Open Access

Sevoflurane alleviates oxygen‑glucose deprivation/reoxygenation‑induced injury in HT22 cells through regulation of the PI3K/AKT/GSK3β signaling pathway

  • Authors:
    • Qiong Yu
    • Haofei Dai
    • Yinan Jiang
    • Yifeng Zha
    • Jie Zhang
  • View Affiliations

  • Published online on: February 19, 2021     https://doi.org/10.3892/etm.2021.9807
  • Article Number: 376
  • Copyright: © Yu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sevoflurane (Sev), a volatile anesthetic, has been reported to exhibit beneficial effects on different ischemia/reperfusion (I/R)‑injured organs. However, the neuroprotective effect of Sev on cerebral I/R injury is poorly understood. In the present study, the effects of Sev on HT22 cells exposed to oxygen‑glucose deprivation/reperfusion (OGD/R) injury are investigated. The present study demonstrated that OGD/R suppressed the cell viability and increased lactate dehydrogenase (LDH) release from the cells, and these effects were attenuated by Sev treatment. The results also demonstrated that Sev alleviated OGD/R‑induced cell apoptosis via flow cytometry and caspase‑3 activity determination. Biochemical analysis results revealed that Sev significantly protected against OGD/R‑induced oxidative stress by reducing ROS generation and improving antioxidant defense markers. Western blot analysis demonstrated that Sev reactivated the PI3K/AKT/glycogen synthase kinase‑3β (GSK3β) signaling pathway, which was inhibited by OGD/R. In addition, wortmannin, a selective PI3K inhibitor was used to investigate the underlying pathways. Notably, the neuroprotective effect of Sev on apoptosis and reactive oxygen species production was found to be suppressed by wortmannin. Collectively, these results demonstrated that Sev may protect neuronal cells against OGD/R‑induced injury through the activation of the PI3K/AKT/GSK3β signaling pathway. The findings from the present study provide a novel insight into understanding the neuroprotective effect of Sev on cerebral I/R injury.

Introduction

Cerebral ischemia/reperfusion (I/R) injury, one of the most common types of stroke, often causes a variety of cerebrovascular and cardiovascular diseases that lead to human disability and mortality worldwide (1,2). Nerve cell injury and apoptosis are considered the first steps in the pathogenesis of cerebral I/R injury (3). Once cerebral I/R occurs, a large number of oxygen free radicals and acute inflammatory reactions are induced, resulting in apoptosis and therefore aggravating the ischemic brain tissue injury (4). Therefore, extensive research is needed to find effective anti-apoptosis and anti-oxidative stress agents that can inhibit apoptosis and oxidative stress, ameliorating cerebral I/R injury.

Sevoflurane (Sev), a volatile anesthetic, has potential therapeutic effects on I/R-induced injury, especially towards the pulmonary and hepatic injuries (5,6). For example, Xiong et al (7) showed that Sev could attenuate ventilator-induced lung injury by suppressing pulmonary inflammation in mice. In addition, Li et al (8) found that Sev preconditioning ameliorated spinal cord I/R-induced neuronal injury by inhibiting microglial MMP-9 expression in rats. Previous research has also demonstrated the neuroprotective effect of Sev on severe cerebral ischemia in rats (9-11). For example, pretreatment with Sev improved spatial learning and memory impairment after cerebral I/R injury in rats (12). However, the underlying mechanisms responsible for the protective effects of Sev against cerebral I/R injury are still elusive.

The present study determined the protective effects of Sev in a model of neuronal HT22 cell with oxygen-glucose deprivation/reperfusion (OGD/R), and investigate the involvement of the PI3K/AKT/GSK3β signaling pathway in molecular mechanisms. The results of the present study suggest that Sev may be an improved anesthetic option for patients with cerebral I/R injury.

Materials and methods

Cell culture

The mouse hippocampal neuronal HT22 cell line was purchased from the American Type Culture Collection (ATCC) and cultured in DMEM supplemented with 10% fetal bovine serum (FBS; both from Sigma-Aldrich; Merck KGaA) with 4.5 g/l glucose, 100 U/ml penicillin and 100 µg/ml streptomycin (Sigma-Aldrich; Merck KGaA) in a humidified incubator with 5% CO2 at 37˚C. The medium was replaced every 2 days.

