1
|
Ma H, Hou T, Wu J, Zhao J, Cao H, Masula M, Wang J. Sevoflurane postconditioning attenuates cardiomyocytes hypoxia/reoxygenation injury via PI3K/AKT pathway mediated HIF-1α to regulate the mitochondrial dynamic balance. BMC Cardiovasc Disord 2024; 24:280. [PMID: 38811893 PMCID: PMC11134705 DOI: 10.1186/s12872-024-03868-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 03/30/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (I/RI) is a major cause of perioperative cardiac-related adverse events and death. Studies have shown that sevoflurane postconditioning (SpostC), which attenuates I/R injury and exerts cardioprotective effects, regulates mitochondrial dynamic balance via HIF-1α, but the exact mechanism is unknown. This study investigates whether the PI3K/AKT pathway in SpostC regulates mitochondrial dynamic balance by mediating HIF-1α, thereby exerting myocardial protective effects. METHODS The H9C2 cardiomyocytes were cultured to establish the hypoxia-reoxygenation (H/R) model and randomly divided into 4 groups: Control group, H/R group, sevoflurane postconditioning (H/R + SpostC) group and PI3K/AKT blocker (H/R + SpostC + LY) group. Cell survival rate was determined by CCK-8; Apoptosis rate was determined by flow cytometry; mitochondrial membrane potential was evaluated by Mito Tracker™ Red; mRNA expression levels of AKT, HIF-1α, Opa1and Drp1 were detected by quantitative real-time polymerase chain reaction (qRT-PCR); Western Blot assay was used to detect the protein expression levels of AKT, phosphorylated AKT (p-AKT), HIF-1α, Opa1 and Drp1. RESULTS Compared with the H/R group, the survival rate of cardiomyocytes in the H/R + SpostC group increased, the apoptosis rate decreased and the mitochondrial membrane potential increased. qRT-PCR showed that the mRNA expression of HIF-1α and Opa1 were higher in the H/R + SpostC group compared with the H/R group, whereas the transcription level of Drp1 was lower in the H/R + SpostC group. In the H/R + SpostC + LY group, the mRNA expression of HIF-1α was lower than the H/R + SpostC group. There was no difference in the expression of Opa1 mRNA between the H/R group and the H/R + SpostC + LY group. WB assay results showed that compared with the H/R group, the protein expression levels of HIF-1α, Opa1, P-AKT were increased and Drp1 protein expression levels were decreased in the H/R + SpostC group. HIF-1α, P-AKT protein expression levels were decreased in the H/R + SpostC + LY group compared to the H/R + SpostC group. CONCLUSION SpostC mediates HIF-1α-regulated mitochondrial fission and fusion-related protein expression to maintain mitochondrial dynamic balance by activating the PI3K/AKT pathway and increasing AKT phosphorylation, thereby attenuating myocardial I/R injury.
Collapse
MESH Headings
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Animals
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/enzymology
- Sevoflurane/pharmacology
- Signal Transduction
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/prevention & control
- Myocardial Reperfusion Injury/genetics
- Myocardial Reperfusion Injury/enzymology
- Mitochondrial Dynamics/drug effects
- Cell Line
- Rats
- Apoptosis/drug effects
- Phosphatidylinositol 3-Kinase/metabolism
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondria, Heart/enzymology
- Membrane Potential, Mitochondrial/drug effects
- Cell Hypoxia
- Dynamins/metabolism
- Dynamins/genetics
- GTP Phosphohydrolases/metabolism
- GTP Phosphohydrolases/genetics
- Phosphoinositide-3 Kinase Inhibitors/pharmacology
- Cytoprotection
- Ischemic Postconditioning
- Phosphorylation
Collapse
Affiliation(s)
- Haiping Ma
- The First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - Tianliang Hou
- The First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - Jianjiang Wu
- The First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - Jiyao Zhao
- The First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - Haoran Cao
- The First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - Maisitanguli Masula
- The First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - Jiang Wang
- The First Affiliated Hospital of Xinjiang Medical University, 393 Xinyi Road, Xinjiang Uygur Autonomous Region, Urumqi, 830000, China.
