Open Access

MicroRNA‑217 inhibition relieves cerebral ischemia/reperfusion injury by targeting SIRT1

  • Authors:
    • Gaofeng Rao
    • Wenfu Zhang
    • Shegeng Song
  • View Affiliations

  • Published online on: May 31, 2019     https://doi.org/10.3892/mmr.2019.10317
  • Pages: 1221-1229
  • Copyright: © Rao 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

MicroRNAs (miRs) have been proposed to be involved in the pathological processes of cerebral ischemia/reperfusion (CIR) injury. The present study aimed to investigate the potential role and molecular mechanisms of miR‑217 in the regulation of neuronal survival in CIR injury. To perform the investigation, an in vitro cellular model of CIR injury was established by treating neurons with oxygen‑glucose deprivation and reoxygenation (OGD/R). miR‑217 levels in neurons were detected using reverse transcription‑quantitative PCR. The association between miR‑217 and sirtuin 1 (SIRT1) was identified using TargetScan and validated in a dual‑luciferase reporter assay. Cell viability and apoptosis were measured using a Cell Counting Kit‑8 assay and flow cytometry, respectively. The release of lactate dehydrogenase, and the production of proinflammatory factors and oxidative stress biomarkers were analyzed by ELISAs and using specific assay kits. It was revealed that miR‑217 was significantly upregulated in OGD/R‑treated neurons. SIRT1 was a direct target of miR‑217, and was downregulated in neurons following OGD/R treatment. Downregulation of miR‑217 significantly ameliorated OGD/R‑induced neuronal injury, inflammatory responses and oxidative stress. The effects of miR‑217 inhibitor on OGD/R treated neurons were attenuated by SIRT1 knockdown. Additionally, western blotting revealed that the SIRT1/AMP‑activated protein kinase‑α/NF‑κB pathway was partially involved in the regulation of OGD/R‑induced neuronal injury by miR‑217. In conclusion, the data of the present study indicated that the downregulation of miR‑217 protected neurons against OGD/R‑induced injury by targeting SIRT1.

Introduction

Ischemic stroke is one of the leading causes of disability and mortality globally (1,2). Interventions require the recovery of blood flow, which can lead to reperfusion injury. Cerebral ischemia-reperfusion (CIR) injury is a pathological process in which nerve damage induced by ischemia and hypoxia is further aggravated following the short-term recovery of blood perfusion (3). CIR leads to mitochondrial dysfunction, inflammation, massive release of reactive oxygen species (ROS), excessive glutamate excitotoxicity and cell death, ultimately resulting in irreversible brain damage (4). Progress has been made in reperfusion therapy (5); however, the therapeutic outcomes remain unsatisfactory. Therefore, improved understanding of the pathological process of CIR injury and the identification of novel treatment strategies is required.

MicroRNAs (miRNAs/miRs) are a class of small endogenous noncoding RNAs (~22 nucleotides in length) that can regulate the expression of genes at the post-transcriptional level by binding to the 3′-untranslated region (3′-UTR) of target genes (68). miRNAs have been reported to serve important roles in the regulation of a variety of biological processes, including proliferation, differentiation and apoptosis, via the regulation of various target genes (7,911). Increasing evidence has indicated that miRNAs serve important regulatory roles in normal physiology and disease (12), including in the pathogenesis of CIR injury (13,14). Numerous studies have reported that various miRNAs are dysregulated during CIR injury, and that targeting these miRNAs can effectively relieve neuronal injury in vitro and in vivo (1517).

miRNA-217 has been studied in various types of cancers, including lung adenocarcinoma, acute myeloid leukemia, liver and gastric cancers, and colorectal carcinoma (1822). miR-217 has been reported to be downregulated in glioma tissues and cells, and overexpression of miR-217 was observed to reduce the malignancy of glioma cells in vitro and decrease tumor growth in vivo (23); however, to the best of our knowledge, there is no data regarding the expression of miR-217 in other neurological diseases, and whether miR-217 is involved in regulating CIR injury remains unknown.

Research into damage following CIR has increased in previous years, and a large number of studies have demonstrated that the mechanisms underlying the effects of CIR on brain damage are complex (24). At present, there is no treatment strategy to effectively treat CIR injury. Therefore, finding new and effective CIR injury treatment methods has important clinical significance. Novel roles for miRNAs in the pathogenesis of CIR injury have been identified (1317); however, to the best of our knowledge, the role of miR-217 in CIR injury remains unclear. Therefore, the aims of the present study were to investigate the role of miR-217 in neuronal injury induced by CIR using an in vitro cellular model induced by oxygen-glucose deprivation and reoxygenation (OGD/R).

