Abstract

Purpose. Long non-coding RNAs (LncRNAs) OIP5-AS1 and miR-25-3p play important roles in myocardial injury, whereas their roles in lipopolysaccharide (LPS)-induced myocardial injury remain unknown. The purpose of our study was to investigate the functional mechanisms of OIP5-AS1 and miR-25-3p in LPS-induced myocardial injury. Methods. Rats and H9C2 cells were treated with LPS to establish the model of myocardial injury in vivo and in vitro, respectively. The expression levels of OIP5-AS1 and miR-25-3p were determined by quantitative reverse transcriptase-polymerase chain reaction. Enzyme-linked immunosorbent assay was performed to measure the serum levels of IL-6 and TNF-α. The relationship between OIP5-AS1 and miR-25-3p/NOX4 was determined by luciferase reporter assay and/or RNA immunoprecipitation assay. The apoptosis rate was detected by flow cytometry, and cell viability was detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay. Western blot was performed to detect the protein levels of Bax, Bcl-2, caspase3, c-caspase3, NOX4, and p-NF-κB p65/NF-κB p65. Results. OIP5-AS1 was up-regulated, and miR-25-3p was down-regulated in myocardial tissues of LPS-induced rats and LPS-treated H9C2 cells. Knockdown of OIP5-AS1 relieved the myocardial injury in LPS-induced rats. Knockdown of OIP5-AS1 also inhibited the inflammation and apoptosis of myocardial cells in vivo, which was subsequently confirmed by in vitro experiments. In addition, OIP5-AS1 targeted miR-25-3p. MiR-25-3p mimics reversed the effects of OIP5-AS1 overexpression on promoting cell apoptosis and inflammation and on inhibiting cell viability. Besides, miR-25-3p mimics blocked the NOX4/NF-κB signalling pathway in LPS-induced H9C2 cells. Conclusion. Silencing of lncRNA OIP5-AS1 alleviated LPS-induced myocardial injury by regulating miR-25-3p.

1. Introduction

Myocardial injury is an important cause of adverse cardiovascular outcomes following myocardial ischemia and circulatory arrest [1]. The underlying pathogenesis of myocardial injury includes exaggerated inflammation and oxidative stress, and changes in the mitochondrial permeability transition pores [2]. Although surgery and various drugs have been widely used in the treatment of myocardial injury, the therapeutic efficacy remains unsatisfactory [35]. Thus, it is imperative to develop more effective therapeutic strategies for myocardial injury.

Long non-coding RNAs (lncRNAs), comprising at least 200 nucleotides, are a subset of non-coding RNA transcripts with multiple known regulatory functions [2]. Recent studies have proved that lncRNAs are important regulators in myocardial injury. For example, lncRNA KCNQ1OT1 attenuates lipopolysaccharide (LPS)-induced myocardial injury via increasing the viability and decreasing the apoptosis of injured cardiomyocytes [6]. lncRNA NEAT1 promotes the progression of septic myocardial cell injury by targeting miR-144-3p [3] or miR-590-3p [4]. lncRNA CYTOR attenuates LPS-induced myocardial injury by regulating miR-24/XIAP axis [5]. Previous studies have also reported that lncRNA OIP5 antisense RNA 1 (OIP5-AS1) promotes endothelial vascular injury by sponging miR-195-5p [7] and mitigates reactive oxygen species (ROS)-driven mitochondrial injury and apoptosis following myocardial injury [8]. In addition, Niu et al. have revealed that OIP5-AS1 attenuates myocardial ischemia/reperfusion (I/R) injury via regulating miR-29a-SIRT1/AMPK/PGC1α pathway [6]. However, the potential role of OIP5-AS1 in LPS-induced myocardial injury remains unclear. Because the action mechanisms of OIP5-AS1 in myocardial injury are complex, more downstream axes still need to be revealed.

MicroRNAs (miRNAs) are also important regulators in the pathogenesis and progression of myocardial injury and related diseases [9, 10]. For example, miR-214 alleviates myocardial injury in a septic mouse model [11]. Down-regulation of miR-23b prevents the cardiac dysfunction associated with polymicrobial sepsis [12]. MiR-146a mitigates LPS-induced myocardial injury by inhibiting NF-κB activation and the production of inflammatory factors [8]. Additionally, miR-25 protects cardiomyocytes against hypoxia/reoxygenation (H/R)-induced fibrosis and apoptosis [13], and inhibits LPS-induced apoptosis of cardiomyocytes by targeting PTEN [14]. However, the interaction between OIP5-AS1 and miR-25-3p in the pathogenesis of myocardial injury is rarely known.