| |
Collapse
|
2
|
Chen W, Ma M, Song Y, Hua Y, Jia H, Liu J, Wang Y. Exercise Attenuates Myocardial Ischemia-Reperfusion Injury by Regulating Endoplasmic Reticulum Stress and Mitophagy Through M 2 Acetylcholine Receptor. Antioxid Redox Signal 2024; 40:209-221. [PMID: 37294203 DOI: 10.1089/ars.2022.0168] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Aims: Adaptive changes in the heart by exercise have been shown to reduce the risk of cardiovascular disease, and M2 Acetylcholine receptor (M2AChR), a receptor abundantly present on cardiac parasympathetic nerves, is closely associated with the development of cardiovascular disease. The present study intends to investigate whether exercise can regulate endoplasmic reticulum stress (ERS) and mitophagy through M2AChR to resist myocardial ischemia-reperfusion (I/R) injury and to elucidate its mechanism of action. Results: Exercise enhanced parasympathetic nerve function and increased myocardial M2AChR protein expression in I/R rats. In addition, it promoted the protein expression of MFN2 and inhibited the expression of Drp1, Chop, PINK1/Parkin, and PERK/eIF2α/ATF4 signaling pathways, effectively reducing mitophagy, ERS, and apoptosis. At the cellular level, 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) reduced hypoxia/reoxygenation (H/R)-induced ERS through the downregulated expression of PERK/eIF2α/ATF4 pathway proteins in H9C2 cardiomyocytes. When intervened with M2AChR inhibitors, the levels of ERS and phosphorylation levels of the PERK/eIF2α/ATF4 pathway were increased in H/R cells. Innovation and Conclusion: Exercise intervention activated the parasympathetic state in rats. It inhibited myocardial mitophagy and ERS levels, and reduced myocardial apoptosis through M2AChR, thereby resisting I/R-induced myocardial injury and improving cardiac function. Antioxid. Redox Signal. 40, 209-221.
Collapse
Affiliation(s)
- Wei Chen
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
- School of Educational Science, Shaanxi University of Technology, Hanzhong, China
| | - Mei Ma
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Yinping Song
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Yijie Hua
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hao Jia
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Jiankang Liu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Youhua Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
3
|
Upregulated microRNA-210-3p improves sevoflurane-induced protective effect on ventricular remodeling in rats with myocardial infarction by inhibiting ADCY9. Funct Integr Genomics 2022; 22:279-289. [PMID: 34988676 DOI: 10.1007/s10142-021-00816-6] [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: 07/12/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 11/04/2022]
Abstract
Myocardial infarction (MI) is a significant cause of death and disability, and sevoflurane (sevo) can protect myocardium in clinic. We aim to assess the effects of miR-210-3p on MI rats undergoing sevo treatment with the involvement of adenylyl cyclase type 9 (ADCY9). Rat MI models were constructed by ligation of the left anterior descending, and the modeled rats were respectively treated with sevo, miR-210-3p agomir, antagomir, or overexpressed ADCY9. Then, miR-210-3p and ADCY9 expression, cardiac function, myocardial injury and fibrosis, and cardiomyocyte apoptosis in rats were evaluated. Target relation between miR-210-3p and ADCY9 was detected. miR-210-3p was downregulated while ADCY9 was upregulated in MI rats. Sevo was able to promote cardiac function and attenuate myocardial injury and fibrosis, as well as cardiomyocyte apoptosis in MI rats. These effects of sevo were strengthened by miR-210-3p elevation but abolished by miR-210-3p inhibition in MI rats. The role of elevated miR-210-3p in MI rats was reversed by overexpression of ADCY9. Upregulated miR-210-3p improves sevo-induced protective effect on ventricular remodeling in rats with MI through inhibiting ADCY9.
