Open Access

Inhibition of lncRNA SNHG8 plays a protective role in hypoxia-ischemia-reoxygenation-induced myocardial injury by regulating miR-335 and RASA1 expression

  • Authors:
    • Yanfeng Liu
    • Ping Zhou
    • Fengxiao Wang
    • Xuehong Zhang
    • Dongmei Yang
    • Lang Hong
    • Dongyun Ruan
  • View Affiliations

  • Published online on: June 22, 2021     https://doi.org/10.3892/mmr.2021.12236
  • Article Number: 597
  • Copyright: © Liu 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

Long non‑coding (lnc)RNAs serve a role in a number of diseases, including different types of cancer and acute myocardial infarction. The aim of the present study was to investigate the protective role of lncRNA small nucleolar RNA host gene 8 (SNHG8) in hypoxia‑ischemia‑reoxygenation (HI/R)‑induced myocardial injury and its potential mechanism of action. Cell viability, proliferation, creatine kinase myocardial band, cell apoptosis and protein expression levels were determined by Cell Counting Kit‑8 assay, EdU assay, ELISA, flow cytometry and western blotting, respectively. The association between SNHG8 and microRNA (miR)‑335 was confirmed using a dual‑luciferase reporter gene assay. The effects of the miR‑335 inhibitor transfections had on increasing apoptosis and decreasing H9C2 cell viability were reversed in cells co‑transfected with SNHG8 small interfering (si)RNA. Furthermore, it was found that miR‑335 could regulate RAS p21 protein activator 1 (RASA1) expression and that transfection with SNHG8 siRNA downregulated RASA1 expression. Silencing of RASA1 protected against HI/R‑induced H9C2 cell injury. However, SNHG8 siRNA did not further reduce apoptosis, demonstrating that SNHG8 may act through RASA1, and RASA1 may mediate the protection of SNHG8 siRNA in HI/R myocardial injury. Thus, inhibition of lncRNA SNHG8 alleviated HI/R‑induced myocardial damage by regulating miR‑335 and RASA1.

Introduction

Ischemic heart disease is a major disease with the highest mortality and morbidity rate (1.655%) in the world, an estimated 31% of all deaths worldwide are due to cardiovascular diseases (13). At present, the most effective treatment for myocardial infarction is primary percutaneous coronary intervention, but there is no effective way to prevent myocardial ischemia-reperfusion (I/R) injury (4). Furthermore, continuous perfusion after ischemia can cause myocardial I/R injury, leading to reperfusion arrhythmia, myocardial shock and other pathological changes (5). Therefore, alleviating myocardial I/R injury is considered particularly important for the prevention and treatment of ischemic myocardial injury.

Long non-coding (lnc)RNAs are >200 nucleotides in length and have no significant protein-coding potential. lncRNAs are associated with different cellular biological processes, such as cell apoptosis, invasion and proliferation (6). Furthermore, it has been found that lncRNAs are involved in the pathogenesis of cardiovascular disease (7). Upregulation of lncRNA H19 imprinted maternally expressed transcript reduced myocardial infarction-induced myocardial injury through the regulation of KDM3A expression (7). Inhibition of lncRNA KCNQ1OT1 protected against oxygen glucose deprivation/reperfusion (OGD/R)-induced myocardial cell injury via the p38/MAPK/NF-κB signaling pathway (8). A recent study demonstrated that lncRNAs can act as competing endogenous (ce)RNAs to specifically bind to microRNAs (miRNAs) and reduce the regulatory effect of miRNAs on targeted mRNAs (9). For example, lncRNA downregulation of lncRNA MALAT1 inhibited OGD/R-induced myocardial cell injury by acting as a miRNA (miR)-20b sponge and regulating autophagy (10). Inhibition of lncRNA GAS5 can alleviate myocardial reperfusion injury by acting as a ceRNA for miR-335 and regulating Rho-associated coiled-coil containing protein kinase 1 expression (11). It has been reported that lncRNA SNHG8 acts as a potential biomarker and may participate in ischemia in myocardial cells (12). Results from the present study further identified the association between SNHG8 and miR-335 in protection from hypoxia-ischemia-reoxygenation (HI/R)-induced myocardial cell injury.

Heart failure, myocardial infarction and I/R injury are the main factors in the pathogenesis of heart disease that may result in cell death (13). Cardiomyocytes are lost as a result of apoptosis and necrosis in heart disease (14). Inhibition of lncRNA HOTAIR could increase the apoptotic rate of hypoxia-induced cardiomyocytes (15). LncRNA Gpr19 could protect OGD/R-induced myocardial cell injury by reducing cell apoptosis and activation of the miR-324-5p/mitochondrial fission regulator 1 axis (16). Bcl-2 and Bax proteins, members of the Bcl-2 family, and caspase-3, their downstream target, are considered to be the main regulators of cell apoptosis; they are expressed in the cytoplasm in the form of zymogens and subsequently activate cell apoptosis (17). Knockdown of lncRNA ZFAS1 expression could decrease I/R-induced myocardial cell apoptosis by regulating the changes in Bcl-2, Bax and cleaved caspase-3 expression (18). Overexpression of lncRNA SNHG8 was reported to inhibit chronic cerebral ischemia-induced neuron apoptosis (19). Therefore, the present study hypothesized that SNGH8 may protect against HI/R-induced myocardial injury by regulating apoptosis. Furthermore, it has been reported that the RAS signaling pathways serve a role in the development of a number of diseases, such as cancer and cardiac diseases (20,21). RAS p21 protein activator 1 (RASA1; also known as p120RasGAP) was the first RasGAP protein to be identified; it primarily acts by negatively regulating RAS signaling (22,23). RASA1 has been shown to serve a role in the cardiac myocyte growth induced by hypertrophic stimuli (24).

