miR‑124 inhibits cardiomyocyte apoptosis in myocardial ischaemia‑reperfusion injury by activating mitochondrial calcium uniporter regulator 1

  • Authors:
    • Linlin Guo
    • Chaoying Liu
    • Chunyan Jiang
    • Yanhan Dong
    • Lynn Htet Htet Aung
    • Han Ding
    • Yanyan Gao
  • View Affiliations

  • Published online on: June 13, 2023     https://doi.org/10.3892/mmr.2023.13031
  • Article Number: 144
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Mitochondria‑mediated apoptosis is the primary cause of cardiomyocyte death. Therefore, mitochondria are a key target for treating myocardial injury. Mitochondrial calcium uniporter regulator 1 (MCUR1)‑mediated mitochondrial calcium homeostasis markedly promotes cell proliferation and resistance to apoptosis. However, whether MCUR1 is involved in regulation of cardiomyocyte apoptosis during myocardial ischaemia‑reperfusion remains unknown. microRNA‑124 (miR‑124) is upregulated in cardiovascular disease, suggesting a key role for miR‑124 in the cardiovascular system. Whether miR‑124 affects cardiomyocyte apoptosis and myocardial infarction is not well understood. Western blot showed that miR‑124 and MCUR1 were upregulated in cardiomyocyte apoptosis induced by hydrogen peroxide (H2O2). Flow cytometry assay of cell apoptosis showed that miR‑124 inhibited cardiomyocyte apoptosis by activating MCUR1 following H2O2 treatment. The dual‑luciferase reporter assay confirmed binding of miR‑124 to MCUR1 3'‑UTR and subsequent activation of MCUR1. FISH assay revealed the entry of miR‑124 into the cell nucleus. Therefore, MCUR1 was identified as a novel target of miR‑124, and it was shown that the miR‑124‑MCUR1 axis modulated cardiomyocyte apoptosis induced by H2O2 in vitro. The results indicated induced expression of miR‑124 during acute myocardial infarction and its transport to the nucleus. In the nucleus, miR‑124 transcriptionally activated MCUR1 by binding to its enhancers. These findings reveal a role of miR‑124 as a biomarker for myocardial injury and infarction.

Introduction

Ischaemic heart disease, particularly myocardial infarction (MI), is the leading cause of mortality worldwide (1,2). The risk of 1-year mortality is increased by 7.5% for each 30-min delay in primary angioplasty for ST-segment elevation myocardial infarction (1). Free calcium ion (Ca2+) concentration in mitochondria may serve a key role in the regulation of myocardial ischaemia-reperfusion (I/R) injury, especially in regulation of Ca2+ balance (36). Mitochondria, as Ca2+ storage organelles, regulate Ca2+ homeostasis (7,8). This is important in cellular homeostasis (911). Mitochondrial Ca2+ maintains dynamic balance of cytoplasmic Ca2+ concentration, which is a key regulatory factor of mitochondrial respiration (12,13). Mitochondrial calcium uniporter (MCU) is a key component of the mitochondrial Ca2+ channel (1417). MCU regulator 1 (MCUR1) is a regulatory protein of MCU. MCUR1 is a key component of the mitochondrial unidirectional transporter complex required for mitochondrial Ca2+ uptake and maintenance of normal cell bioenergy. Normal Ca2+ expression and function are key for mitochondrial Ca2+ homeostasis (1823). MCUR1-mediated remodelling of the mitochondrial Ca2+ environment promotes proliferation and resistance to apoptosis, facilitating malignant progression of hepatoma cells (24,25). Thus, MCUR1 is key in the development of hepatocellular carcinoma. Under pathological conditions, mitochondrial Ca2+ uptake mediated by MCU activates the mitochondrial permeability transition pore, resulting in cell death during myocardial I/R (2633). However, the role of MCUR1 in myocardial I/R is unclear.