Materials and methods

Primary neuron culture

Primary rat cerebral cortical neurons were extracted from Sprague-Dawley rat embryos (n=5) at embryonic day 16–18 as previously described (25,26). In brief, brain cortex tissues were extracted and dissected in Hanks' balanced salt solution (HBSS), cut into ~1 mm3 cubes, washed with HBSS, and then digested with 0.25% trypsin (37°C for 15 min). Then, DMEM-F12 (Gibco; Thermo Fisher Scientific, Inc.) with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) was used to stop trypsin digestion. The dissociated cells were seeded in 24-well plates and cultured for 24 h at 37°C. Then, the cells were cultured in neurobasal medium (Gibco; Thermo Fisher Scientific, Inc.) containing 2% B-27, 2 mM glutamine and 50 ug/ml gentamycin, and cells were incubated at 37°C with 5% CO2. Sprague-Dawley rats were obtained from Beijing Vital River Laboratory Animal Technology Co., Ltd., and housed at 25±5°C with 50% humidity under a 12:12-h dark/light cycle. Rats accessed to food and water ad libitum. Animal experiments were conducted according to the the guidelines of the National Institutes of Health for the Care and Use of Laboratory Animals (27). The present study was approved by the Animal Ethics Committee of the First People's Hospital of Wenling (Wenling, China).

OGD/R model establishment

The OGD/R model was established in the present study as previously described (26). In brief, cortical neurons were plated into 6-well plates at a density of 5×105 cells/ml (2 ml) and cultured overnight. Then, the neurons were incubated in glucose-free DMEM in a hypoxic incubator chamber (1% O2, 5% CO2 and 94% N2) at 37°C for 6 h. After three washes with DMEM, cells were incubated in DMEM supplemented with 4.5 g/l glucose at 37°C with 5% CO2 for 24 h.

The same treatment was performed in the control group without OGD/R exposure. Briefly, neurons in the control group were cultured in DMEM under standard conditions for 6 h. After three washes with DMEM, cells were incubated in DMEM at 37°C with 5% CO2 for 24 h.

Cell transfection

Cortical neurons were seeded in a 6-well plate (1×106 cells/well) and incubated at 37°C for 24 h. Then, 100 nM miR-217 inhibitor (5′-UACUGCAUCAGGAACUGAUUGGA-3′; Bioneer Corp.), 100 nM inhibitor control (5′-GCCUCCGGCUUCGCACCUCU-3′; Bioneer Corp.), 2 µl control-small interfering RNA (siRNA; cat. no. sc-36869; Santa Cruz Biotechnology, Inc.), 2 µl sirtuin 1 (SIRT1)-siRNA (cat. no. sc-40986; Santa Cruz Biotechnology, Inc.) or 100 nM miR-217 inhibitor + 2 µl SIRT1-siRNA was transfected into the cells using Lipofectamine® 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. At 24 h after transfection, transfection efficiency was measured using reverse transcription-quantitative PCR (RT-qPCR). At 24 h following transfection with miR-217 inhibitor, inhibitor control, or miR-217 inhibitor+SIRT1-siRNA, cells were subjected to OGD/R induction.

Cell Counting Kit-8 (CCK-8) assay

Cell viability was determined at 24 h following OGD/R induction as previously described (26). Cell viability was determined via a CCK-8 assay. In brief, cells were plated into a 96-well plate (5×103 cells/well) and incubated at 37°C overnight. Then, cells were transfected with or without miR-217 inhibitor, inhibitor control or miR-217 inhibitor + SIRT1-siRNA for 24 h (in equal quantities as aforementioned), followed by OGD/R treatment. Subsequently, 10 µl CCK-8 solution (cat. no. C0038; Beyotime Biotechnology) was added to each well and incubated for another 2 h at 37°C. At the end of the experiment, cell viability was calculated by measuring the absorbance at 450 nm using a FLUOstar® Omega microplate reader (BMG Labtech GmbH).

ELISA

The levels of tumor necrosis factor-α (TNF-α; cat. no. PT516), and interleukin (IL)-6 (cat. no. PI326) and IL-1β (cat. no. PI301; all Beyotime Institute of Biotechnology) in the supernatant of cells were measured via sandwich ELISAs according to the manufacturer's protocol.

Lactate dehydrogenase (LDH) assay

An LDH Cytotoxicity Assay kit (cat. no. C0016; Beyotime Institute of Biotechnology) was used to determine cell injury according to the manufacturer's protocols. In brief, cells were lysed and then incubated with NADH and pyruvate at 37°C for 15 min. The absorbance value at a wavelength of 530 nm was determined using a FLUOstar Omega microplate reader.