Induction of OGD/R injury

The OGD/R model was built as previously described (13). Briefly, the HT22 cells in 6-well plates (1x106 cells/well; Costar 3506, Corning Life Sciences) were cultured under hypoxic conditions for 6 h in glucose-free DMEM in an atmosphere of 5% CO2, 94% N2 and 1% O2 at 37˚C. Subsequently, the medium was discarded and DMEM containing 4.5 g/l glucose was added, followed by culture under normoxic conditions in a humidified 5% CO2 incubator at 37˚C for 24 h. Cells without OGD/R treatment were used as a control.

Exposure to Sev

Cells in 6-well plates (1x106 cells/well) were exposed to Sev in the carrier gas as previously described (14). Briefly, different concentrations of Sev (0, 4.1 or 8%) were delivered by a Sev vaporizer sustained in 21% O2. The control gas was defined as 21% O2. Cells in 6-well plates (1x106 cells/well) were randomly placed in sealed plastic chambers and exposed to Sev or the control gas at 4 l/min for 6 h at 37˚C, followed by induction of OGD/R injury. The concentration of Sev was monitored by an agent analyzer (AbbVie Inc.)

Drug treatment

The PI3K inhibitor, wortmannin (15) was obtained from Merck KGaA and dissolved in dimethyl sulfoxide (DMSO). HT22 cells in 6-well plates (1x106 cells/well) were pretreated with 1 µM wortmannin for 24 h, and then were exposed to 4.1% Sev for 6 h at 37˚C, followed by OGD/R stimulation. The dose of wortmannin (1 µM) was chosen as previously described (16).

Cell viability assay

At 24 h after sevo treatment, HT22 cells in 96 microplates (5x104 cells/well) were subjected to OGD/R, and then the cell viability of HT22 cells was measured with the Cell Counting Kit-8 assay (CCK-8; Beyotime Institute of Biotechnology) according to the manufacturers protocols. A total of 10 µl CCK-8 reagent in DMEM was added to the cells and incubated for another 2 h at 37˚C. Absorbances at 450 nm were measured using a microplate reader (Bio-Rad Laboratories, Inc).

Lactate dehydrogenase (LDH) assay

At 24 h after sevo treatment, HT22 cells were subjected to OGD/R, and LDH release in the HT22 cells was measured using Pierce LDH Cytotoxicity Assay Kit (cat. no. 88953; Pierce; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The OD values were measured at 490 nm using a microplate reader (Bio-Rad Laboratories, Inc).

Cell apoptosis assay

The apoptotic rate of HT22 cells was evaluated by Annexin V-FITC and PI apoptosis detection kit I (BD Pharmingen; BD Biosciences), following the manufacturer's protocol. At 24 h after sevo treatment, HT22 cells were subjected to OGD/R, and then cells were collected, washed with phosphate-buffered saline (PBS) and resuspended in binding buffer, and double-stained with Annexin V-FITC and PI for 10 min at room temperature in the dark. After incubation for 15 min at room temperature in the dark, cell apoptosis was analyzed by FACScan flow cytometer (FCM; Beckman Coulter, Inc.) and CellQuest software version 3.3 (BD Biosciences). The apoptosis rate was the sum of the Q2 and Q3 quadrants of the flow cytometry images.

Caspase-3 activity

The caspase-3 activity of HT22 cells was measured after exposure to the various treatments using the Caspase-3 Assay kit (cat. no. C10427; Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. The optical density was measured at a wavelength of 450 nm using a microplate reader (Bio-Rad Laboratories, Inc.).

Detection of reactive oxygen species (ROS), malonaldehyde (MDA), superoxide dismutase (SOD) and glutathione peroxidase (GPx)

ROS production in HT22 cells was measured using DCFH-DA (Invitrogen; Thermo Fisher Scientific, Inc.) as previously reported (17). At 24 h after sevo treatment, HT22 cells were subjected to OGD/R, and HT22 cells (1x106 cells/well) were seeded in a 6-well plate for 24 h, and then stained with 20 µmol/l DCFH at 37˚C for 30 min, and then cell images were captured using a fluorescence microscope (Olympus Corporation) and analyzed by ImageJ software (version 1.46; National Institutes of Health).