In this study, we determined the expression levels of OIP5-AS1 and miR-25-3p in LPS-induced myocardial cells, evaluated their possible interaction, and further analyzed the potential downstream pathways of miR-25-3p in vitro. We also explored the function of OIP5-AS1 in a rat model of LPS-induced myocardial injury. Our results may provide a novel therapeutic target for myocardial injury.

2. Methods

2.1. Animals

Sprague–Dawley rats () with an weight of were purchased from the Beijing Vital River Laboratory Animal Technologies Co. Ltd. All rats were housed in an independent environment at room temperature with a 12 hour light/dark cycle and free access to food and water. This study was approved by the ethics committee of our hospital, and the experimental procedures complied with the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

2.2. Establishment of a Rat Model of LPS-Induced Myocardial Injury

The short hairpin RNA against OIP5-AS1 (sh-OIP5-AS1) and the negative control (sh-NC) were synthesized by the GeneChem Company (Shanghai, China) and packaged with lentiviral vector pGLV3-GFP (LV-sh-OIP5-AS1 and LV-sh-NC). To knockdown OIP5-AS1 in vivo, rats were infected with plaque forming units (PFUs) LV-sh-OIP5-AS1 (in 50 μL phosphate-buffered saline [PBS]; a retention time of more than one month in vivo) via intratracheal injection for 7 days before LPS (L2630, Sigma–Aldrich, Shanghai, China) treatment, and sepsis was then triggered by peritoneal injection of LPS, as previously described [12]. Briefly, rats were randomly divided into four groups ( in each group): sham, LPS Blank, LPS + sh-NC, and LPS + sh-OIP5-AS1 group. Rats in the experimental groups were intravenously injected with LPS (5 mg/kg, in 100 μL PBS), whereas rats in the sham group were injected with 100 μL PBS. At the end of the experiment, all rats were euthanized via injecting phenobarbital sodium at a dosage of 100 mg/kg. Blood samples (5 mL) were collected from the abdominal aorta of rats, placed for 30 minutes, and centrifuged at 3000 rpm for 10 minutes. The serum samples (supernatant) were used for enzyme-linked immunosorbent assay (ELISA). In addition, myocardial tissues were obtained from the left ventricle of the heart and subjected to hematoxylin and eosin (H&E) staining and quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR).

2.3. Cell Culture

An embryonic rat ventricular myocardial cell line (H9C2) was purchased from the American Type Culture Collection (Manassas, VA, USA), and cultured in Dulbecco’s Modified Eagle’s Medium supplemented with 10% fetal bovine serum, 100 IU/mL penicillin, and 10 μg/mL streptomycin. H9C2 cells were maintained in a humidified incubator supplied with 5% CO2 at 37°C (Supplementary Figure S1).

2.4. Transfection

OIP5-AS1 overexpression plasmid was obtained from GeneChem Company, and the miR-25-3p mimics and its counterpart negative control were synthesized by the Life Technologies Corporation (Carlsbad, CA, USA). The OIP5-AS1 shRNAs (a retention time of about one week in vitro) and overexpression plasmids were transfected into cells using Lipofectamine 3000 (Thermo Fisher Scientific, Carlsbad, CA, USA) for 48 hours following the manufacturer’s instructions.

2.5. Luciferase Reporter Assay

The 3 untranslated region (3-UTR) of OIP5-AS1/NOX4 containing the predicted binding sequences of miR-25-3p was synthesized and cloned into psiCHECK-2 vector (Promega, Madison, WI, USA), referring as OIP5-AS1/NOX4 wild-type (WT). The 3-UTR of OIP5-AS1/NOX4 containing the mutant binding sequences of miR-25-3p was constructed as OIP5-AS1/NOX4 mutation (MUT). Afterward, miRNA mimics/NC was transfected into the cells with OIP5-AS1/NOX4 WT/MUT using Lipofectamine 3000 (Thermo Fisher Scientific). Relative luciferase activity was finally measured by a Multimode Detector (Beckman Coulter, Fullerton, CA, USA).