Collapse
|
4
|
BIAN W, JIAO F, LI G, CHEN W. Fibrinogen-like protein 2 aggravates myocardial ischemia/reperfusion injury in mice following sevoflurane anesthetic through ROS production by PPAR. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.51021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Wen BIAN
- Shandong Provincial ENT Hospital, PR China
| | | | - Guiting LI
- Shandong Provincial ENT Hospital, PR China
| | - Wei CHEN
- Shandong Provincial ENT Hospital, PR China
| |
Collapse
|
5
|
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
|
6
|
Inactivation of TOPK Caused by Hyperglycemia Blocks Diabetic Heart Sensitivity to Sevoflurane Postconditioning by Impairing the PTEN/PI3K/Akt Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6657529. [PMID: 33986917 PMCID: PMC8093075 DOI: 10.1155/2021/6657529] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 01/14/2023]
Abstract
The cardioprotective effect of sevoflurane postconditioning (SPostC) is lost in diabetes that is associated with cardiac phosphatase and tensin homologue on chromosome 10 (PTEN) activation and phosphoinositide 3-kinase (PI3K)/Akt inactivation. T-LAK cell-originated protein kinase (TOPK), a mitogen-activated protein kinase- (MAPKK-) like serine/threonine kinase, has been shown to inactivate PTEN (phosphorylated status), which in turn activates the PI3K/Akt signaling (phosphorylated status). However, the functions of TOPK and molecular mechanism underlying SPostC cardioprotection in nondiabetes but not in diabetes remain unknown. We presumed that SPostC exerts cardioprotective effects by activating PTEN/PI3K/Akt through TOPK in nondiabetes and that impairment of TOPK/PTEN/Akt blocks diabetic heart sensitivity to SPostC. We found that in the nondiabetic C57BL/6 mice, SPostC significantly attenuated postischemic infarct size, oxidative stress, and myocardial apoptosis that was accompanied with enhanced p-TOPK, p-PTEN, and p-Akt. These beneficial effects of SPostC were abolished by either TOPK kinase inhibitor HI-TOPK-032 or PI3K/Akt inhibitor LY294002. Similarly, SPostC remarkably attenuated hypoxia/reoxygenation-induced cardiomyocyte damage and oxidative stress accompanied with increased p-TOPK, p-PTEN, and p-Akt in H9c2 cells exposed to normal glucose, which were canceled by either TOPK inhibition or Akt inhibition. However, either in streptozotocin-induced diabetic mice or in H9c2 cells exposed to high glucose, the cardioprotective effect of SPostC was canceled, accompanied by increased oxidative stress, decreased TOPK phosphorylation, and impaired PTEN/PI3K/Akt signaling. In addition, TOPK overexpression restored posthypoxic p-PTEN and p-Akt and decreased cell death and oxidative stress in H9c2 cells exposed to high glucose, which was blocked by PI3K/Akt inhibition. In summary, SPostC prevented myocardial ischemia/reperfusion injury possibly through TOPK-mediated PTEN/PI3K/Akt activation and impaired activation of this signaling pathway may be responsible for the loss of SPostC cardioprotection by SPostC in diabetes.
Collapse
|
7
|
Sevoflurane protects cardiomyocytes against hypoxia/reperfusion injury via LINC01133/miR-30a-5p axis. Biosci Rep 2021; 40:226934. [PMID: 33175096 PMCID: PMC7692536 DOI: 10.1042/bsr20200713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 10/21/2020] [Accepted: 11/02/2020] [Indexed: 12/28/2022] Open
Abstract
Previous studies failed to elucidate the detailed mechanisms of anesthetic preconditioning as a protective approach against ischemic/reperfusion (I/R) injury in cells. The present study mainly centered on discovering the mechanisms of Sevoflurane (Sev) in preventing cardiomyocytes against I/R injury. Human cardiomyocyte AC16 cell line was used to simulate I/R injury based on a hypoxia/reperfusion (H/R) model. After Sev treatment, cell viability and apoptosis were detected by MTT assay and flow cytometry, respectively. Lactate dehydrogenase (LDH) content was measured using an LDH Detection Kit. Relative mRNA and protein expressions of LINC01133, miR-30a-5p and apoptosis-related proteins were detected using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot as needed. Target gene of miR-30a-5p and their potential binding sites were predicted using Starbase and confirmed by dual-luciferase reporter assay. Cell behaviors were assessed again after miR-30a-5p and LINC01133 transfection. Sev could improve cell viability, reduce LDH leakage, and down-regulate the expressions of apoptosis-related proteins (Bax, cleaved caspase-3 and cleaved caspase-9) and LINC01133 as well as up-regulate miR-30a-5p and Bcl-2 expressions in H/R cells. MiR-30a-5p was the target of LINC01133, and up-regulating miR-30a-5p enhanced the effects of Sev in H/R cells, with a suppression on H/R-induced activation of the p53 signaling pathway. However, up-regulating LINC01133 reversed the enhancing effects of miR-30a-5p on Sev pretreatment in H/R cells. Sev could protect cardiomyocytes against H/R injury through the miR-30a-5p/LINC01133 axis, which may provide a possible therapeutic method for curing cardiovascular I/R injury.