Cell apoptosis assay

To investigate the effects of miR-217 on the apoptosis of cortical neurons, an Annexin V-FITC/propidium iodide (PI) Apoptosis Detection kit (cat no. 70-AP101-100; MultiSciences Biotech Co., Ltd.) was using according to the manufacturer's protocols. Following the aforementioned treatments, cortical neurons were stained with 5 µl Annexin V-FITC and 5 µl PI for 30 min at room temperature in the dark. Subsequently, a flow cytometer (BD Biosciences) was used to analyze cell apoptosis. The apoptosis rate (early + late) was calculated (percentage of cells in the right quadrants) using WinMDI soft-ware (version 2.5; Purdue University Cytometry Laboratories).

Evaluation of ROS, superoxide dismutase (SOD) and malondialdehyde (MDA) levels

To analyze the levels of ROS in cells, a Reactive Oxygen Species Assay Kit (cat. no. S0033; Beyotime Institute of Biotechnology) was used according to the manufacturer's protocols. A Total Superoxide Dismutase Assay kit with WST-8 (cat. no. S0101; Beyotime Institute of Biotechnology) was used to determine the level of SOD in cortical neurons according to the manufacturer's protocols. A Lipid Peroxidation MDA Assay kit (cat no. S0131; Beyotime Institute of Biotechnology) was used to determine the levels of MDA.

Western blot assay

Proteins were extracted from cells using RIPA lysis buffer (cat no. P0013E; Beyotime Institute of Biotechnology) according to the manufacturer's protocols. A bicinchoninic acid assay kit (Pierce; Thermo Fisher Scientific, Inc.) was used to quantify the protein samples according to the manufacturer's protocols. Equal amounts of protein samples (25 µg/lane) were separated via SDS-PAGE on 12% gels, transferred onto polyvinylidene difluoride membranes (EMD Millipore), and blocked in 5% skim milk at room temperature for 1.5 h. Then, the membranes were incubated with primary antibodies against SIRT1 (1:1,000; cat. no. 9475; Cell Signaling Technology, Inc.), phosphorylated (p)-AMP-activated protein kinase-α (AMPK-α; 1:1,000; cat. no. 50081; Cell Signaling Technology, Inc.), AMPK-α (1:1,000; cat. no. 5831; Cell Signaling Technology, Inc.), NF-κB p65 (1:1,000; cat. no. 8242; Cell Signaling Technology, Inc.), p-p65 (1:1,000; cat. no. 3033; Cell Signaling Technology, Inc.), and β-actin (1:1,000; cat. no. 4970; Cell Signaling Technology, Inc.) overnight at 4°C, followed by incubation with the horseradish peroxidase-conjugated anti-rabbit IgG secondary antibody (1:2,000; cat no. 7074; Cell Signaling Technology, Inc.) at room temperature for 2 h. Finally, protein blots were visualized using chemiluminescent ECL reagent (EMD Millipore) and quantified by densitometry (QuantityOne 4.5.0 software; Bio-Rad Laboratories, Inc.).

RT-qPCR

Total RNA was extracted from cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. A TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used for cDNA generation. Reaction conditions for RT were: 50°C for 5 min and 80°C for 2 min. An SYBR Premix Ex Taq™ II (TliRNaseH Plus) kit (Takara Bio, Inc.) was using to analyze the synthesized cDNAs. U6 and GAPDH were used as internal controls for miRNA and mRNA expression, respectively. Primer sequences for PCR were as follows: miR-217, forward, 5′-CGCAGATACTGCATCAGGAA-3′ and reverse, 5′-CTGAAGGCAATGCATTAGGAACT-3′; SIRT1, forward, 5′-AATCCAGTCATTAAACGGTCTACAA-3′ and reverse, 5′-TAGGACCATTACTGCCAGAGG-3′; U6, forward, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ and reverse, 5′CGCTTCACGAATTTGCGTGTCAT3′; and GAPDH, forward, 5′-CTTTGGTATCGTGGAAGGACTC-3′ and reverse, 5′-GTAGAGGCAGGGATGATGTTCT-3′. The thermocycling conditions were as follows: 95°C for 5 min, followed by 38 cycles of denaturation at 95°C for 15 sec and annealing/elongation at 60°C for 30 sec. The relative gene expression was calculated using the 2−ΔΔCq method (28).