Therefore, the aim of the present study was to investigate the protective effects of lncRNA SNHG8 in HI/R-induced myocardial damage. SNHG8 may serve an important role by regulating miR-335 and RASA1 expression, suggesting that SNHG8 could be a novel therapeutic strategy for treatment of myocardial I/R injury.

Materials and methods

Cell culture and HI/R conditions

The embryonic rat cardiomyocyte-derived H9C2 cell line was purchased from the American Type Culture Collection and grown in Gibco Dulbecco's modified Eagle's medium (DMEM; Thermo Fisher Scientific, Inc.), which contained Gibco 10% fetal bovine serum (FBS; Thermo Fisher Scientific, Inc.) and 1% penicillin/streptomycin (MilliporeSigma) in a humidified incubator under standard conditions (5% CO2). After a 24-h incubation at 37°C, cells were washed three times with phosphate-buffered saline (PBS) and the growth media was replaced with glucose- and FBS-free DMEM. Cells were then cultured in a Thermo 3131 hypoxic incubator (<0.1% O2, 5% CO2 and 95% N2; Thermo Fisher Scientific, Inc.) at 37°C for 24 h (Hypoxia-ischemic; HI). Following treatment, the cells then were reoxygenated in an incubator in a 5% CO2 atmosphere for 2 h (Hypoxia-ischemic-reoxygenate; HI/R). H9C2 cells were used to HI/R conditions to induce a myocardial I/R injury model in vitro.

Transfection

H9C2 cells were seeded into 24-well plates at a density of 1×105 cells/well. The cells were cultured overnight before transfection at 37°C. Next, transfection with SNHG8 siRNA (100 nM), RASA1 siRNA (100 nM), negative control (NC; scrambled sequence) siRNA (Santa Cruz Biotechnology, Inc.), RASA1 plasmid, NC (empty plasmid) plasmid (OriGene Technologies, Inc.), miR-335 mimics, NC-mimics inhibitor or NC-inhibitor (Guangzhou RiboBio Co., Ltd.) into H9C2 cells was performed using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The transfection medium was replaced with complete medium 6 h after transfection, and the cells were incubated for the indicated times. All treatments were started 24 h after transfection. The siRNA sequences are presented in Table I and Fig. S1 demonstrates the transfection efficacy as determined by reverse transcription-quantitative PCR (RT-qPCR).

Table I.

siRNAs, miRNA, miRNA inhibitors and NC sequences used for transfections.

Table I.

siRNAs, miRNA, miRNA inhibitors and NC sequences used for transfections.

GeneSequence (5′→3′)
NC siRNA CATAGCGGTGTAGTAAAGCATAATA
SNHG8 siRNA ATTACGATGGATGATGGAAACATA
RASA1 siRNA TAGGATATTACAGTCACGT
NC-mimic UUCUCCGAACGUGUCACGUTT
miR-335 mimicsUCAAGAGCAA UAACGAAAAAUGU
NC inhibitor GTGTAACACGTCTATACGCCCA
miR-335 inhibitor ACAUUUUUCGUUAUUGCUCUUGA

[i] miR, microRNA NC, negative control; RASA1, RAS p21 protein activator 1; si, small interfering RNA; SNHG8, small nucleolar RNA host gene 8.

Cell viability

Cell viability was examined using a Cell Counting Kit-8 (CCK-8) assay (Dojindo Molecular Technologies, Inc.). The H9C2 cells transfected with SNHG8 siRNA, miR-335 mimics, inhibitor, RASA1 siRNA or NC were seeded into 96-well plates at a density of 3×103 cells/well incubation at 37°C for 24 h. Then, 10 µl CCK-8 solution was added to the cells (100 µl/well) and incubated at 37°C for 3 h. An MRX II microplate reader (Dynex Technologies, Inc.) was used to detect the absorbance at 450 nm.