Collapse
|
8
|
Ma H, Li Y, Hou T, Li J, Yang L, Guo H, Li L, Xin M, Gong Z. Sevoflurane Postconditioning Attenuates Hypoxia/Reoxygenation Injury of Cardiomyocytes Under High Glucose by Regulating HIF-1α/MIF/AMPK Pathway. Front Pharmacol 2021; 11:624809. [PMID: 33692685 PMCID: PMC7938236 DOI: 10.3389/fphar.2020.624809] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022] Open
Abstract
Subject: Cardiovascular disease, as a very common and serious coexisting disease in diabetic patients, and is one of the risk factors that seriously affect the prognosis and complications of surgical patients. Previous studies have shown that sevoflurane post-conditioning (SPostC) exerts a protective effect against myocardial ischemia/reperfusion injury by HIF-1α, but the protective effect is weakened or even disappeared under hyperglycemia. This study aims to explore whether regulating the HIF-1α/MIF/AMPK signaling pathway can restore the protective effect and reveal the mechanism of SPostC on cardiomyocyte hypoxia/reoxygenation injury under high glucose conditions. Methods: H9c2 cardiomyocytes were cultured in normal and high-concentration glucose medium to establish a hypoxia/reoxygenation (H/R) injury model of cardiomyocytes. SPostC was performed with 2.4% sevoflurane for 15 min before reoxygenation. Cell damage was determined by measuring cell viability, lactate dehydrogenase activity, and apoptosis; Testing cell energy metabolism by detecting reactive oxygen species (ROS) generation, ATP content and mitochondrial membrane potential; Analysis of the change of HIF-1α, MIF and AMPKα mRNA expression by RT-PCR. Western blotting was used to examine the expression of HIF-1α, MIF, AMPKα and p-AMPKα proteins. HIF-1α and MIF inhibitors and agonists were administered 40 min before hypoxia. Results: 1) SPostC exerts a protective effect by increasing cell viability, reducing LDH levels and cell apoptosis under low glucose (5 μM) after undergoing H/R injury; 2) High glucose concentration (35 μM) eliminated the cardioprotective effect of SPostC, which is manifested by a significantly decrease in the protein and mRNA expression level of the HIF-1α/MIF/AMPK signaling pathway, accompanied by decreased cell viability, increased LDH levels and apoptosis, increased ROS production, decreased ATP synthesis, and decreased mitochondrial membrane potential; 3. Under high glucose (35 μM), the expression levels of HIF-1α and MIF were up-regulated by using agonists, which can significantly increase the level of p-AMPKα protein, and the cardioprotective effect of SPostC was restored. Conclusion: The signal pathway of HIF-1α/MIF/AMPK of H9c2 cardiomyocytes may be the key point of SPostC against H/R injure. The cardioprotective of SPostC could be restored by upregulating the protein expression of HIF-1α and MIF under hyperglycemia.
Collapse
Affiliation(s)
- Haiping Ma
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yongjie Li
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Tianliang Hou
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jing Li
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Long Yang
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hai Guo
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Lili Li
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Mingxiu Xin
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhongcheng Gong
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
9
|
Tan DX, Chen XX, Bai TZ, Zhang J, Li ZF. RETRACTED: Sevoflurane up-regulates microRNA-204 to ameliorate myocardial ischemia/reperfusion injury in mice by suppressing Cotl1. Life Sci 2020; 259:118162. [PMID: 32730836 DOI: 10.1016/j.lfs.2020.118162] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figures 4C+E and 7E, which appear to have a similar phenotype as seen in many other publications, as detailed here: https://pubpeer.com/publications/CE1E814DD630D160BEEBFC2842FE45; and here: https://docs.google.com/spreadsheets/d/1r0MyIYpagBc58BRF9c3luWNlCX8VUvUuPyYYXzxWvgY/edit#gid=262337249. The journal requested that the corresponding author comment on these concerns and provide the raw data. The authors did not respond to this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Dian-Xiang Tan
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang 421001, Hunan, China.
| | - Xiao-Xi Chen
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang 421001, Hunan, China.
| | - Tai-Zhu Bai
- Department of Cardiovascular Medicine, Hengyang City Central Hospital, Hengyang 421001, Hunan, China.
| | - Juan Zhang
- Department of Anesthesiology, Hengyang City Central Hospital, Hengyang 421001, Hunan, China.
| | - Zhen-Fa Li
- Department of General Surgery, Hengyang City Central Hospital, Hengyang 421001, Hunan, China.
