1
|
Tang S, Wang Y, Ma T, Lu S, Huang K, Li Q, Wu M, Yang H, Zhong J. MiR-30d inhibits cardiomyocytes autophagy promoting ferroptosis after myocardial infarction. Panminerva Med 2024; 66:249-255. [PMID: 32720797 DOI: 10.23736/s0031-0808.20.03979-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The aim of this study was to investigate the effect of microRNA-30d (miR-30d) on autophagy and reveal the mechanism of autophagy promoting ferroptosis in H9C2 cells. METHODS First, we detected miR-30d expression of myocardial tissue in the sham and myocardial infarction (MI) group, and then analyzed by biochemical analysis and luciferase Genetic experiments to confirm its downstream target gene of. After using Lentivirus-ATG5 (LV-sh-ATG5) to effectively inhibit autophagy, in order to further clarify the possible mechanism of autophagy leading to ferroptosis in H9C2 cells, we have tested the relevant indicators ferroptosis. RESULTS We first found that miR-30d expression was down-regulated in myocardial tissue after MI, while autophagy increased, and autophagy was reduced when miR-30d was overexpressed, and then analyzed by biochemical analysis and luciferase Genetic experiments confirmed that ATG5 was a downstream target gene of miR-30d. After using Lentivirus-ATG5 (LV-shATG5) to effectively inhibit autophagy and up-regulate the expression of FTH1 and GSH peroxidase (GPX4) in H9C2 cells, reduce the content of MDA, increase the content of GSH, and increase the activity of GPX4, suggesting that autophagy after MI may promote ferroptosis in H9C2 cells. CONCLUSIONS The expression of miR-30d decreased in cardiomyocytes after MI and which can inhibit autophagy by binding to ATG5. Furthermore, autophagy after MI may promote ferroptosis.
Collapse
Affiliation(s)
- Shilin Tang
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yang Wang
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Tanya Ma
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Shijuan Lu
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Kang Huang
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Qiang Li
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Miao Wu
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Hui Yang
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Jianghua Zhong
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China -
| |
Collapse
|
2
|
Effects of Sevoflurane on Apoptosis of Myocardial Cells in IRI Rats. BIOMED RESEARCH INTERNATIONAL 2022; 2021:3347949. [PMID: 35005016 PMCID: PMC8741344 DOI: 10.1155/2021/3347949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 11/18/2022]
Abstract
Background Cardiomyocyte apoptosis functions essentially in ischemia/reperfusion- (I/R-) induced myocardial injury. It is suggested that autophagy is widely implicated in the regulation of cell survival and death. Sevoflurane, as a largely used inhalational general anesthetic, has been shown to have a protective effect on cardiomyocytes. However, it was yet elusive on the underlying mechanisms. Aim The objective of this study is to investigate the association of sevoflurane-mediated cardioprotective effects with autophagy regulation. Methods An in vitro hypoxia model was established in primary cardiomyocytes from fresh myocardial tissue of the rats. The apoptosis rate of myocardial cells treated with hypoxia and treated with sevoflurane was measured. Western blot and immunocytochemical assay were used to measure the protein expression. The cell proliferation rate and cell apoptosis were measured using the MTT assay and flow cytometry, respectively. Results The expression of apoptotic proteins including B cell lymphoma-2 (Bcl-2), CCAAT/enhancer-binding protein homologous protein (CHOP), glucose-regulated protein 78 (GRP78), and Bcl-2-associated X protein (BAX) in myocardium treated with sevoflurane was significantly lower than that in myocardium treated with hypoxia. The expression of adhesion proteins such as intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin in myocardium treated with sevoflurane was higher than that in myocardium treated with hypoxia, suggesting better connectivity of the myocardium. Conclusion Sevoflurane treatment reduced the apoptosis of myocardial cells after hypoxia treatment.