After the designated treatment of interest, the HT22 cells were lysed using lysis buffer (Beyotime Institute of Biotechnology), following by lysate collection for the detection of SOD, MDA and GPx. The MDA content was measured by thiobarbituric acid method using a Lipid Peroxidation MDA assay kit (cat. no. S0131S), the SOD activity was detected by water-soluble tetrazolium salt (WST-8) method using a SOD assay kit (cat. no. S0103) and the activity of GPx was evaluated using a Cellular GPx assay kit (cat. no. S0056), all purchased from Beyotime Institute of Biotechnology, performed according to the manufacturer's instructions.

Western blot analysis

Western blot was performed as previously described (18). Briefly, total protein was isolated from HT22 cells using RIPA lysis buffer (Beyotime Institute of Biotechnology). The concentrations of total cellular protein were quantitated using BCA assay (Pierce; Thermo Fisher Scientific, Inc.). The protein lysates (40 µg) were analyzed on an 8% SDS-PAGE gel and transferred onto PVDF membranes (EMD Millipore), followed blocking with 5% skim milk at 4˚C overnight. The membranes were probed with the following appropriate primary antibodies: Phosphorylated (phosp)-AKT (Ser473; dilution 1:500; cat. no. 4060), AKT (dilution 1:1,000; cat. no. 4691), GSK3β (dilution 1:1,000; cat. no. 5676), phosp-GSK3β (Ser9; dilution 1:1,000; cat. no. 9327) and β-actin (dilution 1:2,000; cat no.4970) overnight at 4˚C, followed by incubation with horseradish peroxidase-conjugated (HRP) antibodies (dilution 1:6,000; cat. no. 45262; Cell Signaling Technology, Inc.) for 1 h at room temperature. All antibodies were obtained from Cell Signaling Technology, Inc. β-actin served as an internal control. The protein bands were visualized using ECL detection reagent (Cytiva). The intensity of protein fragments was quantified with Bio-Rad Laboratories Quantity One software (version 3.0; Bio-Rad Laboratories, Inc.).

Statistical analysis

Statistical analysis was performed by SPSS software version 14.0 (SPSS, Inc.). All data were presented as the mean ± standard deviation. The comparisons among data were calculated by one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Sev ameliorates OGD/R-induced injury in HT22 cells

To evaluate the protective effects of Sev on the injury induced by OGD/R, HT22 cells were exposed to Sev (0, 4.1 or 8%) for 6 h, followed by induction of OGD/R injury. CCK-8 assay showed that cell viability was significantly decreased in OGD/R-induced cells, compared with that in the control group (Fig. 1A). However, Sev treatment (4.1 and 8%) significantly improved the viability of HT22 cells receiving OGD/R stimulation (Fig. 1A). The LDH assay showed that Sev treatment (4.1 and 8%) markedly decreased LDH leakage in OGD/R-induced HT22 cells, compared with the OGD/R group (Fig. 1B). Given that apoptosis is a key event in the pathogenesis of cerebral I/R injury (19), Sev treatment was investigated for its potential to affect the apoptosis in OGD/R-induced HT22 cells. As shown in Fig. 1C, OGD/R significantly increased the caspase-3 activity, compared with the control group, whereas Sev treatment attenuated the promoting effect of OGD/R on the caspase-3 activity. Moreover, it was observed that OGD/R significantly promoted the apoptosis portion of HT22 cells, whereas Sev treatment decreased the apoptosis portion of HT22 cells (Fig. 1D and E). These data indicated that Sev treatment ameliorates OGD/R-induced cell injury and apoptosis in HT22 cells.

Sev inhibits OGD/R-induced oxidative stress in HT22 cells

Oxidative stress is another key factor in brain damage after cerebral I/R (20). Previous studies suggested that cerebral I/R could cause oxidative stress and then lead to over-production of ROS. Excessive ROS promote cell death and result in brain damage. Malondialdehyde (MDA) is the parameter of oxidative stress and is the lipid peroxidation product that is used to determine the level of damage of cell membrane lipid by ROS (21). Superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) are antioxidant enzymes to protect cells from oxidative damage (22). Therefore, the detection of MDA, GSH-Px levels, and SOD activity reflect the degree of oxidative damage in cells and tissues. To examine whether Sev ameliorates OGD/R-induced cell injury by regulating oxidative stress, the markers of oxidative stress, namely ROS, MDA, SOD and GPx, were detected after OGD/R injury and Sev treatment. As shown in Fig. 2A and B, ROS and MDA production levels were significantly increased in OGD/R-treated HT22 cells compared with those in the control group. However, the increased production of ROS and MDA in OGD/R-induced HT22 cells was decreased by Sev treatment (Fig. 2A and B). In addition, it was observed that OGD/R-induced a significant decrease in the activity of SOD and GPx in HT22 cells, whereas Sev attenuated the inhibitory effects of OGD/R on the activity of SOD and GPx (Fig. 2C and D). All data suggest that Sev treatment may improve OGD/R-induced injury by suppressing oxidative stress in HT22 cells.