Dual-luciferase reporter assay

TargetScan bioinformatics software (release 7.1; www.targetscan.org/vert_71) was used to predict the targets of miR-217; a putative binding site between the 3′-UTR of SIRT1 and miR-217 was identified. Then, to determine whether miR-217 directly binds to SIRT1, the target sequence (WT-SIRT1; ATGCAGT) and a mutated sequence (MUT-SIRT1; TACGTCA) were synthesized. To point-mutate the miR-217 binding domain on the 3′-UTR of SIRT1, a QuikChange Site-Directed Mutagenesis kit (Stratagene; Agilent Technologies, Inc.) was used according to the manufacturer's protocols. Following digestion with XhoI and NotI restriction enzymes, a pmiR RB Report™ reporter gene plasmid vector (Guangzhou RiboBio Co., Ltd.) were combined with synthesized WT-SIRT1 or MUT-SIRT1 to construct recombinant plasmids, SIRT1-WT or SIRT1-MUT. Neurons (5×104 cells/well) were co-transfected with 50 ng SIRT1-WT or 50 ng SIRT1-MUT, 25 ng pRL-TK (expressing Renilla luciferase as the internal control; Promega Corporation), and 50 nM miR-217 mimic (5′-UACUGCAUCAGGAACUGAUUGGA-3′) or 50 nM mimic control (5′-UUUGUACUACACAAAAGUACUG-3′) using Lipofectamine 2000 according to the manufacturer's protocols. At 24 h following transfection, luciferase activity was determined using a dual-luciferase assay system (Promega Corporation) and normalized to the Renilla luciferase activity.

Statistical analysis

Experiments in the present study were performed three times. Experimental analysis was conducted using SPSS 17.0 software (SPSS, Inc.). Data were presented as the mean ± standard deviation. Differences between groups were determined using Student's t-test or one-way ANOVA with a Bonferroni post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-217 is upregulated in OGD/R-treated neurons

To investigate whether miR-217 was involved in CIR injury in vitro, the levels of miR-217 in neurons following OGD/R or control treatment was determined via RT-qPCR analysis. As presented in Fig. 1, compared with the control, OGD/R treatment significantly upregulated the expression of miR-217 in neurons.

SIRT1 is a target gene of miR-217

TargetScan was used to predict the potential targets of miR-217, and a putative binding site between SIRT1 and miR-217 was identified (Fig. 2A). To demonstrate the association between miR-217 and SIRT1, a luciferase reporter assay was conducted. miR-217 mimic was used to significantly upregulated miR-217 expression in neurons (Fig. 2B). As presented in Fig. 2C, compared with cells co-transfected with WT-SIRT1 and mimic control, luciferase activity was significantly decreased in neurons co-transfected with WT-SIRT1 and miR-217 mimic. The results indicated that miR-217 directly targeted SIRT1.

Subsequently, the protein and mRNA levels of SIRT1 in neurons following control or OGD/R treatment were determined via western blot and RT-qPCR analyses, respectively. As presented in Fig. 2D, OGD/R treatment markedly reduced SIRT1 protein expression in neurons. Additionally, compared with the control group, the mRNA levels of SIRT1 in neurons were significantly downregulated following OGD/R treatment (Fig. 2E).

Downregulation of miR-217 alleviates OGD/R-induced neuronal injury

To investigate the role of miR-217 in OGD/R-induced neuronal injury, neurons were transfected with miR-217 inhibitor, inhibitor control, control-siRNA, SIRT1-siRNA or miR-217 inhibitor + SIRT1-siRNA for 24 h, then subjected to OGD/R induction. The transfection efficiency was measured by RT-qPCR and/or western blotting. As presented in Fig. 3A, compared with the transfection control group, miR-217 inhibitor significantly downregulated the expression of miR-217 in neurons, whereas the mRNA levels of SIRT1 in neurons were decreased following SIRT1-siRNA transfection (Fig. 3B). The protein levels of SIRT1 in neurons were also reduced following SIRT1-siRNA transfection (Fig. 3D). Furthermore, it was demonstrated that miR-217 inhibitor significantly increased the mRNA expression of SIRT1 in neurons, and that this upregulation was attenuated by SIRT1-siRNA (Fig. 3C). miR-217 inhibitor also enhanced the protein expression of SIRT1 in neurons, in a manner that was attenuated by SIRT1-siRNA (Fig. 3E).

Then, the effects of miR-217 inhibitor on OGD/R-induced injury were determined via CCK-8 and LDH assays. Consistent with a previous study (25), OGD/R treatment significantly reduced cell viability and increased LDH release. The effects of OGD/R treatment were inhibited by miR-217 downregulation, and this inhibition was attenuated by SIRT1-siRNA (Fig. 4A and B). Additionally, miR-217 inhibition suppressed the OGD/R treatment-induced apoptosis of neurons (Fig. 4C and D). Collectively, the data indicated that miR-217 downregulation attenuated OGD/R-induced neuronal injury.