2.6. RNA Immunoprecipitation Assay

H9C2 cells were transfected with miR-25-3p mimics. After 48 hours of transfection, RNA immunoprecipitation (RIP) assay was performed using the Magna RIP RNA-Binding Protein Immunoprecipitation Kit (Millipore, Bedford, MA, USA). RIP lysis buffer supplemented with protease and RNase inhibitor was used to lyse the cells. Afterward, the cell extracts were incubated with RIP buffer containing magnetic beads conjugated with anti-Ago2 antibody (Cell Signaling, Danvers, MA, USA) or negative control Immunoglobulin G (IgG) (Millipore). The co-precipitated RNAs were then synthesized into complementary DNA (cDNA) and evaluated by qRT-PCR.

2.7. Enzyme-Linked Immunosorbent Assay

The levels of TNF-α and IL-6 were measured using a rat TNF-α ELISA kit (catalog No: K1052-100; Biovision, San Francisco, CA, USA) and a rat IL-6 ELISA kit (catalog No: K4145-100; Biovision), respectively. The contents of creatine kinase (CK)-MB and cardiac troponin I (cTnI) in the serum of rats were measured using a CK-MB isoenzyme Assay Kit (catalog No: H197-1-1; Nanjing Jiancheng Bioengineering, Inc., Nanjing, China) and a rat Cardiac Troponin I ELISA ELISA kit (catalog No: ab246529; Abcam, Cambridge, MA, USA), respectively.

2.8. Quantitative Reverse Transcriptase-Polymerase Chain Reaction

Total RNAs were extracted from tissues and cells using TRIzol reagent (Life Technologies Corporation). A Bio-Rad SYBR Green PCR Master Mix (Bio-Rad, Hercules, CA, USA) was then used for qRT-PCR. The primers were listed as follows: OIP5-AS1, forward: 5-AAAGCAAGGTCTCCCCACAAG-3, reverse: 5-GGTCTGTGCTAGATCAAAAGGCA-3; miR-25-3p, forward: 5-CATTGCACTTGTCTCGGTCTGA-3; reverse: 5-GCTGTCAACGATACGCTACGTAACG-3; glyceraldehyde-3-phosphate dehydrogenase (GAPDH), forward: 5-TCCGCCCCTTCCGCTGATG-3, reverse: 5-CACGGAAGGCCATGCCAGTGA-3; and U6, forward: 5- CTCGCTTCGGCAGCACA-3, reverse: 5-AACGCTTCAGAATTTGCGT-3. The expression of miR-25-3p was normalized to U6, and the expression of OIP5-AS1 was normalized to GAPDH. The fold changes were calculated following the 2ΔΔCt method.

2.9. H&E Staining

The myocardial tissues from rats were fixed in 10% neutral formaldehyde solution for 24 hours at 4°C and then dehydrated and vitrified. Following this, they were embedded in paraffin and cut into 6 μm sections. The sections were then de-waxed and stained with H&E. Finally, the stained sections were imaged under a BX50 bioluminescent microscope (Olympus, Tokyo, Japan) to reveal the morphological changes.

2.10. TUNEL Staining

The apoptosis of myocardial cells in rats was detected using an In Situ Cell Death Detection Kit (Roche, Basel, Switzerland) according to the manufacturer’s instructions. Simply, the paraffin-embedded sections were dewaxed with xylene, dehydrated with gradient alcohol, incubated with 20 μg/mL protease K to enhance membrane permeability, and further incubated with 3% H2O2 to block endogenous peroxidase. Subsequently, the sections were incubated with TUNEL reaction mixture for 1 hour at 37°C under darkness. After counterstained with 4,6-diamino-2-phenyl indole (DAPI), the fluorescence was observed under a microscope (Olympus).

2.11. Western Blot

Cells were lysed in RIPA buffer (Cell Signaling Technology), and equivalent amounts of protein extracts were separated using 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS–PAGE). Protein samples were then transferred to polyvinylidene fluoride membranes and blocked with 5% non-fat milk. These membranes were then incubated with primary antibodies, including rabbit anti-Bcl-2 (1 : 2000, ab182858, Abcam), rabbit anti-Bax (1 : 1000, ab32503, Abcam), rabbit anti-c-caspase3 (1 : 500, ab13847, Abcam), rabbit anti-caspase 3 (1 : 5000, ab32351, Abcam), rabbit anti-NOX4 (1 : 2000, ab133303, Abcam), rabbit NF-κB p65 (1 : 2000, ab32536, Abcam), rabbit p-NF-κB p65 (1 : 1000, ab194726, Abcam), and rabbit anti-β-actin (1 : 200; ab115777, Abcam) overnight at 4°C. Afterward, the membranes were gently washed and then incubated with horseradish peroxidase-labelled secondary antibody (goat anti-rabbit, 1 : 2000, ab205718, Abcam) at 25°C for 1 hour. Finally, relative expression level was quantified using a Gel-Pro Analyzer (Media Cybernetics) and normalized to β-actin.