Sev induces phosphorylation of GSK3β via the PI3K/AKT pathway

It has been previously reported that Sev provides neuroprotection in adult animals via the PI3K/Akt/GSK3β signaling pathway, which has a key role in cerebral I/R injury (23,24). To explore the potential molecular mechanisms of Sev against OGD/R-induced cell injury in HT22 cells, western blotting analysis was performed to detect the phosp-AKT, AKT, phosp-GSK3β and GSK3β levels in OGD/R-induced cells. As shown in Fig. 3A, phosp-AKT expression was markedly downregulated in OGD/R-induced cells compared with that in control cells, whereas Sev treatment attenuated the inhibitory effect of OGD/R on the expression of phosp-AKT. Furthermore, Sev treatment ameliorated the decrease in phosp-GSK3β expression induced by OGD/R in HT22 cells (Fig. 3B). These results indicated that Sev could reactivate the PI3K/AKT/GSK3β pathway in HT22 cells after OGD/R induction.

PI3K kinase inhibitor wortmannin inhibits the protective effect of Sev on OGD/R-induced cell injury

To investigate the effects of the PI3K/AKT/GSK3β signaling pathway on the neuroprotective effect exerted by Sev, wortmannin, a selective PI3K inhibitor, was added to HT22 cells in combination with Sev for 30 min before subjecting them to OGD/R at a concentration of 10 µM. As shown in Fig. 4A, Sev treatment (4.1%) markedly promoted the cell viability in OGD/R-induced HT22 cells. However, the protection of Sev was significantly alleviated by the presence of 10 µM wortmannin, whereas wortmannin alone did not influence the cell viability of HT22 cells compared with OGD/R-induced cells alone (Fig. 4A). Additionally, the results showed that pretreatment with wortmannin partially reversed the inhibitory effect of Sev on caspase-3 activity and apoptosis in OGD/R-induced cells (Fig. 4B-D). Moreover, blockade of PI3K by wortmannin also markedly reversed the protective effect of Sev on oxidative stress induced by OGD/R (Fig. 4E-H). All these data suggested that Sev may suppress OGD/R-induced apoptosis and oxidative stress through the PI3K/AKT/GSK3β signaling pathway.

Sev-reactivated PI3K/AKT/GSK3β signaling pathway is inhibited by wortmannin in OGD/R-induced cells

To confirm whether Sev regulated the PI3K/AKT/GSK3β signaling pathway in OGD/R-induced cell injury, phosp-AKT, AKT, phosp-GSK3β and GSK3β expression levels were detected in OGD/R-induced HT22 cells treated with Sev and pretreated with wortmannin. As shown in Fig. 5A and B, Sev treatment significantly increased the levels of phosp-AKT and phosp-GSK3β compared with OGD/R-induced cells alone, but these effects were inhibited by wortmannin treatment. These data indicate that Sev alleviates OGD/R-induced cell injury through reactivating the PI3K/AKT/GSK3β signaling pathway.

Discussion

In the present study, the results demonstrated that Sev improves OGD/R-induced neuronal injury by suppressing cell apoptosis and oxidative stress. Moreover, data revealed that the PI3K/AKT/GSK3β axis is involved in the protective effects of Sev. The findings of the current study suggested that Sev has the potential to protect neurons from cerebral ischemic injury.

Sev, a volatile anesthetic with minimal pungency, low solubility and low toxicity, is used widely in anesthetic practice (25), and has been documented to play a neuroprotective role following severe cerebral ischemia. For example, Canas et al (10) showed that Sev has a neuroprotective effect in an in vitro model of ischemia-reoxygenation. Wise-Faberowski et al (26) found that Sev can attenuate OGD-induced neuronal cell death. Ye et al (19) reported that delayed application of Sev after reperfusion provides neuroprotection by activating the PI3K/Akt pathway, and Wen et al (27) demonstrated the neuroprotection of Sev against I/R-induced brain injury through the inhibition of JNK3/caspase-3 by enhancing the Akt signaling pathway. Together, the results of these previous studies make Sev an attractive agent for the preservation of neuronal function. In the present study, using an OGD/R model, Sev pretreatment was found to markedly increase cell viability and attenuate LDH leakage in HT22 cells following OGD/R. In addition, Sev inhibited OGD/R-induced apoptosis, as evidenced by the decrease in caspase-3 activity and cleaved-caspase-3 protein expression in the HT22 cells. These data suggested that Sev can efficiently attenuate OGD/R-induced neuronal cell injury and apoptosis.