Western blot analysis

The cells were lysed by the cell lysis buffer (Cell Signaling Technology, Inc.) and samples centrifuged at 300 × g for 5 min at 4°C after the lysis treatment. The supernatant was collected and the protein concentrations measured with a BCA Protein Assay kit (Sigma-Aldrich; Merck KGaA). The proteins (20 µg/lane) were separated by 10% SDS-PAGE and then transferred to polyvinylidene difluoride membranes (EMD Millipore). The membranes were blocked with Tris-buffered saline (TBS) and 0.1% Tween 20 (TBS-T) containing 5% bovine serum albumin (BSA; Sigma-Aldrich; Merck KGaA) at room temperature and then incubated with anti-Bax (cat. no. ab182734), anti-Bcl-2 (cat. no. ab194583) or anti-cleaved caspase-3 (cat. no. ab2302; all from Abcam; 1:1,000 in TBST containing 5% BSA) primary antibodies overnight at 4°C. The membranes were washed with TBST three times and then incubated with the appropriate horseradish peroxidase-labeled secondary antibody (1:2,000, Cell Signaling Technology, Inc.; cat. no. 7076) for 2 h at room temperature. Finally, the ECL Plus detection system (EMD Millipore) was used to examine the protein bands and the expression levels of the target proteins were semi-quantified by detecting the optical density value of each band. GAPDH (Cell Signaling Technology, Inc.; cat. no. 5174; 1:1,000) served as an internal control.

EdU assay

The H9C2 cells were seeded in 96-well plates at a density of 3×103 cells/well in culture media. The medium was replaced with serum-free medium to synchronize the cells. After 24 h, the serum-free medium was replaced with growth media for 48 h. Cell proliferation was assessed using a Click-iT EdU imaging kit (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions.

RT-qPCR analysis

TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was used to extract total RNA from the H9C2 cells according to the manufacturer's instructions. Total RNA was then reverse transcribed to cDNA using a PrimeScript RT Reagent kit (Takara Biotechnology, Co., Ltd.; cat. no. RR047A). qPCR was then performed using a SYBR Green RT-PCR Kit (Takara Biotechnology, Co., Ltd.). The PCR conditions were as follows: 40 cycles of 95°C for 30 sec, 60°C for 34 sec and 72°C for 30 sec. U6 (Takara Biotechnology, Co., Ltd.; cat. no. 638315) and β-actin served as the internal controls. The U6 was used for miRNA, and mRNA and lncRNA was normalization by β-actin. The relative expression levels of lncRNA SNHG8, miR-335 and RASA1 expression were assessed using the comparative 2−ΔΔCq method (25) and primer sequences are presented in Table II and Table SI.

Table II.

Primer sequences used for reverse transcription-quantitative PCR.

Table II.

Primer sequences used for reverse transcription-quantitative PCR.

GenePrimer sequence (5′→3′)
SNHG8F: GACACAAGGTGGCTATGGTGCTG
R: CATGGTGGTCGTCGCGCTAAC
miR-335F: GCGGTCAAGAGCAATAACGAA
R: GTGCAGGGTCCGAGGTATTC
RASA1F: CTGGAGATTATTCCCTGTATTTTCG
R: TGTTCTTTCCGATAGTGGTCTATGA
β-actinF: CCTCTATGCCAACACAGTGC
R: CATCGTACTCCTGCTTGCTG

[i] F, forward; miR, microRNA; R, reverse; RASA1, RAS p21 protein activator 1; SNHG8, small nucleolar RNA host gene 8.

Dual-luciferase reporter assays

The cDNA fragments of SNHG8 carrying the wild-type (WT) or mutated (MuT) binding sites of miR-335 and the 3′-untranslated region (UTR) of the amplified RASA1 fragment containing the predicated target sites for miR-335 were synthesized by Shanghai GenePharma Co., Ltd. H9C2 cells (1×105 cells/well) were seeded into 24-well plates and co-transfected with 50 ng recombinant luciferase vectors, 10 ng pGL3 vectors and 50 nM miR-335 mimics, miR-335 inhibitor or NC-mimic, NC-inhibitor using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.). After transfection for 48 h, the cells were lysed and luciferase activities were evaluated using the Dual-Luciferase Reporter Assay System (Promega Corporation) and a luminometer (Glomax20/20; Promega Corporation).

Lactate dehydrogenase (LDH) release analysis

The expression of LDH was determined by Cytotoxicity LDH Assay Kit-WST (Roche Diagnostics). Briefly, 50 µl cell suspension (2.5×104 cells) was added to each well of a 96-well plate after transfection with or without SNHG8 siRNA, RASA1 siRNA or miR-335 inhibitor and incubated at 37°C for 1 h. Lysis buffer (10 µl) was added to each well and incubated at 37°C for 30 min. Then, 100 µl working solution was added to each well. The plate was protected from light and incubated at room temperature for 30 min. Finally, 50 µl stop solution was added to each well and the absorbance was measured at 490 nm using a microplate reader.

Detection of cell apoptosis

The number of apoptotic cells was determined by flow cytometry using the Annexin V-FITC Apoptosis Detection Kit (Dojindo Molecular Technologies, Inc.). Different treatment groups of H9C2 cells were treated with a trypsin-EDTA (0.25%) solution and centrifuged at 300 × g for 3 min at 4°C. Annexin V Binding Solution, diluted 10-fold, was added to make a final cell concentration of 1×106 cells/ml. The cell suspension (100 µl) was transferred to a new tube, 5 µl Annexin V-FITC conjugate was added and then 5 µl PI solution was added to the cell suspension. The samples were protected from light and incubated for 15 min at room temperature. Finally, 10-fold diluted Annexin V binding solution (400 µl) was added and the percentage of early + late apoptotic cells was quantified by flow cytometry with a FACSCalibur system equipped with the CellQuest software (version 5.1; BD Biosciences).