2.12. Cellular Apoptosis Assay

Cell apoptosis was examined by annexin V-phycoerythrin staining (BD Science, Franklin Lakes, NJ, USA). Briefly, cells were rinsed in PBS and stained with annexin V-PE solution for 25 minutes at 37°C in the dark. Subsequently, the stained cells were detected on a flow cytometry (Beckman Coulter), and the data were analyzed using the FlowJo software (Tree Star, Ashland, OR, USA).

2.13. Cell Viability Assay

Cell viability was detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay. Cells were plated into 96-well plates at 3000 cells/100 μL/per well and treated with LPS and/or transfected with mimics or vectors. These samples then incubated with 10 μL MTT (5 mg/mL) for 4 hours at 37°C. Following this, the samples were further incubated with 150 μL dimethyl sulfoxide for 10 minutes. The optical density of each well was measured at 490 nm using a microplate reader (MG Labtech, Durham, NC, USA).

2.14. Statistical Analysis

Each experiment was performed in at least triplicates, and the data were presented as the mean ± standard deviation. One-way analysis of variance (one-way ANOVA) was used for the comparisons among multiple groups (a test for Gaussian distribution was applied before any analysis), and Tukey’s multiple comparisons test was used for pairwise comparisons. The t-test was used for comparisons between two groups. The results were considered statistically significant if the -value was <0.05.

3. Results

3.1. Construction of a Rat Model of Myocardial Injury

The LPS-induced myocardial injury was first assessed in rats. H&E staining showed that LPS treatment resulted in irregular arrangement, myofibrillar fragmentation, and cardiocyte degeneration in myocardial tissues (Figure 1(a)). TUNEL staining revealed more apoptotic myocardial cells in the LPS group than those in the sham group (Figure 1(b)). CK-MB and cTnl are biomarkers of myocardial injury. LPS-induced pro-inflammatory cytokines, IL-6 and TNF-α, are also correlated with myocardial injury [15]. We detected the serum levels of CK-MB, cTnl, IL-6, and TNF-α. The results showed that these indexes in LPS-treated rats were remarkably increased compared with those in control rats (; Figures 1(c), 1(d), 1(e), and 1(f)). These results indicated that the rat model of LPS-induced myocardial injury was successfully constructed.

3.2. OIP5-AS1 Is Up-Regulated by LPS in Myocardial Tissues, and Knockdown of OIP5-AS1 Suppresses Inflammation in LPS-Induced Rats

Subsequently, we investigated the expression of sh-OIP5-AS1. qRT-PCR results demonstrated that OIP5-AS1 in myocardial tissues was up-regulated in LPS-induced rats (; Figure 2(a)). The efficiency of OIP5-AS1 knockdown was evaluated, and sh-OIP5-AS1 significantly decreased the expression of OIP5-AS1 in myocardial tissues (; Figure 2(b)). In addition, the serum levels of IL-6 and TNF-α (two inflammatory factors) were increased by the treatment of LPS in a time-dependent manner (). The increased serum levels of IL-6 and TNF-α at different time points were all significantly weakened by the intervention of sh-OIP5-AS1 in LPS-induced rats (; Figures 2(c) and 2(d)). Similarly, LPS-induced elevation of CK-MB and cTnl levels was significantly offset by sh-OIP5-AS1 (; Figures 2(e) and 2(f)). Besides, as illustrated in Figure 2(g), H&E staining demonstrated that knockdown of OIP5-AS1 alleviated LPS-induced inflammatory cell infiltration, irregular arrangement, and cardiomyocyte degeneration. TUNEL staining also determined that the LPS-induced myocardial apoptosis was weakened by the down-regulation of OIP5-AS1 (Figure 2(h)).

