn1, Mfn2 and Opa1) were assessed by immunofluorescence staining and western blotting was performed to determine the level of protein expression. Results Cardiomyocyte H/R injury resulted in significant increases in LDH release and cell death that were concomitant with reduced cell viability and reduced mitochondrial interconnectivity (mean area/perimeter ratio) and mitochondrial elongation, and with reduced mitochondrial membrane potential and increased mPTP opening. All the above changes were significantly attenuated by SPostC. Furthermore, H/R resulted in significant reductions in mitochondrial fusion proteins Mfn1, Mfn2 and Opa1 and significant enhancement of fission proteins Drp1 and Fis1. SPostC significantly enhanced Mfn2 and Opa1 and reduced Drp1, without significant impact on Mfn1 and Fis1. Conclusions Sevoflurane postconditioning attenuates cardiomyocytes hypoxia/reoxygenation injury (HRI) by restoring mitochondrial fusion/fission balance and morphology.
Collapse
Affiliation(s)
- Jin Yu
- Department of Anethesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Jianjiang Wu
- Department of Anethesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Peng Xie
- Department of Anethesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Yiliyaer Maimaitili
- Department of Anethesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Jiang Wang
- Department of Anethesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| | - Zhengyuan Xia
- Department of Anethesiology, University of Hong Kong , Hongkong , China
| | - Feng Gao
- Department of Aerospace Medicine, School of Basic Medical Sciences, Fourth Military Medical University , Xi'an, Shaanxi , China
| | - Xing Zhang
- Department of Aerospace Medicine, School of Basic Medical Sciences, Fourth Military Medical University , Xi'an, Shaanxi , China
| | - Hong Zheng
- Department of Anethesiology, The First Affiliated Hospital of Xinjiang Medical University , Urumqi, Xinjiang , China
| |
Collapse
|
44
|
Li C, Zhang C, Wang T, Xuan J, Su C, Wang Y. Heme oxygenase 1 induction protects myocardiac cells against hypoxia/reoxygenation-induced apoptosis : The role of JNK/c-Jun/Caspase-3 inhibition and Akt signaling enhancement. Herz 2016; 41:715-724. [PMID: 27220977 DOI: 10.1007/s00059-016-4424-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 02/11/2016] [Accepted: 02/20/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Although recent studies have found that heme oxygenase (HO)-1 plays an important role in myocardiac cell survival, the precise mechanisms occurring during hypoxia/reoxygenation (H/R) injury remain unclear. Therefore, the aim of this study was to investigate the cytoprotective mechanisms of HO-1 against H/R-induced myocardiac cell apoptosis and to explore whether the Akt signaling pathway contributed to the protection provided by HO-1. METHODS Cobalt protoporphyrin (CoPP, a pharmacologic inducer of HO-1) was employed to induce the over-expression of HO-1 before H/R in H9c2 cells. Hoechst staining and flow cytometry were used to examine the extent of apoptosis. Furthermore, the effect of HO-1 on Akt, JNK, and the expression of apoptosis-related proteins (c-JUN and Caspase-3) was determined by Western blotting. RESULTS The results showed that over-expressed HO-1 could significantly protect myocardiac cells against H/R-induced apoptosis in H9c2 cells. Furthermore, the protein expression of p‑Akt increased and of p‑JNK decreased in the H/R injury H9c2 cells when treated with CoPP. The apoptosis-related proteins c‑Jun and caspase-3 were both inhibited by over-expression of HO-1. At the same time, retreatment with zinc protoporphyrin (ZnPP, a specific inhibitor of HO-1 enzymatic activity) or LY294002 (an inhibitor of Akt1) reversed the HO-1-induced changes. CONCLUSION In summary, our results suggest that HO-1 can decrease H/R-induced myocardiac cell apoptosis; the mechanism may be related to the activation of the Akt signaling pathway and, furthermore, to the inhibition of the JNK/c-Jun/caspase-3 signaling pathway.