microRNAs (miRNAs or miRs) are small non-coding RNAs that mediate post-transcriptional gene modulation. miRNAs are key in aging and development of cancer and cardiovascular diseases. Increasing evidence indicates that miRNAs regulate cardiac balance and response to injury (3437). miR-124 is the most abundant miRNA, with a range of biological functions in the central nervous system (38). Previous studies have showed that miR-124 was elevated in acute myocardial infarction and was correlated with myocardial pathophysiology and cardiac function (3941). The expression of miR-124 is upregulated in smokers and is associated with increased risk of subclinical arteriosclerosis due to altered single-cell phenotypes (42). Randomised clinical trials have indicated that serum miR-124 levels may be a useful prognostic indicator of outcomes after cardiac arrest (43,44). Circulating miR-124 is upregulated in patients with acute coronary syndrome, with requirement for urgent coronary occlusion differentiating this syndrome from membranous inflammation (45). Although clinical and preclinical data indicate a key role of miR-124 in the cardiovascular system (4648), data that substantiate this role are limited. Whether miR-124 affects cardiomyocyte apoptosis and MI requires further investigation.

To investigate the role of miR-124 in cardiomyocyte apoptosis and MI, cardiomyocyte apoptosis was induced using hydrogen peroxide (H2O2) to simulate oxidative stress induced by I/R injury. We investigated the mechanism of miR-124 regulated during H2O2-induced cardiomyocyte apoptosis.

Materials and methods

Cell culture and transduction

The H9C2 cell line is a subclone of the original clonal cell line derived from embryonic BD1X rat heart tissue (49). H9C2 cells exhibit a number of skeletal muscle properties. Up to 95% of cells fused to form myotubes are characteristic of skeletal muscle (50). The length, diameter, and arrangement of sarcomeres and fine myofilaments in their myotubes are similar to those of developing skeletal and cardiac myofilaments in vitro. Cells were obtained from the Whole Gold Cell Bank (Beijing, China) and cultured in Dulbecco's Modified Eagle's Medium (Merck KGaA) containing 10% FBS (Thermo Fisher Scientific, Inc.) in an atmosphere of 95% oxygen and 5% carbon dioxide at 37°C. After starvation in serum-free DMEM (37°C) for 12 h, oxidative stress was induced by treatment at 37°C with 200 µM H2O2 for 0, 2, 4, 6, 8 and 10 h. Cells were collected at each time point for subsequent analysis.

RNA constructs miR-124 mimics (5′uaaggcacgcggugaaugcc3′), anti-miR-124 (5′ATCAAGGTCCGCTGTG3′) and corresponding negative controls (NCs) were purchased from GenePharma and transfected into H9C2 cells using FuGENE® HD (Promega Corporation) at room temperature for 15 min, according to the manufacturer's instructions. miRNA (10 µM) and 10 µl FuGENE co-transfected in H9C2 cells. Following incubation at 37°C for 48 h, subsequent experiments were performed. In addition, miR-124 mimic, MCUR1 3′-untranslated region (UTR) plasmid, an MCUR1 overexpression plasmid vector [pEX-3(pGCMV/MCS/Neo)] (GenScript), NC (mimic Ctrl and pcDNA3.1) and an MCUR1 small interfering RNA (siRNA, 5′GCCAGAGACAGACAAUACUTT3′; GenePharma) were transfected using FuGENE® HD (Promega Corporation).

Cell Counting Kit-8 (CCK-8) proliferation assay

Cells were harvested in the exponential growth phase and suspended in complete medium. Medium containing cells (100 µl) were added to the wells and incubated at 37°C for 24 h. Subsequently, 200 µM (12.5 µl) H2O2 was added. Each well was incubated with 10 µl CCK-8 (Beijing Solarbio; cat. no. CA1210) for 1–4 h. The signal was measured by microplate reader at an absorption wavelength of 450 nm and a reference wavelength of 600–650 nm. Six samples was tested in triplicate.