Collapse
|
3
|
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: 2.3] [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
|
4
|
He W, Su J, Liu D, Huang K. Mannan Oligosaccharide Could Attenuate Ochratoxin A-Induced Immunosuppression with Long-Time Exposure Instead of Immunostimulation with Short-Time Exposure. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:11461-11469. [PMID: 34542274 DOI: 10.1021/acs.jafc.1c04485] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Our previous study showed that ochratoxin A (OTA), one of the most common mycotoxins in feed, could induce immunosuppression with long-time exposure but immunostimulation with short-time exposure. However, limited studies for the control of OTA-induced two-way immune toxicity were carried out. This study explored the effects of mannan oligosaccharide (MOS), a glucomannoprotein complex with immunoregulatory capability derived from the yeast cell wall, on OTA-induced immune toxicity and its underlying mechanisms. Surprisingly, the results showed that MOS significantly attenuated immunosuppression induced by long-time OTA treatment but did not provide protection against immunostimulation induced by short-time OTA treatment on porcine alveolar macrophages (PAMs), as demonstrated by the expressions of inflammatory cytokines and the capability of migration and phagocytosis. Further, MOS increased the OTA-inhibited autophagy level and the JNK phosphorylation level on PAMs with long-time OTA treatment. In addition, the inhibition of autophagy by 3-MA or the inhibition of JNK phosphorylation by SP600125 could partly block the protective effects of MOS on OTA-induced immunosuppression. Importantly, the inhibition of JNK phosphorylation down-regulated the MOS-promoted autophagy level. In conclusion, MOS could attenuate OTA-induced immunosuppression with short-time exposure on PAMs through activating JNK-mediated autophagy but had no significant effects on OTA-induced immunostimulation with short-time exposure. Our study provides new insights into the application of MOS as an immunoregulator against mycotoxin-induced immune toxicity.
Collapse
Affiliation(s)
- Wenmiao He
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Jiarui Su
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Dandan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| |
Collapse
|
5
|
Qian Q, Xie Y. Propofol protects H9C2 cells against hypoxia/reoxygenation injury through miR-449a and NR4A2. Exp Ther Med 2021; 22:1181. [PMID: 34475971 PMCID: PMC8406901 DOI: 10.3892/etm.2021.10615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 06/25/2021] [Indexed: 12/31/2022] Open
Abstract
Propofol has been revealed to protect cardiomyocytes against myocardial ischemia injury, although the underlying mechanism remains incompletely understood. H9C2 cells were used to generate a hypoxia/reoxygenation (H/R) in vitro model for the present study. Reverse transcription-quantitative PCR and western blotting were performed to measure the expression levels of microRNA (miR)-449a and nuclear receptor subfamily 4 group A member 2 (NR4A2). The CCK-8, BrdU, EdU, and caspase-3 activity assays and western blot analysis were employed to detect cell viability, proliferation, and apoptosis. The target relationship between miR-449a and NR4A2 was verified through dual-luciferase reporter assays. The results confirmed that exposure of the cells to H/R resulted in severe cell injury. However, the presence of propofol improved cell activity by promoting cell viability and proliferation and inhibiting cell apoptosis. The beneficial effect of propofol on H/R-mediated injury could be abrogated by the inhibition of NR4A2 mediated by miR-449a. Thus, the present study demonstrated that propofol counteracted cardiomyocyte H/R injury by inhibiting miR-449a to upregulate NR4A2.