Downregulation of miR-217 alleviates OGD/R-induced inflammatory response

To further investigate the biological function of miR-217 in the regulation of OGD/R injury, the effects of miR-217 inhibitor on OGD/R-induced inflammation were determined. The levels of TNF-α, IL-6 and IL-1β were detected by ELISA. As presented in Fig. 5, the increased levels of TNF-α, IL-6 and IL-1β following OGD/R were significantly reduced by miR-217 inhibitor; however, these reductions were prevented by SIRT1-siRNA.

Downregulation of miR-217 alleviates OGD/R-induced oxidative stress

It was then investigated as to whether transfection with miR-217 inhibitor induced an effect on OGD/R-induced oxidative stress; it was observed that compared with the control, OGD/R treatment significantly enhanced the levels of ROS and MDA, and reduced those of SOD in neurons. Downregulation of miR-217 significantly decreased ROS production (Fig. 6A), reduced the levels of MDA (Fig. 6B) and increased those of SOD (Fig. 6C) in OGD/R-treated neurons, whereas these alterations were attenuated by SIRT1 silencing. The results indicated that miR-217 downregulation attenuated OGD/R-induced oxidative stress.

Effects of miR-217 inhibitor on the SIRT1/AMPK-α/NF-κB pathway in OGD/R-treated neurons

Finally, the SIRT1/AMPK-α/NF-κB pathway in OGD/R-treated neurons was analyzed to investigate the molecular mechanisms underlying the effects of miR-217 on OGD/R-induced neuronal injury. As presented in Fig. 7, compared with the control, OGD/R treatment significantly reduced SIRT1 mRNA and protein levels (Fig. 7A and B), decreased AMPK-α protein phosphorylation levels (Fig. 7A and C) and promoted p65 phosphorylation (Fig. 7A and D) in neurons. Compared with the OGD/R treatment group, miR-217 downregulation significantly upregulated SIRT1 and p-AMPK-α expression, and decreased p-p65 levels; these effects were eliminated by SIRT1 downregulation.

Discussion

Recent studies have reported that miR-217 serves an important role in the development of tumors (1822). miR-217 is involved in the apoptosis of human podocyte cells by targeting TNF ligand superfamily member 11 in membranous nephropathy (29). Another study demonstrated that miR-217 promotes fibroblast senescence (30). Additionally, miR-217 was identified as a key regulator in ethanol-induced hepatic inflammation (31); however, the role of miR-217 in neuronal injury induced by CIR injury remains unclear. In the present study, the results indicated that miR-217 was upregulated in OGD/R-treated neurons, and its downregulation relieved OGD/R treatment-induced neuronal injury, as revealed by increased cell viability, reduced LDH release and decreased cell apoptosis. Additionally, it was observed that miR-217 downregulation notably inhibited OGD/R-induced inflammatory responses and oxidative stress. Furthermore, the findings of the present study revealed that SIRT1 was a direct target of miR-217, and that the effects of miR-217 inhibitor on OGD/R-treated neurons were eliminated by SIRT1 silencing.

A number of experimental studies have revealed that the mechanisms underlying the effects of CIR on brain damage are complex (24,25). At present, there is no strategy to effectively treat CIR injury. Therefore, finding novel and effective methods has important clinical relevance. The emerging roles of miRNAs in the pathogenesis of CIR injury have been identified in numerous studies (1216). In the present study, the role of miR-217 in neuron injury induced by OGD/R was investigated.

An in vitro cellular model of CIR injury was established by treating neurons with OGD/R. The OGD/R model is a cell injury model widely used to study CIR injury in vitro (24,26,32). Then, the levels of miR-217 in neurons subjected to control or OGD/R treatment were determined, and the results revealed that compared with the control group, miR-217 was significantly upregulated in OGD/R-treated neurons. Subsequently, it was revealed that SIRT1 was a direct target of miR-217 that was downregulated in neurons treated with OGD/R. SIRT1 is a NAD+-dependent deacetylase important for apoptosis, cell cycle, mitochondrial function and metabolism (33), and serves an important role in the regulation of inflammatory responses and oxidative stress (3337). To explore the potential function of miR-217 in the regulation of OGD/R-induced neuronal injury, loss-of-function experiments were performed by transfection with miR-217 inhibitor. The results suggested that miR-217 downregulation significantly mitigated OGD/R-induced neuronal injury, as determined by increased cell viability, reduced LDH release and decreased cell apoptosis. Additionally, miR-217 downregulation notably suppressed inflammatory responses and oxidative stress enhanced by OGD/R treatment in neurons.