| |
Collapse
|
10
|
Wang F, Tong H. Precondition of sevoflurane upregulates TIMP3 expression to alleviate myocardial ischemia/reperfusion injury. Perfusion 2020; 36:717-723. [PMID: 33016228 DOI: 10.1177/0267659120960306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Previous studies have pointed out that sevoflurane (Sef) preconditioning could relieve myocardial ischemia/reperfusion (I/R) injury, but the mechanisms is still unknown. METHODS C57BL/6 mice model of myocardial I/R injury was established to evaluate the function of Sef. Briefly, Sef was inhaled before I/R operation. The levels of TIMP3, oxidative damage-related factors, and mitogen activated protein kinases (MAPKs) pathway-related factors were measured by qRT-PCR and western blot. Myocardial infarction (MI) area was detected by triphenyl tetrazolium chloride (TTC) staining assay. RESULTS Sef preconditioning reduced MI area in myocardial I/R injury mice and upregulated TIMP3 expression in myocardial tissues of I/R mice. In addition, downregulation of TIMP3 reversed the alleviating effects of Sef pretreatment on myocardial oxidative damage and inhibited the effect of Sef pretreatment on MAPKs pathway activity. CONCLUSION Sef preconditioning ameliorated myocardial I/R injury by modulating MAPKs pathway activity via upregulating TIMP3.
Collapse
Affiliation(s)
- Fen Wang
- Department of Cardiovascular Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Hua Tong
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
11
|
Ma M, Li R, Sun W, Wang Q, Yu H, Yu H. Sevoflurane preconditioning inhibits cardiomyocyte injury induced by oxygen‑glucose deprivation by modulating TXNIP. Int J Mol Med 2020; 46:889-897. [PMID: 32626926 DOI: 10.3892/ijmm.2020.4639] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 05/21/2020] [Indexed: 11/05/2022] Open
Abstract
The thioredoxin interaction protein (TXNIP) has been reported to be closely related to cell oxidative stress, apoptosis and inflammation. TXNIP is involved in the regulation of oxidative stress in lung and renal injury. However, it is unclear as to whether it participates in the protective effects of sevoflurane preconditioning in cardiomyocyte injury caused by oxidative stress in ischemia. In the present study, H9c2 cardiomyocytes were cultured with 0, 1.5, 2, 3.5, 5 or 6% sevoflurane for 3 h, followed by exposure to oxygen and glucose deprivation. The results demonstrated that oxygen and glucose deprivation induced an increase in TXNIP expression, lactate dehydrogenase (LDH) release, caspase‑3 activity, reactive oxygen species and malondialdehyde production. Preconditioning of the H9c2 cells with 3.5% sevoflurane suppressed TXNIP expression, LDH leakage, caspase‑3 activity, reactive oxygen species and malondialdehyde production, and it promoted cell viability. TXNIP overexpression reversed the effects of 3.5% sevoflurane preconditioning on caspase‑3 activity, reactive oxygen production and cell viability. Furthermore, TXNIP modulated p27 expression via PKB (protein kinase B/AKT) phosphorylation following preconditioning with 3.5% sevoflurane, and oxygen and glucose deprivation. On the whole these findings indicated that sevoflurane preconditioning protected the H9c2 cells against injury induced by oxygen and glucose deprivation by modulating TXNIP, AKT activation and p27 signaling.
Collapse
Affiliation(s)
- Meina Ma
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Rui Li
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Wenbo Sun
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Qi Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Hong Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Hongmei Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|
12
|
Atrial fibrillation promotion in a rat model of heart failure induced by left ventricle radiofrequency ablation. IJC HEART & VASCULATURE 2018; 21:22-28. [PMID: 30258978 PMCID: PMC6153117 DOI: 10.1016/j.ijcha.2018.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/11/2018] [Indexed: 12/19/2022]
Abstract
Background Atrial fibrillation (AF) frequently coexists with congestive heart failure (CHF). The increased susceptibility to AF in CHF has been attributed to a variety of structural and electrophysiological changes in the atria, particularly dilation and interstitial fibrosis. We evaluated atrial remodeling and AF vulnerability in a rat model of CHF induced by left ventricle (LV) radiofrequency (RF) ablation. Methods Wistar rats were divided into 3 groups: RF-induced CHF (Ab, n = 36), CHF animals treated with spironolactone (AbSpi, n = 20) and sham controls (Sham, n = 29). After 12 weeks, animals underwent echocardiographic and electrophysiological evaluation and were sacrificed for histological (atrial fibrosis) and Western blotting (TGF-β1, collagen I/III, connexin 43 and CaV1.2) analysis. Results Mild LV dysfunction and marked atrial enlargement were noted in both ablated groups. AF inducibility (episodes ≥2 s) increased in the Ab group compared to sham animals (31/36, 86%; vs. 15/29, 52%; p = 0.005), but did not differ from the AbSpi group (16/20, 80%; p = NS). Sustained AF (>30 s) was also more frequent in the Ab group compared to shams (56% vs. 28%; p = 0.04). Spironolactone reduced atrial fibrosis (p < 0.01) as well as TGF-β1 (p < 0.01) and collagen I/III (p < 0.01) expression but did not affect connexin 43 and CaV1.2 expression. Conclusions Rats with RF-induced CHF exhibit pronounced atrial structural remodeling and enhanced AF vulnerability. This model may be useful for studying AF substrate in CHF.