It is well known that oxidative stress is an important factor in the pathogenesis of cerebral I/R injury, which is caused by increased production of ROS and decreased activity levels of scavenger enzymes, such as SOD and GPx (28). ROS can interfere with the regulation of gene expression, thus altering the relative expression levels of signaling proteins, influencing intracellular signaling cascades (29). Under pathological conditions, such as cerebral ischemia, the aberrant production of ROS can result in the oxidation of cellular components, leading to cell damage (30). Hence, investigating whether Sev plays a role in ROS production after cerebral I/R injury was a focus of the present study. Sev was found to decrease the production of ROS and MDA, and to elevate the SOD and GPx activity induced by OGD/R in HT22 cells, suggesting that Sev protects neuronal cells from OGD/R injury by inhibiting oxidative stress.

PI3K/AKT, an anti-apoptotic and pro-survival kinase signaling cascade, plays an important role in I/R induced brain injury (31-33). Activation of AKT induces the phosphorylation of GSK3β at serine 9 (34,35). In fact, the PI3K/AKT/GSK3β signaling pathway plays a key role in modulating cellular survival/death after hypoxia and I/R (36-38). Recently, Gerace et al (39) indicated that inhibition of AKT/GSK3β signaling pathway by the PI3K inhibitor LY294002 or two GSK3β inhibitors exerts a neuroprotective effect against cerebral I/R injury induced by OGD/R (39). In addition, suppression of PI3K/AKT/GSK3β signaling pathway by the PI3K inhibitor LY294002 and GSK3β inhibitor LiCl exhibited neuroprotective properties against cerebral ischaemia-induced apoptosis (40). Therefore, we hypothesize that the neuroprotective effect of Sev on cerebral I/R injury occurs through the modulation of the PI3K/AKT/GSK3β signaling pathway. Consistent with the aforementioned studies, the activation of the PI3K/Akt pathway was also demonstrated to mediate the protective effect of Sev on OGD/R-induced cell injury in the present study. However, it is worthy of note that wortmannin, a selective PI3K inhibitor, abolished the neuroprotective effect of Sev by suppressing the phosphorylation of AKT and GSK3β proteins. Collectively, these results suggested that Sev improves OGD/R-induced neuronal injury by activating the PI3K/AKT/GSK3β signaling pathway.

In summary, the results of the present study demonstrated that Sev alleviates OGD/R-induced neuronal injury through regulating the PI3K/AKT/GSK3β signaling pathway. These findings suggest that the application of Sev may help to protect neurons against cerebral I/R injury.

Acknowledgements

Not applicable.

Funding

Funding: This study was supported by Shanghai Municipal Commission of Health and Family Planning (201740008).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

QY, HD, YJ and YZ performed the experiments, analyzed the data and wrote the study. JZ conceived and designed the study and contributed reagents to the study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Feigin VL: Stroke epidemiology in the developing world. Lancet. 365:2160–2161. 2005.PubMed/NCBI View Article : Google Scholar

2 

Surinkaew P, Sawaddiruk P, Apaijai N, Chattipakorn N and Chattipakorn SC: Role of microglia under cardiac and cerebral ischemia/reperfusion (I/R) injury. Metab Brain Dis. 33:1019–1030. 2018.PubMed/NCBI View Article : Google Scholar

3 

Schrepfer E and Scorrano L: Mitofusins, from mitochondria to metabolism. Mol Cell. 61:683–694. 2016.PubMed/NCBI View Article : Google Scholar

4 

Wu H, Tang C, Tai LW, Yao W, Guo P, Hong J, Yang X, Li X, Jin Z, Ke J and Wang Y: Flurbiprofen axetil attenuates cerebral ischemia/reperfusion injury by reducing inflammation in a rat model of transient global cerebral ischemia/reperfusion. Biosci Rep. 38(BSR20171562)2018.PubMed/NCBI View Article : Google Scholar