Bioinformatics analysis

Bioinformatics analysis was performed using StarBase v 3.0 (http://starbase.sysu.edu.cn/index.php) and miRTarBase (http://mirtarbase.cuhk.edu.cn/php/index.php) to determine the association between miRNA and lncRNA (26,27).

Statistical analysis

Data are expressed as the mean ± SD, and statistical analysis was performed using GraphPad Prism 8 software (GraphPad Software, Inc.). Statistical comparisons were conducted using unpaired Student's t-test or one-way analysis of variance with Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Protective effect of SNHG8 on HI/R myocardial injury

To determine the protective effect of lncRNA on HI/R myocardial injury, the expression levels of lncRNA were detected in the control group and HI/R myocardial cells. The results revealed that lncRNA SNHG8 was the most markedly upregulated lncRNA (Fig. 1A). It was also found that treatment with SNHG8 siRNA significantly increased HI/R-induced cell viability and proliferation, as analyzed by the CCK-8 assay and EdU analysis. LDH analysis demonstrated that treatment with SNHG8 siRNA reduced HI/R-induced cell damage (Fig. 1B-D). Subsequently, apoptotic rates and the change of apoptotic-related protein expressions following SNHG8 siRNA treatment in the HI/R myocardial cells was examined. As presented in Fig. 1E and F, HI/R-induction increased apoptosis and the protein expression levels of Bax and cleaved caspase-3 and reduced the expression of Bcl-2 protein, whereas HI/R treated cells transfected with SNHG8 siRNA exhibited decreased cell apoptosis and Bcl-2 and cleaved caspase-3 expression and increased the expression of Bcl-2 protein. These data indicated that inhibition of SNHG8 may protect against HI/R-induced myocardial cell injury.

SNHG8 regulates miR-335 expression

It has previously been reported that lncRNA mediates HI/R injury through the regulation of miRNAs, such as lncRNA MALAT1 which can regulate miRNA-20b after HI/R injury (9,28). Bioinformatics analysis was performed to screen potential miRNAs that have complementary base pairing with SNHG8. We firstly determined the expression of miRNA after under HI/R condition, showing that miR-335 was (Fig. S2A). StarBase analysis identified miR-335 as a potential target of SNHG8 (Fig. 2A). Consistent with our predication, the dual-luciferase reporter assay verified that miR-335 mimics could decrease the luciferase activity of pGL3-SNHG8-wild-type (WT), whereas no significant difference on pGL3-SNHG8-mutant (Mut) was observed (Fig. 2B). The schematic diagram of the relationship between SNHG8 and miR-335 was shown in Fig. 2C. Furthermore, silencing of SNHG8 resulted in upregulation of miR-335 expression (Fig. 2D).

SNHG8 serves a protective role in HI/R myocardial injury by regulating miR-335

To further determine the role of miR-335 on HI/R myocardial injury, H9C2 cells were co-transfected with a miR-335 inhibitor and SNHG8 siRNA. As shown in Fig. 3A, treatment with the miR-335 inhibitor reduced cell viability compared with HI/R, whereas inhibition of SNHG8 increased cell viability after co-transfected with the miR-335 inhibitor on HI/R-induced H9C2 myocardial cell injury. Furthermore, SNHG8 siRNA combined with the miR-335 inhibitor was able to enhance cell proliferation and reduce LDH release compared with the miR-335 inhibitor alone group (Fig. 3B and C, respectively). SNHG8 siRNA reduced the miR-335 inhibitor-induced increase in the number of apoptotic cells under the HI/R condition in H9C2 cells (Fig. 3D). Western blot analysis of apoptosis-related proteins demonstrated that, Bax was decreased and Bcl-2 was increased following co-transfection with SNHG8 siRNA and miR-335 inhibitor (Fig. 3E).

miR-335 negatively regulates the expression of RASA1

Based on the aforementioned results, TargetScan and miRTarBase were used to investigate the binding site of miR-335 (29,30). The overlapping analysis of the 196 differentially expressed mRNAs identified by TargetScan and the predicted single mRNA identified by miRTarBase indicated that only RASA1 interacted with miR-335 (Fig. S2B). The results indicated that miR-335 targets the 3′-UTR of the RASA1 mRNA (Fig. 4A). The schematic diagram of the relationship between miR-335 and RASA1 is shown in Fig. 4B. A dual-luciferase reporter assay was used to further verify this prediction, and the results indicated that miR-335 mimics decreased the luciferase activity of pGL3-RASA1-WT, whereas no effect on pGL3-RASA1-Mut was observed (Fig. 4C). In addition, it was demonstrated that overexpression of miR-335 significantly downregulated the expression level of RASA1 mRNA, whereas transfection with the miR-335 inhibitor had the opposite effect on the expression of RASA1 (Fig. 4D). The expression of RASA1 following transfection of HI/R-induced H9C2 cells with SNHG8 siRNA was also confirmed. The results demonstrated that SNHG8 siRNA treatment significantly downregulated the expression of RASA1 (Fig. 4E).