3.3. Sh-OIP5-AS1 Inhibits LPS-Induced Production of Inflammatory Cytokines in H9C2 Cells

Next, we investigate the role of OIP5-AS1 in H9C2 cells and found that the relative expression of OIP5-AS1 was increased in LPS-treated H9C2 cells (; Figure 3(a)). We then silenced OIP5-AS1 and discovered that cells transfected with sh-OIP5-AS1-1 and sh-OIP5-AS1-2 both exhibited decreased OIP5-AS1 expression compared with the control (; Figure 3(b)). We also measured the levels of two inflammatory cytokines, including IL-6 and TNF-α. The levels of IL-6 and TNF-α in LPS-treated cells were approximately three times higher than those in the control group. Meanwhile, the promoting effects of LPS on the levels of IL-6 and TNF-α were reversed by sh-OIP5-AS1-1 (; Figures 3(c) and 3(d)). In summary, knockdown of OIP5-AS1 could partially attenuate LPS-induced production of inflammatory cytokines, including IL-6 and TNF-α in H9C2 cells.

3.4. Knockdown of OIP5-AS1 Inhibits the Apoptosis and Promotes the Proliferation of LPS-Treated H9C2 Cells

To explore the function of OIP5-AS1 on the apoptosis of H9C2 cells, OIP5-AS1 was knockdown, and the apoptosis rate of H9C2 cells exposed to LPS was detected. Flow cytometry assay demonstrated that the apoptosis rate of H9C2 cells was increased after LPS treatment, whereas it was decreased due to the transfection of sh-OIP5-AS1-1 (; Figure 4(a)). Bcl-2 is an apoptotic suppressor, whereas c-caspase 3 is a promoter of apoptosis [16, 17]. We then measured the expression of these two proteins in H9C2 cells using western blot. The results showed that the expression of Bcl-2 was decreased, and the expression of Bax and c-caspase3/caspase3 ratio was increased following LPS treatment. Moreover, the effects of LPS on these proteins were reversed by the transfection of sh-OIP5-AS1-1 in H9C2 cells (; Figure 4(b)). In addition, knockdown of OIP5-AS1 also reversed LPS-induced inhibition on the viability of H9C2 cells (; Figure 4(c)). Taken together, these results indicated that the knockdown of OIP5-AS1 could prevent H9C2 cells from LPS-induced apoptosis and inhibition of cell proliferation.

3.5. OIP5-AS1 Directly Targets miR-25-3p

Based on the predicting results of starBase4.0, WT OIP5-AS1 could target miR-25-3p (; Figure 5(a)). The expression of miR-25-3p was increased by up to threefold after OIP5-AS1 knockdown (; Figure 5(b)). The transfection of miR-25-3p mimics effectively up-regulated miR-25-3p in cells (; Figure 5(c)). The potential interaction between OIP5-AS1 and miR-25-3p was then verified by luciferase reporter assay. Cells co-transfected with OIP5-AS1-wt and miR-25-3p mimics displayed decreased relative luciferase activity compared with other groups, which indicated that OIP5-AS1 interacted with miR-25-3p (; Figure 5(d)). Furthermore, the expression of miR-25-3p was negatively correlated with the expression of OIP5-AS1 (; Figure 5(f)). This finding was supported by the results of RIP assay that more OIP5-AS1 was co-precipitated when miR-25-3p was overexpressed. These results indicated that OIP5-AS1 directly interacts with miR-25-3p.

3.6. The Regulatory Role of OIP5-AS1 in Inflammation Is Associated with miR-25-3p

MiR-25-3p was down-regulated both in LPS-induced rats and H9C2 cells compared with controls (; Figures 6(a) and 6(b)). Relative expression of OIP5-AS1 in H9C2 cells was significantly increased after transfection with pcDNA-OIP5-AS1 (; Figure 6(c)). Overexpression of miR-25-3p reduced IL-6 and TNF-α levels in LPS-treated H9C2 cells. The same effects of miR-25-3p were also discovered even though OIP5-AS1 was overexpressed in LPS-treated H9C2 cells, which indicated that OIP5-AS1 contributed to LPS-induced inflammation via regulating miR-25-3p (; Figures 6(d) and 6(e)). Taken together, these results indicated that the regulatory effect of OIP1-AS1 on LPS-induced inflammation was associated with miR-25-3p.