The activation of the AMPK-α-SIRT1 pathway can inhibit NF-κB inflammation (38). Activation of AMPK/SIRT1 signaling attenuated TNF-α-induced MMP-3 expression in human nucleus pulposus cells (39). Additionally, activation of the AMPK/SIRT1 pathway protected PC12 cells against cisplatin-induced neurotoxicity (40). The pathway has also been reported to relieve oxidized low-density lipoprotein-induced endothelial cell injury (41). These results indicated that AMPK/SIRT1 signaling serves important roles in cell injury and inflammation regulation. Furthermore, a recent study revealed that AMPK/SIRT1 pathway activation was involved in the protective effects of lncRNA SNHG12 on CIR injury (42). Therefore, to investigate the molecular mechanisms underlying the effects of miR-217 inhibitor on OGD/R-induced neuronal injury, the activity of the SIRT1/AMPK-α/NF-κB pathway was analyzed in the present study. Results revealed that OGD/R treatment significantly reduced SIRT1 mRNA and protein levels, decreased AMPK-α phosphorylation, and increased p65 phosphorylation level in neurons; these effects were reversed by miR-217 inhibitor. Of note, all the observed effects of miR-217 inhibitor on OGD/R-treated neurons were attenuated by SIRT1 silencing.

In conclusion, the results of the present study demonstrated that miR-217 was upregulated in OGD/R-treated neurons, and that its inhibition attenuated OGD/R-induced neuronal injury via the upregulation of SIRT1 and the SIRT1/AMPK-α/NF-κB pathway. Of note, this is a preliminary study of the role of miR-217 in CIR injury, and further research is required to more comprehensively and convincingly determine the precise roles and mechanisms of miR-217 in CIR injury. For example, the effects of miR-217 on CIR injury in vivo should be investigated, in addition to the effects of SIRT1 manipulation alone on CIR injury. Furthermore, in the present study, experiments were performed at only one time point (24 h following OGD/R treatment); thus, additional time points should be investigated in the future. Finally, an investigation into the association between miR-217 expression and the clinical characteristics and prognosis of patients with CIR injury is required to confirm the clinical relevance of miR-217 in CIR injury. In-depth research into these issues will be conducted in the future.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Medical and Health Science and Technology Planning Project of Zhejiang Province (grant no. 2018KY918).

Availability of data and materials

The analyzed data sets generated during the present study are available from the corresponding author on reasonable request.

Authors' contributions

GR and WZ contributed to study design, data collection, statistical analysis, data interpretation and manuscript preparation. SS contributed to data collection and interpretation. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Animal Ethics Committee of The First People's Hospital of Wenling.

Patient consent for publication

Not applicable.

Competing interests

All authors declare that they have no competing interests.

References

1 

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

2 

Goldstein LB, Adams R, Becker K, Furberg CD, Gorelick PB, Hademenos G, Hill M, Howard G, Howard VJ, Jacobs B, et al: Primary prevention of ischemic stroke A statement for healthcare professionals from the stroke council of the American heart association. Circulation. 103:163–182. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Jean WC, Spellman SR, Nussbaum ES and Low WC: Reperfusion injury after focal cerebral ischemia: The role of inflammation and the therapeutic horizon. Neurosurgery. 43:1382–1397. 1998. View Article : Google Scholar : PubMed/NCBI

4 

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

5 

Bhaskar S, Stanwell P, Cordato D, Attia J and Levi C: Reperfusion therapy in acute ischemic stroke: Dawn of a new era? BMC Neurol. 18:82018. View Article : Google Scholar : PubMed/NCBI

6 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Hammond SM: An overview of microRNAs. Adv Drug Deliv Rev. 87:3–14. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Ghildiyal M and Zamore PD: Small silencing RNAs: An expanding universe. Nat Rev Genet. 10:94–108. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Soifer HS, Rossi JJ and Saetrom P: MicroRNAs in disease and potential therapeutic applications. Mol Ther. 15:2070–2079. 2017. View Article : Google Scholar

10 

Krol J, Loedige I and Filipowicz W: The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet. 11:597–610. 2010. View Article : Google Scholar : PubMed/NCBI