Collapse
Affiliation(s)
- C Li
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, 273500, Jining, Shandong, P.R. China.
| | - C Zhang
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, 273500, Jining, Shandong, P.R. China
| | - T Wang
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, 273500, Jining, Shandong, P.R. China
| | - J Xuan
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, 273500, Jining, Shandong, P.R. China
| | - C Su
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, 273500, Jining, Shandong, P.R. China
| | - Y Wang
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, 273500, Jining, Shandong, P.R. China
| |
Collapse
|
45
|
Zhang Y, Ren J. Epigenetics and obesity cardiomyopathy: From pathophysiology to prevention and management. Pharmacol Ther 2016; 161:52-66. [PMID: 27013344 DOI: 10.1016/j.pharmthera.2016.03.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Uncorrected obesity has been associated with cardiac hypertrophy and contractile dysfunction. Several mechanisms for this cardiomyopathy have been identified, including oxidative stress, autophagy, adrenergic and renin-angiotensin aldosterone overflow. Another process that may regulate effects of obesity is epigenetics, which refers to the heritable alterations in gene expression or cellular phenotype that are not encoded on the DNA sequence. Advances in epigenome profiling have greatly improved the understanding of the epigenome in obesity, where environmental exposures during early life result in an increased health risk later on in life. Several mechanisms, including histone modification, DNA methylation and non-coding RNAs, have been reported in obesity and can cause transcriptional suppression or activation, depending on the location within the gene, contributing to obesity-induced complications. Through epigenetic modifications, the fetus may be prone to detrimental insults, leading to cardiac sequelae later in life. Important links between epigenetics and obesity include nutrition, exercise, adiposity, inflammation, insulin sensitivity and hepatic steatosis. Genome-wide studies have identified altered DNA methylation patterns in pancreatic islets, skeletal muscle and adipose tissues from obese subjects compared with non-obese controls. In addition, aging and intrauterine environment are associated with differential DNA methylation. Given the intense research on the molecular mechanisms of the etiology of obesity and its complications, this review will provide insights into the current understanding of epigenetics and pharmacological and non-pharmacological (such as exercise) interventions targeting epigenetics as they relate to treatment of obesity and its complications. Particular focus will be on DNA methylation, histone modification and non-coding RNAs.
Collapse
Affiliation(s)
- Yingmei Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| |
Collapse
|
46
|
Sulforaphane prevents rat cardiomyocytes from hypoxia/reoxygenation injury in vitro via activating SIRT1 and subsequently inhibiting ER stress. Acta Pharmacol Sin 2016; 37:344-53. [PMID: 26775664 DOI: 10.1038/aps.2015.130] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/26/2015] [Indexed: 02/06/2023] Open
Abstract
AIM Sulforaphane (SFN), a natural dietary isothiocyanate, is found to exert beneficial effects for cardiovascular diseases. This study aimed to investigate the mechanisms underlying the protective effects of SFN in a model of myocardial hypoxia/reoxygenation (H/R) injury in vitro. METHODS Cultured neonatal rat cardiomyocytes pretreated with SFN were subjected to 3-h hypoxia followed by 3-h reoxygenation. Cell viability and apoptosis were detected. Caspase-3 activity and mitochondrial membrane potential (ΔΨm) was measured. The expression of ER stress-related apoptotic proteins were analyzed with Western blot analyses. Silent information regulator 1 (SIRT1) activity was determined with SIRT1 deacetylase fluorometric assay kit. RESULTS SFN (0.1-5 μmol/L) dose-dependently improved the viability of cardiomyocytes, diminished apoptotic cells and suppressed caspase-3 activity. Meanwhile, SFN significantly alleviated the damage of ΔΨm and decreased the expression of ER stress-related apoptosis proteins (GRP78, CHOP and caspase-12), elevating the expression of SIRT1 and Bcl-2/Bax ratio in the cardiomyocytes. Co-treatment of the cardiomyocytes with the SIRT1-specific inhibitor Ex-527 (1 μmol/L) blocked the SFN-induced cardioprotective effects. CONCLUSION SFN prevents cardiomyocytes from H/R injury in vitro most likely via activating SIRT1 pathway and subsequently inhibiting the ER stress-dependent apoptosis.