Collapse
Affiliation(s)
- Qiu Qian
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Yingxiang Xie
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
6
|
miR-378a-3p inhibits ischemia/reperfusion-induced apoptosis in H9C2 cardiomyocytes by targeting TRIM55 via the DUSP1-JNK1/2 signaling pathway. Aging (Albany NY) 2020; 12:8939-8952. [PMID: 32463795 PMCID: PMC7288954 DOI: 10.18632/aging.103106] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/09/2020] [Indexed: 12/31/2022]
Abstract
MicroRNAs (miRNAs) are involved in many pathological and biological processes, such as ischemia/reperfusion (I/R) injury by modulating gene expression. Increasing evidence indicates that miR-378a-3p might provide a potential cardioprotective effect against ischemic heart disease. Cell apoptosis is a crucial mechanism in I/R injury. As such, this study evaluated the protective effects and underlying mechanisms of action of miR-378a-3p on H9C2 cardiomyocyte apoptosis following I/R injury. We found that I/R-induced H9C2 cardiomyocytes exhibited a decrease in miR-378a-3p expression, while treatment with a miR-378a-3p mimic suppressed cell apoptosis, JNK1/2 activation, cleavage of PARP and caspase-3, and Bax/Bcl-2 ratio but increased DUSP1 expression, which subsequently inhibited JNK1/2 phosphorylation. TRIM55 was shown to be a target of miR-378a-3p and its downregulation inhibited the miR-378a-3p inhibitor-induced increase in cell apoptosis and JNK1/2 activation. TRIM55 inhibited DUSP1 protein expression through ubiquitination of DUSP1. Moreover, DUSP1 overexpression inhibited the TRIM55 overexpression-induced increase in cell apoptosis and JNK1/2 activation. The protective effect of miR-378a-3p was subsequently confirmed in a rat myocardial I/R model, as evidenced by a decrease in cardiomyocyte apoptosis of cardiomyocytes, TRIM55 expression, and JNK1/2 activation. Taken together, these results suggest that miR-378a-3p may protect against I/R-induced cardiomyocyte apoptosis via TRIM55/DUSP1/JNK signaling.
Collapse
|
7
|
Wang H, Peng X, Huang Y, Xiao Y, Wang Z, Zhan L. Propofol Attenuates Hypoxia/Reoxygenation-Induced Apoptosis and Autophagy in HK-2 Cells by Inhibiting JNK Activation. Yonsei Med J 2019; 60:1195-1202. [PMID: 31769251 PMCID: PMC6881709 DOI: 10.3349/ymj.2019.60.12.1195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/13/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022] Open
Abstract
PURPOSE The aim of this study was to investigate whether propofol could attenuate hypoxia/reoxygenation-induced apoptosis and autophagy in human renal proximal tubular cells (HK-2) by inhibiting JNK activation. MATERIALS AND METHODS HK-2 cells were treated with or without propofol or JNK inhibitor SP600125 for 1 hour and then subjected to 15 hours of hypoxia and 2 hours of reoxygenation (H/R). Cell viability and LDH release were measured with commercial kits. Cell apoptosis was evaluated by flow cytometry. The expressions of p-JNK, cleaved-caspase-3, Bcl-2, and autophagy markers LC3 and p62 were measured by Western blot or immunofluorescence. RESULTS HK-2 cells exposed to H/R insult showed higher cell injury (detected by increased LDH release and decreased cell viability), increased cell apoptosis index and expression of cleaved-caspase-3, a decrease in the expression of Bcl-2 accompanied by increased expression of p-JNK and LC3II, and a decrease in expression of p62. All of these alterations were attenuated by propofol treatment. Similar effects were provoked upon treatment with the JNK inhibitor SP600125. Moreover, the protective effects were more obvious with the combination of propofol and SP600125. CONCLUSION These results suggest that propofol could attenuate hypoxia/reoxygenation induced apoptosis and autophagy in HK-2 cells, probably through inhibiting JNK activation.
Collapse
Affiliation(s)
- Huaxin Wang
- Department of Anesthesiology, Wuhan University Renmin Hospital, Wuhan, China
| | - Xuan Peng
- Department of Anesthesiology, Wuhan University Renmin Hospital, Wuhan, China
| | - Yayi Huang
- Department of Anesthesiology, Wuhan University Renmin Hospital, Wuhan, China
| | - Yeda Xiao
- Department of Anesthesiology, Wuhan University Renmin Hospital, Wuhan, China
| | - Zhuo Wang
- Department of Anesthesiology, Wuhan University Renmin Hospital, Wuhan, China
| | - Liying Zhan
- Department of Anesthesiology, Wuhan University Renmin Hospital, Wuhan, China.