11 

O'Connell RM, Rao DS, Chaudhuri AA and Baltimore D: Physiological and pathological roles for microRNAs in the immune system. Nat Rev Immunol. 10:111–122. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Mendell JT and Olson EN: MicroRNAs in stress signaling and human disease. Cell. 148:1172–1187. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Di Y, Lei Y, Yu F, Changfeng F, Song W and Xuming M: MicroRNAs expression and function in cerebral ischemia reperfusion injury. J Mol Neurosci. 53:242–250. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Hu Y, Deng H, Xu S and Zhang J: MicroRNAs regulate mitochondrial function in cerebral ischemia-reperfusion injury. Int J Mol Sci. 16:24895–24917. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Wang P, Liang X, Lu Y, Zhao X and Liang J: MicroRNA-93 downregulation ameliorates cerebral ischemic injury through the Nrf2/HO-1 defense pathway. Neurochem Res. 41:2627–2635. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Wang N, Zhang L, Lu Y, Zhang M, Zhang Z, Wang K and Lv J: Down-regulation of microRNA-142-5p attenuates oxygen-glucose deprivation and reoxygenation-induced neuron injury through up-regulating Nrf2/ARE signaling pathway. Biomed Pharmacother. 89:1187–1195. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Shi F, Dong Z, Li H, Liu X, Liu H and Dong R: MicroRNA-137 protects neurons against ischemia/reperfusion injury through regulation of the notch signaling pathway. Exp Cell Res. 352:1–8. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Liu AN, Qu HJ, Yu CY and Sun P: Knockdown of LINC01614 inhibits lung adenocarcinoma cell progression by up-regulating miR-217 and down-regulating FOXP1. J Cell Mol Med. 22:4034–4044. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Wang LP, Wang JP and Wang XP: HOTAIR contributes to the growth of liver cancer via targeting miR-217. Oncol Lett. 15:7963–7972. 2018.PubMed/NCBI

20 

Yan J, Wu G, Chen J, Xiong L, Chen G and Li P: Downregulated miR-217 expression predicts a poor outcome in acute myeloid leukemia. Cancer Biomark. 22:73–78. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Safaralizadeh R, Ajami N, Nemati M, Hosseinpourfeizi M, Azimzadeh Isfanjani A and Moaddab SY: Disregulation of miR-216a and miR-217 in gastric cancer and their clinical significance. J Gastrointest Cancer. 50:78–83. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Yu B, Du Q, Li H, Liu HY, Ye X, Zhu B, Zhai Q and Li XX: Diagnostic potential of serum exosomal colorectal neoplasia differentially expressed long non-coding RNA (CRNDE-p) and microRNA-217 expression in colorectal carcinoma. Oncotarget. 8:83745–83753. 2017.PubMed/NCBI

23 

Zheng J, Liu X, Xue Y, Gong W, Ma J, Xi Z, Que Z and Liu Y: TTBK2 circular RNA promotes glioma malignancy by regulating miR-217/HNF1β/Derlin-1 pathway. J Hematol Oncol. 10:522017. View Article : Google Scholar : PubMed/NCBI

24 

Sahu S, Nag DS, Swain A and Samaddar DP: Biochemical changes in the injured brain. World J Biol Chem. 8:21–31. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Zhang J, Zhao F, Zhao Y, Wang J, Pei L, Sun N and Shi J: Hypoxia induces an increase in intracellular magnesium via transient receptor potential melastatin 7 (TRPM7) channels in rat hippocampal neurons in vitro. J Biol Chem. 286:20194–20207. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Liu X, Li M, Hou M, Huang W and Song J: MicroRNA-135a alleviates oxygen-glucose deprivation and reoxygenation-induced injury in neurons through regulation of GSK-3β/Nrf2 signaling. J Biochem Mol Toxicol. 2:e221592018. View Article : Google Scholar

27 

Bayne K: Revised guide for the care and use of laboratory animals available. American physiological society. Physiologist. 39:199208–199211. 1996.

28 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Li J, Liu B, Xue H, Zhou QQ and Peng L: miR-217 Is a useful diagnostic biomarker and regulates human podocyte cells apoptosis via targeting TNFSF11 in membranous nephropathy. Biomed Res Int. 2017:21687672017.PubMed/NCBI

30 

Wang B, Du R, Xiao X, Deng ZL, Jian D, Xie HF and Li J: Microrna-217 modulates human skin fibroblast senescence by directly targeting DNA methyltransferase 1. Oncotarget. 8:33475–33486. 2017.PubMed/NCBI