SNHG8 protects against HI/R myocardial injury through regulation of miR-335 and RASA1

To investigate whether miR-335 and RASA1 mediated the protective effect of SNHG8 on HI/R-induced H9C2 cell injury, a series of experiments was performed. Knockdown of RASA1 protected HI/R-induced H9C2 cells by increasing cell viability and cell proliferation, and decreasing LDH release, whereas compared with RASA1 siRNA + HI/R group, there was no significant difference in cell viability, proliferation and LDH following treatment with SNHG8 siRNA and RASA1 siRNA (Fig. 5A-C). Moreover, following RASA1 silencing, SNHG8 did not further reduce apoptosis, again demonstrating that SNHG8 may act through RASA1 (Fig. 5D). Western blot analysis showed that, compared with the RASA1 siRNA group, there was no significant difference in Bax and Bcl-2 expression after combined treatment with RASA1 siRNA and SNHG8 siRNA (Fig. 5E).

The protective effect of SNHG8 combined with RASA1 overexpression plasmid on HI/R induced H9C2 cells was analyzed further. The data demonstrated that RASA1 overexpression reduced cell viability, whereas SNHG8 siRNA combined with RASA1 overexpression increased viability (Fig. S2C). These results demonstrated that lncRNA SNHG8 may regulate the expression of RASA1 and that RASA1 may mediate the effects SNHG8 on the damage to cells. These data indicated that SNHG8 may protect HI/R-induced H9C2 cell injury through the regulation of miR-335 and RASA1 expression.

Discussion

Myocardial I/R injury is one of the major causes of mortality worldwide (31). Myocardial ischemia leads to hypoxia and cell apoptosis, further aggravating myocardial tissue damage (32). Therefore, investigating strategies to reduce myocardial injury during hypoxia are essential for treatment of heart failure and angina pectoris.

Studies have indicated that lncRNAs are related to cerebral ischemia and that these lncRNAs could regulate the development of ischemic cerebrovascular disease (33,34). They also act as ceRNAs, regulating specific RNA transcription by competitively binding with miRNAs. This lncRNA-miRNA crosstalk might be particularly important for the control and treatment of myocardial I/R injury and recovery after ischemia, and lncRNA-miRNA crosstalk have also emerged as new regulators in myocardial injury (31). Upregulation of lncRNA HULC could reduce HI/R-induced H9C2 cell apoptosis and inflammation through the miR-377-5p-NLRP3/caspase-1/IL-1β axis (35). Silencing of lncRNA PVT1 could reduce HI/R injury-induced cell apoptosis and autophagy via the miR-186/Beclin-1 signaling pathway (36). lncRNA SNHG8, located on 4qq26, belongs to the lncRNA family and is 1,062 nt in length (37). A recent study indicated that lncRNA SNHG8 was significantly upregulated in AMI and that it may be correlated with regulation of myocardial cell necrosis and apoptosis (12). The present study found that, after HI/R induction, SNHG8 exhibited the highest expression among the lncRNAs examined, and it was associated with ischemic myocardial cells, which was consistent with studies by Zhuo et al (12) and Zhang and Bian (38). Inhibition of SNHG8 could protect against HI/R-induced myocardial injury by increasing cell viability and proliferation, and by decreasing cell apoptosis.

In the present study, a bioinformatics approach was used to predict that the SNHG8 transcript contained a miR-335 binding region. It has previously been reported that miR-335 serves an important role in myocardial I/R injury (39), which is consistent with the results of the present study. The present findings also demonstrated that miR-335 inhibitor treatment could increase cell apoptosis and reduce cell viability or proliferation in HI/R-induced H9C2 cells, with co-transfection with SNHG8 siRNA, the effect of the miR-335 inhibitor on increasing apoptosis was also reversed.

miRNAs serve an important role in cardiovascular disease by interacting with downstream mRNAs (40,41). For example, upregulation of miR-149 protected against myocardial I/R damage by inhibiting forkhead box O3 expression (42). miR-7b overexpression reduced HI/R-induced H9C2 apoptosis via the hypoxia inducible factor-1α/p38 signaling pathway (43). The present study demonstrated that miR-335 negatively regulates the expression of RASA1. Combined with the aforementioned results, it was confirmed that knockdown of SNHG8 could decrease RASA1 expression. Moreover, the present study confirmed that RASA1 mediated the protective effect of SNHG8 in HI/R-induced myocardial cell injury. Therefore, it was demonstrated that SNHG8 may control HI/R-induced myocardial cell injury by regulating miR-335 and RASA1.