Dual-luciferase reporter assay

MCUR1 3′-UTR was cloned into the gp-mirGLO vector (GenScript) and mutant 3′-UTR was obtained by target mutation (GenScript). The luciferase reporter plasmid (gp-miRGLO, GenePharma) and miR-124 mimic (GenePharma; 5′-uaaggcacgcggugaaugcc3′)/miR-NC (5′UUCUCCGAACGUGUCACGUTT3′, GenePharma) were co-transfected into H9C2 cells (2×104) using FuGENE® HD (Promega Corporation) into H9C2 cells. Following incubation at 37°C for 48 h, a Dual-Glo® Luciferase Assay System (Promega Corporation, E2920) was used according to the manufacturer's instructions and luciferase activity was detected. Renilla luciferase activity was compared with that of the control group.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from H9C2 cells using TRIzol® (Thermo Fisher Scientific, Inc.) at room temperature according to the manufacturer's instructions. RNA was reverse-transcribed to cDNA using Titanium One-Step RT-PCR Kit (Clontech). qPCR was performed using SYBR® Premix Ex Taq II (Takara Bio, Inc.) and detected using an ABI 7,500 Fast Real-Time PCR System (Thermo Fisher Scientific, Inc.) with 50 ng cDNA and 500 nM each forward or reverse primers (Table I). The thermocycling conditions were as follows: 50°C for 1 h, 94°C for 5 min, 25–40 cycles (94°C for 30 sec, 65°C for 30 sec, 68°C for 1 min) and 68°C for 2 min. Data were normalized according to the ΔCq method (51). The reference genes is U6 or GAPDH.

Table I.

Sequences of primers used for reverse transcription-quantitative PCR.

Table I.

Sequences of primers used for reverse transcription-quantitative PCR.

GeneSequence, 5′→3′
microRNA-F: CGTGTTCACAGCGGAC
124-5PR: ATCAAGGTCCGCTGTG
U6F: CTCGCTTCGGCAGCACATATACT
R: ACGCTTCACGAATTTGCGTGTC
MCUR1 F:GCTCTACTTTGACACCCACGCTCTGG
R:GTCTCCGTAATCTTGACCAATGC
GAPDHF: GGACATTGTTGCCATCAACGA
R: GGGTAGAGTCATACTGGAACATGT

[i] MCUR1, mitochondrial calcium uniporter regulator 1; F, forward; R, reverse.

Western blotting

Protein was extracted from H9C2 cells using the RIPA extraction buffer (Beijing Solarbio Science & Technology Co., Ltd.) containing phenylmethylsulfonyl fluoride for 30 min and prepared to measure protein concentration by BCA. The same amount (15–40 µg) of protein was separated by 15% SDS-PAGE, transferred to a pre-treated PVDF membrane, blocked with 5% milk at room temperature for 1 h and incubated with primary antibody at 4°C (MCUR1 1:500, BOSTER, cat. no. A08547-1; β-actin 1:5,000, Beyotime, AF5003) over night. After washing with TBST (1% Tween-20) three times, the washed membrane was incubated with HRP-conjugated, goat anti-Rabbit IgG (Abbkin, cat. no. A21020, 1:10,000) at room temperature for 2.5 h and visualised using a chemiluminescence kit (Vazyme, E412-02). The results were visualized with a chemiluminescence analyser. Protein quantification was analysed using ImageJ software (ImageJ 1.51j8, National Institutes of Health).

Fluorescence in situ hybridisation (FISH)

Cell samples were fixed in paraformaldehyde (4%) at room temperature for 24 h and embedded in paraffin at 42°C, 4~8 h. Paraffin sections (3–4 µm) were removed using xylene, dehydrated and washed. FISH was performed according to a previously described protocol (52). Briefly, the slides were permeabilized by proteinase K (Solarbio, P1120) at 37°C for 20 min. The slides were incubated with pre-hybridization buffer 5×SSC (saline sodium citrate) for 1 h at 37°C. The hybridization buffer 5×SSC (Solarbio, S1030) was transferred to the sample, and a single gene probe (working solution, 50 ng/µl, synthesis by Shenggong Biotechnology Company) was added. Slides were incubated at 37°C overnight to hybridise the probe with target DNA. The slides were washed with 37°C preheated washing buffer (2×SSC) for 10 min, then washed with 37°C preheated washing buffer (1×SSC) for 10 min and 37°C preheated washing buffer (0.5×SSC) for 10 min. DAPI was incubated in the dark for 8 min and sealed with anti-fluorescence quenching sealing agent (Solarbio). The signal from each probe was observed under a Leica TCS SP8 MP laser scanning confocal microscope.