Collapse
|
47
|
Gurianova V, Stroy D, Ciccocioppo R, Gasparova I, Petrovic D, Soucek M, Dosenko V, Kruzliak P. Stress response factors as hub-regulators of microRNA biogenesis: implication to the diseased heart. Cell Biochem Funct 2015; 33:509-18. [PMID: 26659949 DOI: 10.1002/cbf.3151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 09/21/2015] [Accepted: 10/02/2015] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are important regulators of heart function and then an intriguing therapeutic target for plenty of diseases. The problem raised is that many data in this area are contradictory, thus limiting the use of miRNA-based therapy. The goal of this review is to describe the hub-mechanisms regulating the biogenesis and function of miRNAs, which could help in clarifying some contradictions in the miRNA world. With this scope, we analyse an array of factors, including several known agents of stress response, mediators of epigenetic changes, regulators of alternative splicing, RNA editing, protein synthesis and folding and proteolytic systems. All these factors are important in cardiovascular function and most of them regulate miRNA biogenesis, but their influence on miRNAs was shown for non-cardiac cells or some specific cardiac pathologies. Finally, we consider that studying the stress response factors, which are upstream regulators of miRNA biogenesis, in the diseased heart could help in (1) explaining some contradictions concerning miRNAs in heart pathology, (2) making the role of miRNAs in pathogenesis of cardiovascular disease more clear, and therefore, (3) getting powerful targets for its molecular therapy.
Collapse
Affiliation(s)
- Veronika Gurianova
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Dmytro Stroy
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Rachele Ciccocioppo
- Clinica Medica I; Fondazione IRCCS Policlinico San Matteo, Università degli Studi di Pavia, Italy
| | - Iveta Gasparova
- Institute of Biology, Genetics and Medical Genetics, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovak Republic
| | - Daniel Petrovic
- Institute of Histology and Embryology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Miroslav Soucek
- Second Department of Internal Medicine, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Victor Dosenko
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Peter Kruzliak
- Second Department of Internal Medicine, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovak Republic.,Laboratory of Structural Biology and Proteomics, Faculty of Pharmacy, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| |
Collapse
|
48
|
Ham O, Lee SY, Lee CY, Park JH, Lee J, Seo HH, Cha MJ, Choi E, Kim S, Hwang KC. let-7b suppresses apoptosis and autophagy of human mesenchymal stem cells transplanted into ischemia/reperfusion injured heart 7by targeting caspase-3. Stem Cell Res Ther 2015; 6:147. [PMID: 26296645 PMCID: PMC4546263 DOI: 10.1186/s13287-015-0134-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 03/17/2015] [Accepted: 07/17/2015] [Indexed: 12/15/2022] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) have therapeutic potential for the repair of myocardial injury. The efficacy of MSC therapy for myocardial regeneration mainly depends on the survival of cells after transplantation into the infarcted heart. In the transplanted regions, reactive oxygen species (ROS) can cause cell death, and this process depends on caspase activation and autophagosome formation. Methods A Software TargetScan was utilized to search for microRNAs (miRNAs) that target caspase-3 mRNA. Six candidate miRNAs including let-7b were selected and transfected into human MSCs in vitro. Expression of MEK-EKR signal pathways and autophagy-related genes were detected. Using ischemia/reperfusion model (I/R), the effect of MSCs enriched with let-7b was determined after transplantation into infarcted heart area. Miller catheter was used to evaluate cardiac function. Results Here, we report that let-7b targets caspase-3 to regulate apoptosis and autophagy in MSCs exposed to ROS. Let-7b-transfected MSCs (let-7b-MSCs) showed high expression of survival-related proteins, including p-MEK, p-ERK and Bcl-2, leading to a decrease in Annexin V/PI- and TUNEL-positive cells under ROS-rich conditions. Moreover, autophagy-related genes, including Atg5, Atg7, Atg12 and beclin-1, were significantly downregulated in let-7b-MSCs. Using a rat model of acute myocardial infarction, we found that intramyocardial injection of let-7b-MSCs markedly enhanced left ventricular (LV) function and microvessel density, in accordance with a reduced infarct size and the expression of caspase-3. Conclusions Taken together, these data indicate that let-7b may protect MSCs implanted into infarcted myocardium from apoptosis and autophagy by directly targeting caspase-3 signaling.