5 

Zhang Y, Li D, Luo J, Chen S, Dou X, Han M and Zhang H: Pharmacological postconditioning with sevoflurane activates PI3K/AKT signaling and attenuates cardiopulmonary bypass-induced lung injury in dog. Life Sci. 173:68–72. 2017.PubMed/NCBI View Article : Google Scholar

6 

Lu CC, Tsai CS, Ho ST, Chen WY, Wong CS, Wang JJ, Hu OYP and Lin CY: Pharmacokinetics of sevoflurane uptake into the brain and body. Anaesthesia. 58:951–956. 2003.PubMed/NCBI View Article : Google Scholar

7 

Xiong XQ, Lin LN, Wang LR and Jin LD: Sevoflurane attenuates pulmonary inflammation and ventilator-induced lung injury by upregulation of HO-1 mRNA expression in mice. Int J Nanomedicine. 6:1075–1081. 2013.PubMed/NCBI View Article : Google Scholar

8 

Li XQ, Cao XZ, Wang J, Fang B, Tan WF and Ma H: Sevoflurane preconditioning ameliorates neuronal deficits by inhibiting microglial MMP-9 expression after spinal cord ischemia/reperfusion in rats. Mol Brain. 7(69)2014.PubMed/NCBI View Article : Google Scholar

9 

Park HP, Jeong EJ, Kim MH, Hwang JW, Lim YJ, Min SW, Kim CS and Jeon YT: Effects of sevoflurane on neuronal cell damage after severe cerebral ischemia in rats. Korean J Anesthesiol. 61:327–331. 2011.PubMed/NCBI View Article : Google Scholar

10 

Canas PT, Velly LJ, Labrande CN, Guillet BA, Sautou-Miranda V, Masmejean FM, Nieoullon AL, Gouin FM, Bruder NJ and Pisano PS: Sevoflurane protects rat mixed cerebrocortical neuronal-glial cell cultures against transient oxygen-glucose deprivation: Involvement of glutamate uptake and reactive oxygen species. Anesthesiology. 105:990–998. 2006.PubMed/NCBI View Article : Google Scholar

11 

Wang Z, Ye Z, Huang G, Wang N, Wang E and Guo Q: Sevoflurane post-conditioning enhanced hippocampal neuron resistance to global cerebral ischemia induced by cardiac arrest in rats through PI3K/Akt survival pathway. Front Cell Neurosci. 10(271)2016.PubMed/NCBI View Article : Google Scholar

12 

Hu X, Zhang Y, Li W, Liu J and Li Y: Preconditioning with sevoflurane ameliorates spatial learning and memory deficit after focal cerebral ischemia-reperfusion in rats. Int J Dev Neurosci. 31:328–333. 2013.PubMed/NCBI View Article : Google Scholar

13 

Wang G, Wang T, Zhang Y, Li F, Yu B and Kou J: Schizandrin protects against OGD/R-induced neuronal injury by suppressing autophagy: Involvement of the AMPK/mTOR pathway. Molecules. 24(3624)2019.PubMed/NCBI View Article : Google Scholar

14 

Liang H, Gu M, Yang C, Wang H, Wen X and Zhou Q: Sevoflurane inhibits invasion and migration of lung cancer cells by inactivating the p38 MAPK signaling pathway. J Anesth. 26:381–392. 2012.PubMed/NCBI View Article : Google Scholar

15 

Pyo H, Yang MS, Jou I and Joe EH: Wortmannin enhances lipopolysaccharide-induced inducible nitric oxide synthase expression in microglia in the presence of astrocytes in rats. Neurosci Lett. 346:141–144. 2003.PubMed/NCBI View Article : Google Scholar

16 

Rössler OG, Giehl KM and Thiel G: Neuroprotection of immortalized hippocampal neurones by brain-derived neurotrophic factor and Raf-1 protein kinase: Role of extracellular signal-regulated protein kinase and phosphatidylinositol 3-kinase. J Neurochem. 88:1240–1252. 2004.PubMed/NCBI View Article : Google Scholar

17 

Tang Y, Vater C, Jacobi A, Liebers C, Zou X and Stiehler M: Salidroside exerts angiogenic and cytoprotective effects on human bone marrow-derived endothelial progenitor cells via Akt/mTOR/p70S6K and MAPK signalling pathways. Br J Pharmacol. 171:2440–2456. 2014.PubMed/NCBI View Article : Google Scholar