The main factors leading to reperfusion injury are oxidative stress, inflammation and apoptosis (44). Myocardial apoptosis is also related to ventricular dysfunction subsequent to cardiac surgery (45). Therefore, inhibition of myocardial cell apoptosis after ischemia might be considered an effective treatment to improve diastolic function after ischemia. The Bcl-2 family, including Bcl-2 and Bax, are related to the regulation of cell apoptosis (46). The present study revealed that SNHG8 could regulate apoptotic-related proteins Bax, Bcl-2 and cleaved caspase-3 by regulating miR-335 and RASA1. However, the present study also had certain limitations. The protective effect of SNHG8 was verified under HI/R-induced H9C2 cells in vitro; however, the experiment could be extended to demonstrate the effect in vivo. Additionally, further identification and confirmation of the precise mechanisms underlying RASA1 are required, such as whether RASA1 regulates Ras/MEK1/2/ERK1/2 and PI3K/Akt signaling.

In conclusion, the present study demonstrated that transfection with SNHG8 siRNA protected against HI/R-induced injury in H9C2 cells by mediating the regulation of miR-335 and RASA1, thus indicating that SNHG8 may be an effective target for treatment of myocardial I/R injury.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The current study was funded by The Traditional Chinese Medicine Research Project of Jiangxi Province (grant no. 2019A151).

Availability of data and materials

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

Authors' contributions

DR and LH conceived the idea. YL and PZ performed the experiments. FW, XZ and DY analyzed the data. DR and YL wrote the manuscript. All authors have read and approved the final manuscript. DR and LH confirm the authenticity of all the raw data.

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 

Severino P, D'Amato A, Pucci M, Infusino F, Adamo F, Birtolo LI, Netti L, Montefusco G, Chimenti C, Lavalle C, et al: Ischemic heart disease pathophysiology paradigms overview: From plaque activation to microvascular dysfunction. Int J Mol Sci. 21:81182020. View Article : Google Scholar : PubMed/NCBI

2 

Reed GW, Rossi JE and Cannon CP: Acute myocardial infarction. Lancet. 389:197–210. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Devereaux PJ and Szczeklik W: Myocardial injury after non-cardiac surgery: Diagnosis and management. Eur Heart J. 41:3083–3091. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Jokinen E: Coronary artery disease in patients with congenital heart defects. J Intern Med. 288:383–389. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Khan MA, Hashim MJ, Mustafa H, Baniyas MY, Al Suwaidi SKBM, AlKatheeri R, Alblooshi FMK, Almatrooshi MEAH, Alzaabi MEH, Al Darmaki RS and Lootah SNAH: Global epidemiology of ischemic heart disease: Results from the global burden of disease study. Cureus. 12:e93492020.PubMed/NCBI

6 

Li M, Duan L, Li Y and Liu B: Long noncoding RNA/circular noncoding RNA-miRNA-mRNA axes in cardiovascular diseases. Life Sci. 233:1164402019. View Article : Google Scholar : PubMed/NCBI

7 

Huang Y: The novel regulatory role of lncRNA-miRNA-mRNA axis in cardiovascular diseases. J Cell Mol Med. 22:5768–5775. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Li X, Dai Y, Yan S, Shi Y, Han B, Li J, Cha L and Mu J: Down-regulation of lncRNA KCNQ1OT1 protects against myocardial ischemia/reperfusion injury following acute myocardial infarction. Biochem Biophys Res Commun. 491:1026–1033. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Sui C, Dong Z, Yang C, Zhang M, Dai B, Geng L, Lu J, Yang J and Xu M: LncRNA FOXD2-AS1 as a competitive endogenous RNA against miR-150-5p reverses resistance to sorafenib in hepatocellular carcinoma. J Cell Mol Med. 23:6024–6033. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Wang S, Yao T, Deng F, Yu W, Song Y, Chen J and Ruan Z: LncRNA MALAT1 promotes oxygen-glucose deprivation and reoxygenation induced cardiomyocytes injury through sponging miR-20b to Enhance beclin1-mediated autophagy. Cardiovasc Drugs Ther. 33:675–686. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Wu N, Zhang X, Bao Y, Yu H, Jia D and Ma C: Down-regulation of GAS5 ameliorates myocardial ischaemia/reperfusion injury via the miR-335/ROCK1/AKT/GSK-3β axis. J Cell Mol Med. 23:8420–8431. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Zhuo LA, Wen YT, Wang Y, Liang ZF, Wu G, Nong MD and Miao L: LncRNA SNHG8 is identified as a key regulator of acute myocardial infarction by RNA-seq analysis. Lipids Health Dis. 18:2012019. View Article : Google Scholar : PubMed/NCBI