| |
Collapse
|
8
|
Guan G, Yang L, Huang W, Zhang J, Zhang P, Yu H, Liu S, Gu X. Mechanism of interactions between endoplasmic reticulum stress and autophagy in hypoxia/reoxygenation‑induced injury of H9c2 cardiomyocytes. Mol Med Rep 2019; 20:350-358. [PMID: 31115545 PMCID: PMC6580049 DOI: 10.3892/mmr.2019.10228] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 04/25/2019] [Indexed: 12/13/2022] Open
Abstract
Endoplasmic reticulum (ER) stress and autophagy are involved in myocardial ischemia-reperfusion (I/R) injury; however, their roles in this type of injury remain unclear. The present study investigated the roles of ER stress and autophagy, and their underlying mechanisms, in H9c2 cells during hypoxia/reoxygenation (H/R) injury. Cell viability was detected by CCK-8 assay. The autophagy flux was monitored with mCherry-GFP-LC3-adenovirus transfection. The expression levels of autophagy-related proteins and ER stress-related proteins were measured by western blotting. Apoptosis was detected by flow cytometry and western blotting. The results indicated that autophagy was induced, ER stress was activated and apoptosis was promoted in H9c2 cells during H/R injury. The inhibition of ER stress by 4-phenylbutyrate or C/EBP homologous protein (CHOP)-targeting small interfering RNA (siRNA) decreased autophagy and ameliorated cell apoptosis during H/R injury. Activation of autophagy by rapamycin attenuated ER stress and ameliorated cell apoptosis. Inhibition of autophagy by 3-methyladenine or Beclin1-targeting siRNA aggravated ER stress and exacerbated cell apoptosis, and activation of ER stress by thapsigargin decreased autophagy and induced cell apoptosis. Collectively, the findings of the present study demonstrated that H/R induced apoptosis and autophagy via ER stress in H9c2 cells, and that CHOP may serve an important role in ER stress-induced autophagy and apoptosis. Autophagy, as an adaptive response, was activated by ER stress and alleviated ER stress-induced cell apoptosis during H/R injury.
Collapse
Affiliation(s)
- Gaopeng Guan
- Department of Cardiology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Lei Yang
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Wenyin Huang
- Department of Cardiology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Jun Zhang
- Department of Cardiology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Puhua Zhang
- Department of Cardiology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Huan Yu
- Key Laboratory of System Bio‑Medicine of Jiangxi Province, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Shengyuan Liu
- Department of Cardiology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Xiang Gu
- Department of Cardiology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| |
Collapse
|
9
|
Qiao SG, Sun Y, Sun B, Wang A, Qiu J, Hong L, An JZ, Wang C, Zhang HL. Sevoflurane postconditioning protects against myocardial ischemia/reperfusion injury by restoring autophagic flux via an NO-dependent mechanism. Acta Pharmacol Sin 2019; 40:35-45. [PMID: 30002490 DOI: 10.1038/s41401-018-0066-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 06/05/2018] [Indexed: 12/14/2022] Open
Abstract
Volatile anesthetics improve postischemic cardiac function and reduce infarction even when administered for only a brief time at the onset of reperfusion. A recent study showed that sevoflurane postconditioning (SPC) attenuated myocardial reperfusion injury, but the underlying mechanisms remain unclear. In this study, we examined the effects of sevoflurane on nitric oxide (NO) release and autophagic flux during the myocardial ischemia/reperfusion (I/R) injury in rats in vivo and ex vivo. Male rats were subjected to 30 min ischemia and 2 h reperfusion in the presence or absence of sevoflurane (1.0 minimum alveolar concentration) during the first 15 min of reperfusion. We found that SPC significantly improved hemodynamic performance after reperfusion, alleviated postischemic myocardial infarction, reduced nicotinamide adenine dinucleotide content loss, and cytochrome c release in heart tissues. Furthermore, SPC significantly increased the phosphorylation of endothelial nitric oxide synthase (NOS) and neuronal nitric oxide synthase, and elevated myocardial NOS activity and NO production. All these effects were abolished by treatment with an NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME, 10 mg/kg, i.v.). We also observed myocardial I/R-induced accumulation of autophagosomes in heart tissues, as evidenced by increased ratios of microtubule-associated protein 1 light chain 3 II/I, up-regulation of Beclin 1 and P62, and reduced lysosome-associated membrane protein-2 expression. SPC significantly attenuated I/R-impaired autophagic flux, which were blocked by L-NAME. Moreover, pretreatment with the autophagic flux blocker chloroquine (10 mg/kg, i.p.) increased autophagosome accumulation in SPC-treated heart following I/R and blocked SPC-induced cardioprotection. The same results were also observed in a rat model of myocardial I/R injury ex vivo, suggesting that SPC protects rat hearts against myocardial reperfusion injury by restoring I/R-impaired autophagic flux via an NO-dependent mechanism.