18 

Landucci E, Lattanzi R, Gerace E, Scartabelli T, Balboni G, Negri L and Pellegrini-Giampietro DE: Prokineticins are neuroprotective in models of cerebral ischemia and ischemic tolerance in vitro. Neuropharmacology. 108:39–48. 2016.PubMed/NCBI View Article : Google Scholar

19 

Ye Z, Xia P, Cheng ZG and Guo Q: Neuroprotection induced by sevoflurane-delayed post-conditioning is attributable to increased phosphorylation of mitochondrial GSK-3β through the PI3K/Akt survival pathway. J Neurol Sci. 348:216–225. 2015.PubMed/NCBI View Article : Google Scholar

20 

Chen H, Yoshioka H, Kim GS, Jung JE, Okami N, Sakata H, Maier CM, Narasimhan P, Goeders CE and Chan PH: Oxidative stress in ischemic brain damage: Mechanisms of cell death and potential molecular targets for neuroprotection. Antioxid Redox Signal. 14:1505–1517. 2011.PubMed/NCBI View Article : Google Scholar

21 

Xu HX, Pan W, Qian JF, Liu F, Dong HQ and Liu QJ: MicroRNA-21 contributes to the puerarin-induced cardioprotection via suppression of apoptosis and oxidative stress in a cell model of ischemia/reperfusion injury. Mol Med Rep. 20:719–727. 2019.PubMed/NCBI View Article : Google Scholar

22 

Siti HN, Kamisah Y and Kamsiah J: The role of oxidative stress, antioxidants and vascular inflammation in cardiovascular disease (Review). Vascul Pharmacol. 71:40–56. 2015.PubMed/NCBI View Article : Google Scholar

23 

Lai Z, Zhang L, Su J, Cai D and Xu Q: Sevoflurane postconditioning improves long-term learning and memory of neonatal hypoxia-ischemia brain damage rats via the PI3K/Akt-mPTP pathway. Brain Res. 1630:25–37. 2016.PubMed/NCBI View Article : Google Scholar

24 

Li P, Zhang Y and Liu H: The role of Wnt/β-catenin pathway in the protection process by dexmedetomidine against cerebral ischemia/reperfusion injury in rats. Life Sci. 236(116921)2019.PubMed/NCBI View Article : Google Scholar

25 

Patel SS and Goa KL: Sevoflurane. A review of its pharmacodynamic and pharmacokinetic properties and its clinical use in general anaesthesia. Drugs. 51:658–700. 1996.PubMed/NCBI View Article : Google Scholar

26 

Wise-Faberowski L, Raizada MK and Sumners C: Desflurane and sevoflurane attenuate oxygen and glucose deprivation-induced neuronal cell death. J Neurosurg Anesthesiol. 15:193–199. 2003.PubMed/NCBI View Article : Google Scholar

27 

Wen XR, Fu YY, Liu HZ, Wu J, Shao XP, Zhang XB, Tang M, Shi Y, Ma K, Zhang F, et al: Neuroprotection of sevoflurane against ischemia/reperfusion-induced brain injury through inhibiting JNK3/caspase-3 by enhancing akt signaling pathway. Mol Neurobiol. 53:1661–1671. 2016.PubMed/NCBI View Article : Google Scholar

28 

Coyle JT and Puttfarcken P: Oxidative stress, glutamate, and neurodegenerative disorders. Science. 262:689–695. 1993.PubMed/NCBI View Article : Google Scholar

29 

Shen MH, Zhang CB, Zhang JH and Li PF: Electroacupuncture attenuates cerebral ischemia and reperfusion injury in middle cerebral artery occlusion of rat via modulation of apoptosis, inflammation, oxidative stress, and excitotoxicity. Evid Based Complement Alternat Med. 2016(9438650)2016.PubMed/NCBI View Article : Google Scholar

30 

Sosunov SA, Ameer X, Niatsetskaya ZV, Utkina-Sosunova I, Ratner VI and Ten VS: Isoflurane anesthesia initiated at the onset of reperfusion attenuates oxidative and hypoxic-ischemic brain injury. PLoS One. 10(e0120456)2015.PubMed/NCBI View Article : Google Scholar

31 

Chen L, Wei X, Hou Y, Liu X, Li S, Sun B, Liu X and Liu H: Tetramethylpyrazine analogue CXC195 protects against cerebral ischemia/reperfusion-induced apoptosis through PI3K/Akt/GSK3β pathway in rats. Neurochem Int. 66:27–32. 2014.PubMed/NCBI View Article : Google Scholar