Flow cytometry measurement of apoptosis using Annexin-V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) staining

H9C2 cells were treated with 200 µM hydrogen peroxide at 37°C for 6 h and transfected with miRNA. Treated fresh cells were collected resuspended in 100 µl PBS. PI or Annexin-V-FITC or PI plus Annexin-V were added for staining. After 15 min incubation at 4°C, C6 flow cytometry was performed using a flow cytometer (BD accuri, C6 flow cytometry) equipped with FITC signal filtered by a FL1 detector at 530/30 nm and PI signal filtered by a FL2 detector at 585/42 nm. The flow cytometry data are expressed as percentages of initial cell count. The data was analyzed by FLOWJO v10 (Becton, Dickinson & Company) software.

Flow cytometry-based measurement of mitochondrial membrane potential

Cells were digested with trypsin, resuspended in PBS and treated with rhodamine 123 (5 mg/l) at 37°C for 30 min, and washed once with PBS followed by centrifugation at 300 g, 10 min) at room temperature. Flow cytometric analysis (BD Accuri C6 Flow Cytometer, BD Company) was used to detect cells at an excitation wavelength of 488 nm. The data was analyzed by FLOWJO v10 (Becton, Dickinson & Company) software.

Online databases analysis

TargetScan was used to predict miRNA-binding sites in mammals (targetscan.org/vert_80/). MiRanda (bioinformatics.com.cn/local_miranda_miRNA_target_prediction_120) and miRDB (mirdb.org/) online databases analysis was performed according to previously described protocol (53,54).

Statistical analysis

The experimental data were analysed using the GraphPad Prism 6.02 software (Dotmatics). All experiments were repeated three times (n=3). The data are presented as the mean ± SD. Independent-sample t test was used for comparisons between two groups. One-way ANOVA followed by Dunnett's post hoc test was used for comparisons between >2 groups. Pearson's correlation coefficient was used for correlation analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-124 and MCUR1 are upregulated in H2O2-induced cardiomyocyte apoptosis

Cardiomyocytes undergo apoptosis and necrosis induced by hypoxia and oxidative stress during coronary artery occlusion and subsequent MI (55,56). CCK-8 viability assay was used to test cell activity following H2O2 treatment (Fig. 1A). To determine the involvement of caspase-3 in H2O2-induced apoptosis in cardiomyocytes, cleaved caspase-3 was measured by western blotting (Fig. 1B). The release of cytochrome c into the cytoplasm is a key step in the apoptotic process and plays an important role in the apoptotic mechanism. We therefore measured the release of cytochrome c by western blot (Fig. 1B). Flow cytometry (Fig. 2G) showed H2O2 induced both apoptosis and necrosis in cardiomyocytes, and H2O2 was used to generate oxidative stress to mimic I/R injury. The results indicated that H2O2 decreased cardiomyocyte viability, induced apoptosis and decreased mitochondrial membrane potential (Figs. 1A-D and 2G-I). H2O2 treatment significantly increased mRNA expression of miR-124 in H9C2 cells (Fig. 1E). These data suggested that upregulation of miR-124 may be related to H2O2-induced cardiomyocyte apoptosis. The online databases TargetScan, miRanda and miRDB were used to predict the potential targets of miR-124. Computational prediction (Fig. 2A) showed miR-124 binding sites exist in the 3′ UTR of MCUR1. MCUR1 was identified as a potential target gene of miR-124 in cardiomyocyte apoptosis. A prior study demonstrated that MCUR1-mediated mitochondrial Ca2+ environment remodelling markedly promotes proliferation and apoptosis resistance of hepatoma cell lines (25). Therefore, expression of MCUR1 was investigated during cardiomyocyte apoptosis induced by H2O2. MCUR1 protein expression increased in a time-dependent manner following H2O2 treatment (Fig. 1F and G). These results indicated that the MCUR1 protein expression was increased during cardiomyocyte apoptosis.