13 

Shekhar A, Heeger P, Reutelingsperger C, Arbustini E, Narula N, Hofstra L, Bax JJ and Narula J: Targeted imaging for cell death in cardiovascular disorders. JACC Cardiovasc Imaging. 11:476–493. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Xu T, Ding W, Ao X, Chu X, Wan Q, Wang Y, Xiao D, Yu W, Li M, Yu F and Wang J: ARC regulates programmed necrosis and myocardial ischemia/reperfusion injury through the inhibition of mPTP opening. Redox Biol. 20:414–426. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Gao L, Liu Y, Guo S, Yao R, Wu L, Xiao L, Wang Z, Liu Y and Zhang Y: Circulating long noncoding RNA HOTAIR is an essential mediator of acute myocardial infarction. Cell Physiol Biochem. 44:1497–1508. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Huang L, Guo B, Liu S, Miao C and Li Y: Inhibition of the LncRNA Gpr19 attenuates ischemia-reperfusion injury after acute myocardial infarction by inhibiting apoptosis and oxidative stress via the miR-324-5p/Mtfr1 axis. IUBMB Life. 72:373–383. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Siddiqui WA, Ahad A and Ahsan H: The mystery of BCL2 family: Bcl-2 proteins and apoptosis: An update. Arch Toxicol. 89:289–317. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Huang P, Yang D, Yu L and Shi Y: Downregulation of lncRNA ZFAS1 protects H9c2 cardiomyocytes from ischemia/reperfusion-induced apoptosis via the miR5903p/NFκB signaling pathway. Mol Med Rep. 22:2300–2306. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Liu J, An P, Xue Y, Che D, Liu X, Zheng J, Liu Y, Yang C, Li Z and Yu B: Mechanism of Snhg8/miR-384/Hoxa13/FAM3A axis regulating neuronal apoptosis in ischemic mice model. Cell Death Dis. 10:4412019. View Article : Google Scholar : PubMed/NCBI

20 

Soleimani A, Rahmani F, Saeedi N, Ghaffarian R, Khazaei M, Ferns GA, Avan A and Hassanian SM: The potential role of regulatory microRNAs of RAS/MAPK signaling pathway in the pathogenesis of colorectal cancer. J Cell Biochem. 120:19245–19253. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Liu X, Xu Y, Deng Y and Li H: MicroRNA-223 regulates cardiac fibrosis after myocardial infarction by targeting RASA1. Cell Physiol Biochem. 46:1439–1454. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Tao H, Yang JJ, Chen ZW, Xu SS, Zhou X, Zhan HY and Shi KH: DNMT3A silencing RASSF1A promotes cardiac fibrosis through upregulation of ERK1/2. Toxicology. 323:42–50. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Trial J, Entman ML and Cieslik KA: Mesenchymal stem cell-derived inflammatory fibroblasts mediate interstitial fibrosis in the aging heart. J Mol Cell Cardiol. 91:28–34. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Diao X, Shen E, Wang X and Hu B: Differentially expressed microRNAs and their target genes in the hearts of streptozotocin-induced diabetic mice. Mol Med Rep. 4:633–640. 2011.PubMed/NCBI

25 

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

26 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Huang HY, Lin YC, Li J, Huang KY, Shrestha S, Hong HC, Tang Y, Chen YG, Jin CN, Yu Y, et al: MiRTarBase 2020: Updates to the experimentally validated microRNA-target interaction database. Nucleic Acids Res. 48(D1): D148–D154. 2020.PubMed/NCBI

28 

Yang L, Lu Y, Ming J, Pan Y, Yu R, Wu Y and Wang T: SNHG16 accelerates the proliferation of primary cardiomyocytes by targeting miRNA-770-5p. Exp Ther Med. 20:3221–3227. 2020.PubMed/NCBI

29 

Han J, LaVigne CA, Jones BT, Zhang H, Gillett F and Mendell JT: A ubiquitin ligase mediates target-directed microRNA decay independently of tailing and trimming. Science. 370:eabc95462020. View Article : Google Scholar : PubMed/NCBI

30 

Shi CY, Kingston ER, Kleaveland B, Lin DH, Stubna MW and Bartel DP: The ZSWIM8 ubiquitin ligase mediates target-directed microRNA degradation. Science. 370:eabc93592020. View Article : Google Scholar : PubMed/NCBI

31 

Gu S, Tan J, Li Q, Liu S, Ma J, Zheng Y, Liu J, Bi W, Sha P, Li X, et al: Downregulation of LAPTM4B contributes to the impairment of the autophagic flux via unopposed activation of mTORC1 signaling during myocardial ischemia/reperfusion injury. Circ Res. 127:e148–e165. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Liang S, Ren K, Li B, Li F, Liang Z, Hu J, Xu B and Zhang A: LncRNA SNHG1 alleviates hypoxia-reoxygenation-induced vascular endothelial cell injury as a competing endogenous RNA through the HIF-1α/VEGF signal pathway. Mol Cell Biochem. 465:1–11. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Ghafouri-Fard S, Shoorei H and Taheri M: Non-coding RNAs participate in the ischemia-reperfusion injury. Biomed Pharmacother. 129:1104192020. View Article : Google Scholar : PubMed/NCBI

34 

Bao MH, Szeto V, Yang BB, Zhu SZ, Sun HS and Feng ZP: Long non-coding RNAs in ischemic stroke. Cell Death Dis. 9:2812018. View Article : Google Scholar : PubMed/NCBI