Collapse
Affiliation(s)
- Onju Ham
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City, 404-834, Republic of Korea.
| | - Se-Yeon Lee
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City, 404-834, Republic of Korea.
| | - Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Graduate School, Yonsei University, Seoul, 120-752, Republic of Korea.
| | - Jun-Hee Park
- Department of Integrated Omics for Biomedical Sciences, Graduate School, Yonsei University, Seoul, 120-752, Republic of Korea.
| | - Jiyun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 120-752, Republic of Korea.
| | - Hyang-Hee Seo
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 120-752, Republic of Korea.
| | - Min-Ji Cha
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City, 404-834, Republic of Korea. .,Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, 210-701, Republic of Korea.
| | - Eunhyun Choi
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City, 404-834, Republic of Korea. .,Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, 210-701, Republic of Korea.
| | - Soonhag Kim
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City, 404-834, Republic of Korea. .,Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, 210-701, Republic of Korea.
| | - Ki-Chul Hwang
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City, 404-834, Republic of Korea. .,Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, 210-701, Republic of Korea.
| |
Collapse
|
49
|
Protective Effect of Sevoflurane Postconditioning against Cardiac Ischemia/Reperfusion Injury via Ameliorating Mitochondrial Impairment, Oxidative Stress and Rescuing Autophagic Clearance. PLoS One 2015; 10:e0134666. [PMID: 26263161 PMCID: PMC4532466 DOI: 10.1371/journal.pone.0134666] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 07/13/2015] [Indexed: 01/29/2023] Open
Abstract
Background and Purpose Myocardial infarction leads to heart failure. Autophagy is excessively activated in myocardial ischemia/reperfusion (I/R) in rats. The aim of this study is to investigate whether the protection of sevoflurane postconditioning (SPC) in myocardial I/R is through restored impaired autophagic flux. Methods Except for the sham control (SHAM) group, each rat underwent 30 min occlusion of the left anterior descending coronary (LAD) followed by 2 h reperfusion. Cardiac infarction was determined by 2,3,5-triphenyltetrazolium chloride triazole (TTC) staining. Cardiac function was examined by hemodynamics and echocardiography. The activation of autophagy was evaluated by autophagosome accumulation, LC3 conversion and p62 degradation. Potential molecular mechanisms were investigated by immunoblotting, real-time PCR and immunofluorescence staining. Results SPC improved the hemodynamic parameters, cardiac dysfunction, histopathological and ultrastructural damages, and decreased myocardial infarction size after myocardial I/R injury (P < 0.05 vs. I/R group). Compared with the cases in I/R group, myocardial ATP and NAD+ content, mitochondrial function related genes and proteins, and the expressions of SOD2 and HO-1 were increased, while the expressions of ROS and Vimentin were decreased in the SPC group (P < 0.05 vs. I/R group). SPC significantly activated Akt/mTOR signaling, and inhibited the formation of Vps34/Beclin1 complex via increasing expression of Bcl2 protein (P < 0.05 vs. I/R group). SPC suppressed elevated expressions of LC3 II/I ratio, Beclin1, Atg5 and Atg7 in I/R rat, which indicated that SPC inhibited over-activation of autophagy, and promoted autophagosome clearance. Meanwhile, SPC significantly suppressed the decline of Opa1 and increases of Drp1 and Parkin induced by I/R injury (P < 0.05 vs. I/R group). Moreover, SPC maintained the contents of ATP by reducing impaired mitochondria. Conclusion SPC protects rat hearts against I/R injury via ameliorating mitochondrial impairment, oxidative stress and rescuing autophagic clearance.
Collapse
|
50
|
Regulation of skeletal muscle development and homeostasis by gene imprinting, histone acetylation and microRNA. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:309-16. [DOI: 10.1016/j.bbagrm.2015.01.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/17/2014] [Accepted: 01/10/2015] [Indexed: 12/13/2022]
|