Collapse
|
13
|
Wenlan L, Zhongyuan X, Shaoqing L, Liying Z, Bo Z, Min L. MiR-34a-5p mediates sevoflurane preconditioning induced inhibition of hypoxia/reoxygenation injury through STX1A in cardiomyocytes. Biomed Pharmacother 2018; 102:153-159. [PMID: 29554593 DOI: 10.1016/j.biopha.2018.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/27/2018] [Accepted: 03/02/2018] [Indexed: 01/11/2023] Open
Abstract
Anesthetic preconditioning is a cellular protective approach whereby exposure to a volatile anesthetic renders cardio injury. Sevoflurane preconditioning has been shown to exhibit cardio protective effect on hypoxia/reoxygenation (H/R) injury, but the underlying mechanism is unclear. Syntaxin 1A (STX1A), an important regulator in cardio disease, was predicted to be the target gene of microRNA-34a-5p (miR-34a-5p). The current research was designed to delineate the role of miR-34a-5p in regulating sevoflurane preconditioning in cardiomyocytes injury. In this study, the results demonstrated that the expression of STX1A was significantly increased, while miR-34a-5p was dramatically decreased in sev-preconditioning H9c2 cells as compared with cells only under H/R stimulation. Moreover, miR-34a-5p regulated the protective effect of sev-preconditioning in injured H9c2 cells by mediating cell proliferation and cell apoptosis. Additionally, the luciferase report confirmed the targeting reaction between STX1A and miR-34a-5p. Taken together, our study suggested that miR-34a-5p regulated sev-preconditioning induced inhibition of hypoxia/reoxygenation injury through mediating STX1A, provided a potential therapeutic target for anesthetic protection in cardio disease.
Collapse
Affiliation(s)
- Li Wenlan
- Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xia Zhongyuan
- Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Lei Shaoqing
- Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhan Liying
- Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhao Bo
- Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Liu Min
- Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
14
|
Sevoflurane Ameliorates Myocardial Cell Injury by Inducing Autophagy via the Deacetylation of LC3 by SIRT1. Anal Cell Pathol (Amst) 2017; 2017:6281285. [PMID: 29104855 PMCID: PMC5635469 DOI: 10.1155/2017/6281285] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/27/2017] [Indexed: 11/24/2022] Open
Abstract
Misfolded and aberrant proteins have been found to be associated with myocardial cell injury. Thus, increased clearance of misfolded or aggregated proteins via autophagy might be a potential option in preventing myocardial cell injury. Sevoflurane may ameliorate myocardial cell injury by affecting sirtuin 1- (SIRT1-) mediated autophagy. Rat models with myocardial cell injury were induced by limb ischemia reperfusion. The model rats received different treatments: sevoflurane, nicotinamide, and autophagy inhibitor 3-methyladenine (3-MA). Autophagy was observed by SEM. The levels of SIRT1 and microtubule-associated protein 1A/1B-light chain 3 (LC3) were measured. Present findings demonstrated that limb ischemia reperfusion induced autophagy. Sevoflurane increased the level of SIRT1, which deacetylated LC3 and further increased autophagic rates. On the other hand, the autophagy was inhibited by sevoflurane and or the inhibitors of SIRT1 and LC3. Present results demonstrated a novel molecular mechanism by which sevoflurane induced autophagy by increasing the level of SIRT1 and reducing the acetylation of LC3.
Collapse
|
15
|
Sevoflurane pretreatment inhibits the myocardial apoptosis caused by hypoxia reoxygenation through AMPK pathway: An experimental study. ASIAN PAC J TROP MED 2017; 10:148-151. [DOI: 10.1016/j.apjtm.2017.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/17/2016] [Accepted: 01/16/2017] [Indexed: 11/19/2022] Open
|