Collapse
|
10
|
Sevoflurane prevents hypoxia/reoxygenation-induced cardiomyocyte apoptosis by inhibiting PI3KC3-mediated autophagy. Hum Cell 2018; 32:150-159. [DOI: 10.1007/s13577-018-00230-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/03/2018] [Indexed: 12/30/2022]
|
11
|
Sun B, Ou H, Ren F, Huan Y, Zhong T, Gao M, Cai H. Propofol inhibited autophagy through Ca 2+/CaMKKβ/AMPK/mTOR pathway in OGD/R-induced neuron injury. Mol Med 2018; 24:58. [PMID: 30470173 PMCID: PMC6251140 DOI: 10.1186/s10020-018-0054-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 09/24/2018] [Indexed: 11/22/2022] Open
Abstract
Background The neuroprotective role of propofol (PPF) in cerebral ischemia-reperfusion (I/R) has recently been highlighted. This study aimed to explore whether the neuroprotective mechanisms of PPF were linked to its regulation of Ca2+/CaMKKβ (calmodulin-dependent protein kinase kinase β)/AMPK (AMP-activated protein kinase)/mTOR (mammalian target of rapamycin)/autophagy pathway. Methods Cultured primary rat cerebral cortical neurons were treated with oxygen-glucose deprivation and re-oxygenation (OGD/R) to mimic cerebral I/R injury in vitro. Results Compared with the control neurons, OGD/R exposure successfully induced neuronal I/R injury. Furthermore, OGD/R exposure notably caused autophagy induction, reflected by augmented LC3-II/LC3-I ratio and Beclin 1 expression, decreased p62 expression, and increased LC3 puncta formation. Moreover, OGD/R exposure induced elevation of intracellular Ca2+ concentration ([Ca2+]i). However, PPF treatment significantly antagonized OGD/R-triggered cell injury, autophagy induction, and [Ca2+]i elevation. Further investigation revealed that both autophagy induction by rapamycin and [Ca2+]i elevation by the Ca2+ ionophore ionomycin significantly reversed the PPF-mediated amelioration of OGD/R-triggered cell injury. Importantly, ionomycin also significantly abrogated the PPF-mediated suppression of autophagy and CaMKKβ/AMPK/mTOR signaling in OGD/R-exposed neurons. Additionally, activation of CaMKKβ/AMPK/mTOR signaling abrogated the PPF-mediated autophagy suppression. Conclusion Our findings demonstrate that PPF antagonized OGD/R-triggered neuronal injury, which might be mediated, at least in part, via inhibition of autophagy through Ca2+/CaMKKβ/AMPK/mTOR pathway. Electronic supplementary material The online version of this article (10.1186/s10020-018-0054-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bei Sun
- Department of Anesthesiology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, People's Republic of China
| | - Hao Ou
- Department of Emergency and Critical Care Medicine, The Third Xiangya Hospital, Central South University, 410013, Changsha, Hunan, People's Republic of China.,Translational Medicine Center of Sepsis, Department of Pathophysiology, The Third Xiangya Hospital, Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Fei Ren
- Department of Anesthesiology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, People's Republic of China
| | - Ye Huan
- Department of Anesthesiology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, People's Republic of China
| | - Tao Zhong
- Department of Anesthesiology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, People's Republic of China
| | - Min Gao
- Department of Emergency and Critical Care Medicine, The Third Xiangya Hospital, Central South University, 410013, Changsha, Hunan, People's Republic of China.,Translational Medicine Center of Sepsis, Department of Pathophysiology, The Third Xiangya Hospital, Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Hongwei Cai
- Department of Anesthesiology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, People's Republic of China.
| |
Collapse
|