35 

Liang H, Li F, Li H, Wang R and Du M: Overexpression of lncRNA HULC attenuates myocardial ischemia/reperfusion injury in rat models and apoptosis of hypoxia/reoxygenation cardiomyocytes via targeting miR-377-5p through NLRP3/Caspase1/IL1β signaling pathway inhibition. Immunol Invest. Jul 17–2020.(Epub ahead of print). View Article : Google Scholar

36 

Ouyang M, Lu J, Ding Q, Qin T, Peng C and Guo Q: Knockdown of long non-coding RNA PVT1 protects human AC16 cardiomyocytes from hypoxia/reoxygenation-induced apoptosis and autophagy by regulating miR-186/Beclin-1 axis. Gene. 754:1447752020. View Article : Google Scholar : PubMed/NCBI

37 

Zhang P, Li S, Chen Z, Lu Y and Zhang H: LncRNA SNHG8 promotes proliferation and invasion of gastric cancer cells by targeting the miR-491/PDGFRA axis. Hum Cell. 33:123–130. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Zhang Y and Bian Y: Long non-coding RNA SNHG8 plays a key role in myocardial infarction through affecting hypoxia-induced cardiomyocyte injury. Med Sci Monit. 26:e9240162020. View Article : Google Scholar : PubMed/NCBI

39 

Wu N, Zhang X, Du S, Chen D and Che R: Upregulation of miR-335 ameliorates myocardial ischemia reperfusion injury via targeting hypoxia inducible factor 1-alpha subunit inhibitor. Am J Transl Res. 10:4082–4094. 2018.PubMed/NCBI

40 

Zhang Y, Fang J and Ma H: Inhibition of miR-182-5p protects cardiomyocytes from hypoxia-induced apoptosis by targeting CIAPIN1. Biochem Cell Biol. 96:646–654. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Xiao X, Lu Z, Lin V, May A, Shaw DH, Wang Z, Che B, Tran K, Du H and Shaw P: MicroRNA miR-24-3p reduces apoptosis and regulates Keap1-Nrf2 pathway in mouse cardiomyocytes responding to ischemia/reperfusion injury. Oxid Med Cell Longev. 2018:70421052018. View Article : Google Scholar : PubMed/NCBI

42 

Lin J, Lin H, Ma C, Dong F, Hu Y and Li H: MiR-149 aggravates pyroptosis in myocardial ischemia-reperfusion damage via silencing FoxO3. Med Sci Monit. 25:8733–8743. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Sheng Z, Lu W, Zuo Z, Wang D, Zuo P, Yao Y and Ma G: MicroRNA-7b attenuates ischemia/reperfusion-induced H9C2 cardiomyocyte apoptosis via the hypoxia inducible factor-1/p-p38 pathway. J Cell Biochem. 120:9947–9955. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Liu X, Zhang L, Qin H, Han X, Zhang Z, Zhang Z, Qin SY and Niu J: Inhibition of TRAF3 expression alleviates cardiac ischemia reperfusion (IR) injury: A mechanism involving in apoptosis, inflammation and oxidative stress. Biochem Biophys Res Commun. 506:298–305. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Pereira RM, Mekary RA, da Cruz Rodrigues KC, Anaruma CP, Ropelle ER, da Silva ASR, Cintra DE, Pauli JR and de Moura LP: Protective molecular mechanisms of clusterin against apoptosis in cardiomyocytes. Heart Fail Rev. 23:123–129. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Banjara S, D Sa J, Hinds MG and Kvansakul M: The structural basis of Bcl-2 mediated cell death regulation in hydra. Biochem J. 477:3287–3297. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2021
Volume 24 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
Liu Y, Zhou P, Wang F, Zhang X, Yang D, Hong L and Ruan D: Inhibition of lncRNA <em>SNHG8</em> plays a protective role in hypoxia-ischemia-reoxygenation-induced myocardial injury by regulating miR-335 and RASA1 expression. Mol Med Rep 24: 597, 2021
APA
Liu, Y., Zhou, P., Wang, F., Zhang, X., Yang, D., Hong, L., & Ruan, D. (2021). Inhibition of lncRNA <em>SNHG8</em> plays a protective role in hypoxia-ischemia-reoxygenation-induced myocardial injury by regulating miR-335 and RASA1 expression. Molecular Medicine Reports, 24, 597. https://doi.org/10.3892/mmr.2021.12236
MLA
Liu, Y., Zhou, P., Wang, F., Zhang, X., Yang, D., Hong, L., Ruan, D."Inhibition of lncRNA <em>SNHG8</em> plays a protective role in hypoxia-ischemia-reoxygenation-induced myocardial injury by regulating miR-335 and RASA1 expression". Molecular Medicine Reports 24.2 (2021): 597.
Chicago
Liu, Y., Zhou, P., Wang, F., Zhang, X., Yang, D., Hong, L., Ruan, D."Inhibition of lncRNA <em>SNHG8</em> plays a protective role in hypoxia-ischemia-reoxygenation-induced myocardial injury by regulating miR-335 and RASA1 expression". Molecular Medicine Reports 24, no. 2 (2021): 597. https://doi.org/10.3892/mmr.2021.12236