3.7. OIP5-AS1 Promotes Cellular Apoptosis and Represses Cellular Proliferation via Regulating miR-25-3p in LPS-Induced H9C2 Cells

The apoptosis of LPS-induced H9C2 cells was significantly repressed by miR-25-3p mimics in cells transfected or not transfected with pcDNA-OIP5-AS1 (; Figure 7(a)). Western blot assays showed that overexpression of miR-25-3p increased Bcl-2 expression, and reduced Bax expression and the c-caspase3/caspase3 ratio in LPS-induced H9C2 cells regardless of OIP5-AS1 overexpression or not (; Figure 7(b)). The viability of LPS-induced H9C2 cells was increased when cells were transfected with miR-25-3p mimics (; Figure 7(c)). Collectively, these results demonstrated that OIP5-AS1 promoted the apoptosis and repressed the proliferation of LPS-induced H9C2 cells via regulating miR-25-3p.

3.8. MiR-25-3p Blocks the NOX4/NF-κB Signalling Pathway in LPS-Induced H9C2 Cells

To further reveal the downstream mechanisms of miR-25-3p in myocardial injury, the target mRNAs of miR-25-3p were predicated by miRDB. NOX4 was determined as a potential target of miR-25-3p (Figure 8(a)). Luciferase reporter assay showed that the luciferase activity was lower in cells co-transfected with NOX4 WT and miR-25-3p mimics than those co-transfected with NOX4 WT and miR-NC (; Figure 8(b)), illustrating the target relationship between NOX4 and miR-25-3p. In addition, the protein expression of NOX4 and p-NF-κB p65/NF-κB p65 was elevated in LPS-induced H9C2 cells compared with the controls (). The transfection of miR-25-3p mimics significantly decreased the high expression of NOX4 and p-NF-κB p65/NF-κB p65 in LPS-induced H9C2 cells (; Figure 8(c)).

4. Discussion

Myocardial injury is a serious pathological change associated with a high risk of mortality. Until now, many strategies have been developed in establishing the model of myocardial injury, such as isoproterenol [18], Adriamycin [13], I/R [14], and sepsis [19]. Since myocardial injury is one of the dominant symptoms of sepsis, the model of sepsis-induced myocardial injury is widely used in animal and cell experiments [20]. In inducing the model of sepsis-induced myocardial injury, LPS is the most commonly used agent with the advantages of simplicity, repeatability, and rapidity, which can activate acute inflammatory response, oxidative stress, and apoptosis [16]. In this study, the model of LPS-induced myocardial injury was established in rats and H9C2 cells. We demonstrated that OIP5-AS1 was up-regulated, but miR-25-3p was down-regulated by the treatment of LPS in rats and H9C2 cells. Loss-of-function analyses indicated that knockdown of OIP5-AS1 relieved LPS-induced apoptosis and inflammation, and LPS-induced inhibition of proliferation by interacting with miR-25-3p. Besides, the NOX4/NF-κB signalling pathway was blocked by miR-25-3p in LPS-induced H9C2 cells.

Recent studies have shown that myocardial injury is associated with the abnormal expression of diverse lncRNAs [17, 21]. OIP5-AS1 is a lncRNA that overexpressed in a human umbilical vein endothelial cell (HUVEC) model of atherosclerosis. In HUVECs treated with oxidative low-density lipoprotein, OIP5-AS1 promotes apoptosis and inhibits proliferation [22]. In our study, we demonstrated that OIP5-AS1 was up-regulated in a rat model of LPS-induced myocardial injury and also in LPS-induced H9C2 cells. In addition, knockdown of OIP5-AS1 increased the viability and decreased the apoptosis of LPS-induced H9C2 cells, indicating that OIP5-AS1 may be involved in the progression of myocardial injuries. It has been demonstrated that down-regulation of OIP5-AS1 promotes cell proliferation and suppresses apoptosis in oxidative low-density lipoprotein-mediated endothelial cell injury [23]. OIP5-AS1 can also regulate the inflammatory response. In LPS-activated nucleus pulposus cells, inflammation response was repressed when OIP5-AS1 was silenced [24], which is in agreement with our results that knockdown of OIP5-AS1 led to a decrease in the expression of IL-6 and TNF-α. Similar expression alteration of OIP5-AS1 and relevant effects on cell proliferation, apoptosis, and inflammation are also observed in LPS-induced acute lung injury [6]. Thus, silencing of OIP5-AS1 may be a suppressor in LPS-induced myocardial injury and a meaningful target for treatments.