32 

Liu H, Liu X, Wei X, Chen L, Xiang Y, Yi F and Zhang X: Losartan, an angiotensin II type 1 receptor blocker, ameliorates cerebral ischemia-reperfusion injury via PI3K/Akt-mediated eNOS phosphorylation. Brain Res Bull. 89:65–70. 2012.PubMed/NCBI View Article : Google Scholar

33 

Zhang J, Deng Z, Liao J, Song C, Liang C, Xue H, Wang L, Zhang K and Yan G: Leptin attenuates cerebral ischemia injury through the promotion of energy metabolism via the PI3K/Akt pathway. J Cereb Blood Flow Metab. 33:567–574. 2013.PubMed/NCBI View Article : Google Scholar

34 

Martin M, Rehani K, Jope RS and Michalek SM: Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3. Nat Immunol. 6:777–784. 2005.PubMed/NCBI View Article : Google Scholar

35 

Jope RS and Johnson GVW: The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci. 29:95–102. 2004.PubMed/NCBI View Article : Google Scholar

36 

Arslan F, Lai RC, Smeets MB, Akeroyd L, Choo A, Aguor ENE, Timmers L, van Rijen HV, Doevendans PA, Pasterkamp G, et al: Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 10:301–312. 2013.PubMed/NCBI View Article : Google Scholar

37 

Li Y, Zhu W, Tao J, Xin P, Liu M, Li J and Wei M: Fasudil protects the heart against ischemia-reperfusion injury by attenuating endoplasmic reticulum stress and modulating SERCA activity: The differential role for PI3K/Akt and JAK2/STAT3 signaling pathways. PLoS One. 7(e48115)2012.PubMed/NCBI View Article : Google Scholar

38 

Zhang JF, Zhang L, Shi LL, Zhao ZH, Xu H, Liang F, Li HB, Zhao Y, Xu X, Yang K and Tian YF: Parthenolide attenuates cerebral ischemia/reperfusion injury via Akt/GSK-3β pathway in PC12 cells. Biomed Pharmacother. 89:1159–1165. 2017.PubMed/NCBI View Article : Google Scholar

39 

Gerace E, Scartabelli T, Pellegrini-Giampietro DE and Landucci E: Tolerance induced by (S)-3,5-dihydroxyphenylglycine postconditioning is mediated by the PI3K/Akt/GSK3β signalling pathway in an in vitro model of cerebral ischemia. Neuroscience. 433:221–229. 2020.PubMed/NCBI View Article : Google Scholar

40 

Lee DH, Lee YJ and Kwon KH: Neuroprotective effects of astaxanthin in oxygen-glucose deprivation in SH-SY5Y cells and global cerebral ischemia in rat. J Clin Biochem Nutr. 47:121–129. 2010.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2021
Volume 21 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu Q, Dai H, Jiang Y, Zha Y and Zhang J: Sevoflurane alleviates oxygen‑glucose deprivation/reoxygenation‑induced injury in HT22 cells through regulation of the PI3K/AKT/GSK3β signaling pathway. Exp Ther Med 21: 376, 2021
APA
Yu, Q., Dai, H., Jiang, Y., Zha, Y., & Zhang, J. (2021). Sevoflurane alleviates oxygen‑glucose deprivation/reoxygenation‑induced injury in HT22 cells through regulation of the PI3K/AKT/GSK3β signaling pathway. Experimental and Therapeutic Medicine, 21, 376. https://doi.org/10.3892/etm.2021.9807
MLA
Yu, Q., Dai, H., Jiang, Y., Zha, Y., Zhang, J."Sevoflurane alleviates oxygen‑glucose deprivation/reoxygenation‑induced injury in HT22 cells through regulation of the PI3K/AKT/GSK3β signaling pathway". Experimental and Therapeutic Medicine 21.4 (2021): 376.
Chicago
Yu, Q., Dai, H., Jiang, Y., Zha, Y., Zhang, J."Sevoflurane alleviates oxygen‑glucose deprivation/reoxygenation‑induced injury in HT22 cells through regulation of the PI3K/AKT/GSK3β signaling pathway". Experimental and Therapeutic Medicine 21, no. 4 (2021): 376. https://doi.org/10.3892/etm.2021.9807