31 

Yin H, Liang X, Jogasuria A, Davidson NO and You M: miR-217 regulates ethanol-induced hepatic inflammation by disrupting sirtuin 1-lipin-1 signaling. Am J Pathol. 185:1286–1296. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Xin L, Junhua W, Long L, Jun Y and Yang X: Exogenous hydrogen sulfide protects SH-SY5Y cells from OGD/rinduced injury. Curr Mol Med. 17:563–567. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Nogueiras R, Habegger KM, Chaudhary N, Finan B, Banks AS, Dietrich MO, Horvath TL, Sinclair DA, Pfluger PT and Tschöp MH: Sirtuin 1 and sirtuin 3: Physiological modulators of metabolism. Physiol Rev. 92:1479–1514. 2012. View Article : Google Scholar : PubMed/NCBI

34 

da Cunha MSB and Arruda SF: Tucum-do-cerrado (Bactris setosa Mart.) may promote anti-aging effect by Upregulating SIRT1-Nrf2 pathway and attenuating oxidative stress and inflammation. Nutrients. 9:E12432017. View Article : Google Scholar : PubMed/NCBI

35 

Rada P, Pardo V, Mobasher MA, García-Martínez I, Ruiz L, González-Rodríguez Á, Sanchez-Ramos C, Muntané J, Alemany S, James LP, et al: SIRT1 controls acetaminophen hepatotoxicity by modulating inflammation and oxidative stress. Antioxid Redox Signal. 28:1187–1208. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Chan SH, Hung CH, Shih JY, Chu PM, Cheng YH, Lin HC and Tsai KL: SIRT1 inhibition causes oxidative stress and inflammation in patients with coronary artery disease. Redox Biol. 13:301–309. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Cheng YY, Kao CL, Ma HI, Hung CH, Wang CT, Liu DH, Chen PY and Tsai KL: SIRT1-related inhibition of pro-inflammatory responses and oxidative stress are involved in the mechanism of nonspecific low back pain relief after exercise through modulation of Toll-like receptor 4. J Biochem. 158:299–308. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Tian Y, Ma J, Wang W, Zhang L, Xu J, Wang K and Li D: Resveratrol supplement inhibited the NF-κB inflammation pathway through activating AMPKα-SIRT1 pathway in mice with fatty liver. Mol Cell Biochem. 422:75–84. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Wang XH, Zhu L, Hong X, Wang YT, Wang F, Bao JP, Xie XH, Liu L and Wu XT: Resveratrol attenuated TNF-α-induced MMP-3 expression in humannucleus pulposus cells by activating autophagy via AMPK/SIRT1 signaling pathway. Exp Biol Med (Maywood). 241:848–853. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Ferreira RS, Dos Santos NAG, Bernardes CP, Sisti FM, Amaral L, Fontana ACK and Dos Santos AC: Caffeic acid phenethyl ester (CAPE) protects PC12 cells against cisplatin-induced neurotoxicity by activating the AMPK/SIRT1, MAPK/Erk, and PI3k/Akt signaling pathways. Neurotox Res. Apr 23–2019.(Epub ahead of print). doi: 10.1007/s12640-019-00042-w. View Article : Google Scholar

41 

Zhao D, Sun X, Lv S, Sun M, Guo H, Zhai Y, Wang Z, Dai P, Zheng L, Ye M and Wang X: Salidroside attenuates oxidized low-density lipoprotein-induced endothelial cell injury via promotion of the AMPK/SIRT1 pathway. Int J Mol Med. 43:2279–2290. 2019.PubMed/NCBI

42 

Yin WL, Yin WG, Huang BS and Wu LX: LncRNA SNHG12 inhibits miR-199a to upregulate SIRT1 to attenuate cerebral ischemia/reperfusion injury through activating AMPK signaling pathway. Neurosci Lett. 690:188–195. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 20 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Rao G, Zhang W and Song S: MicroRNA‑217 inhibition relieves cerebral ischemia/reperfusion injury by targeting SIRT1. Mol Med Rep 20: 1221-1229, 2019
APA
Rao, G., Zhang, W., & Song, S. (2019). MicroRNA‑217 inhibition relieves cerebral ischemia/reperfusion injury by targeting SIRT1. Molecular Medicine Reports, 20, 1221-1229. https://doi.org/10.3892/mmr.2019.10317
MLA
Rao, G., Zhang, W., Song, S."MicroRNA‑217 inhibition relieves cerebral ischemia/reperfusion injury by targeting SIRT1". Molecular Medicine Reports 20.2 (2019): 1221-1229.
Chicago
Rao, G., Zhang, W., Song, S."MicroRNA‑217 inhibition relieves cerebral ischemia/reperfusion injury by targeting SIRT1". Molecular Medicine Reports 20, no. 2 (2019): 1221-1229. https://doi.org/10.3892/mmr.2019.10317