MiRNAs have drawn increasing attention as possible therapeutic targets for myocardial injury [9, 25, 26]. MiR-25-3p is poorly expressed in ox-LDL-induced coronary vascular endothelial cells (CVECs) and vascular tissues. Exosomal miRNA-25-3p derived from platelets represses the inflammatory response in CVECs [27]. MiR-25 protects cardiomyocytes from LPS-induced injury via regulating the expression of inflammatory cytokines [28]. In addition, it has been reported that miR-25 is decreased in LPS-induced cardiomyocytes and its overexpression can inhibit the apoptosis of cardiomyocyte [29]. These results are consistent with our findings that miR-25-3p was down-regulated in LPS-induced H9C2 cells, and its overexpression suppresses the apoptosis and the production of inflammatory cytokines (IL-6 and TNF-α), and promotes the viability of LPS-induced H9C2 cells. Taken together, our results indicate that miR-25-3p may relieve LPS-induced myocardial injury by inhibiting inflammation and apoptosis, and promoting proliferation.

MiR-25-3p has also been reported to interact with various lncRNAs in diverse diseases [3033]. LncRNA can act as competitive endogenous RNA via directly sponging target miRNAs. Here, we found that lncRNA OIP5-AS1 interacted with miR-25-3p, which reduced the expression of miR-25-3p in LPS-treated rat and H9C2 cells. Overexpression of miR-25-3p repressed apoptosis and inflammatory response, and enhanced proliferation in the presence of overexpressed OIP5-AS1. These results indicate that OIP5-AS1 may regulate cell apoptosis, proliferation, and inflammatory response by sponging miR-25-3p. Similar interactions between miR-25-3p and other lncRNAs in cardiovascular diseases have also been reported in previous studies, for example, MALAT1 plays an essential role in myocardial infarction by sponging miR-25-3p [34]. Here, we identified the regulatory interaction of OIP5-AS1 with miR-25-3p in LPS-induced myocardial injury.

LncRNAs and miRNAs are involved in the progression of myocardial injury by targeting diverse mRNAs. It has been reported that Krüppel-like factor 4 (KLF4) is targeted by miR-25-3p, and overexpression of KLF4 promotes hypoxia-induced injury in H9C2 cells [35]. Up-regulation of miR-25-3p facilitates the activation of the PI3K/Akt pathway and represses myocardial I/R injury [8]. In this study, NOX4 was determined as a downstream target of miR-25-3p. The following assays revealed that miR-25-3p mimics blocked the NOX4/NF-κB signalling pathway in LPS-induced H9C2 cells. NOX4 is a key enzyme for the production of ROS, which contributes to myocardial injury through inducing ROS [36]. Fan et al. have found that CAPE-pNO2 relieves heart injury in mice with diabetic cardiomyopathy via inhibiting the NOX4/NF-κB pathway [37]. Therefore, the blocking of the NOX4/NF-κB pathway may be involved in the action mechanisms of OIP5-AS1-miR-25-3p axis in LPS-induced myocardial injury.

However, this study still has some limitations. For example, whether there is a correlation between OIP5-AS1 expression and clinical characteristics of myocardial injury in human. The function of the NOX4/NF-κB pathway in LPS-induced myocardial injury is not verified. The NOX4/NF-κB pathway is also not the only downstream target of OIP5-AS1-miR-25-3p. More downstream regulators involving cell inflammation, apoptosis, and proliferation in LPS-induced myocardial injury remain need to be studied. In addition, the upstream mechanisms of OIP5-AS1 in LPS-induced myocardial injury are also unclear. The up-regulation of lncRNAs involves frequent somatic copy number alteration, transcription factors, histone modification, and posttranscriptional destabilization [3841]. Therefore, the upstream mechanisms of LPS-induced up-regulation of OIP5-AS1 also need further investigation.

In summary, our study demonstrates that knockdown of OIP5-AS1 may function as a suppressor in apoptosis and inflammation, and as an inhibitor in proliferation in LPS-induced myocardial injury via down-regulating miR-25-3p.

Data Availability

Data supporting this research article are available from the corresponding author or first author on reasonable request.

Ethical Approval

The animal experiments were approved by the Ethics Committee of Suzhou Ninth People’s Hospital (Jiangsu, China), and the experimental procedures complied with the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

Conflicts of Interest

The author(s) declare(s) that they have no conflicts of interest.

Authors’ Contributions

JM conceived and designed the present study. HQ and HZ performed the experiments, analyzed the data, and drafted the article. JM revised the article critically for important intellectual content. JM, HQ, and HZ confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Supplementary Materials

Figure S1. The morphology of H9C2 cells. (Supplementary Materials)