miR-124 binds to MCUR1 3′-UTR and inhibits cardiomyocyte apoptosis by activating MCUR1

Compared with miR-NC, miR-124 overexpression increased the protein expression of MCUR1 compared with miR-NC (Fig. 2B-D). Treatment with H2O2 increased the protein expression of MCUR1 and miR-124 further increased this following treatment with H2O2 (Fig. 3C and D). Simultaneously, the addition of the anti-124 in presence of H2O2 significantly decreased expression of MCUR1 (Fig. 3A and B). MCUR1 siRNA treatment (100 nM) decreased the expression of MCUR1 (Fig. 2E and F). In the presence of H2O2, miR-124 inhibitor (anti-124), or MCUR1 siRNA inhibited MCUR1 protein expression. Simultaneous treatment with miR-124 inhibitor and MCUR1 siRNA further inhibited MCUR1 expression (Fig. 3E and F). Flow cytometry showed that H2O2 enhanced apoptosis, while miR-124 significantly decreased apoptosis induced by H2O2. However, when cells were co-treated with MCUR1-siRNA and miR-124, H2O2-induced apoptosis was restored (Fig. 2G and H). These results indicated that miR-124 inhibited cardiomyocyte apoptosis by activating MCUR1 following H2O2 treatment. Overexpression of miR-124 restored mitochondrial membrane potential in H2O2-treated cells (Fig. 2I). Moreover, when MCUR1-siRNA and miR-124 were applied simultaneously in H2O2-treated cells, mitochondrial membrane potential was significantly decreased (Fig. 2I and J). The dual-luciferase reporter assay confirmed binding of miR-124 to MCUR1 3′-UTR. The relative luciferase activity was increased in H9C2 cells co-transfected with MCUR1 3′UTR and miR-124 mimic compared to 3′UTR and miR-NC co-transfected (Fig. 3G). These findings indicated that miR-124 may activate expression of MCUR1 by binding to MCUR1 3′-UTR, decreasing cardiomyocyte apoptosis induced by H2O2.

miR-124 enters the nucleus and targets MCUR1 enhancer

Next, the association between miR-124 overexpression and increased MCUR1 expression was investigated. By screening miRNA database, the positions of numerous miRNAs including miR-24-1, lin-4 in the genome were coincident with enhancer regions (57). Most miRNAs are localized in the nucleus. These miRNAs bind to enhancers and activate gene expression at the genome level (5760). miR-24-1 activated gene transcription by targeting enhancers (45). It was reported that overexpression of miR-26a-1 increased the transcription of neighboring ITGA9 and VILL genes (57). TargetScan (61), miRanda (53) and miRDB online databases are computational approaches have been used to predict mRNA-miRNA interaction and microRNA targets (54). In this study, these online databases were used to predict MCUR1 enhancers. Comparison of the miR-124 sequences revealed binding of MCUR1 enhancer to miR-124 (Fig. 4A). FISH at the cellular level revealed the entry of miR-124 into the nucleus (Fig. 4B), which may have increased MCUR1 expression.

Discussion

The present study revealed the key role of miR-124 in both cardiomyocyte apoptosis and MI and its underlying mechanism. The findings further demonstrated that MCUR1 was a novel target of miR-124, and that the miR-124-MCUR1 axis modulated cardiomyocyte apoptosis induced by H2O2. The findings were consistent with the hypothesis that the expression level of miR-124 increases during oxidative stress and that miR-124 enters the nucleus to combine with the MCUR1 enhancer. The subsequent high expression level of MCUR1 confers resistance to apoptosis in cardiomyocytes (Fig. 4C). Thus, miR-124 may be a biomarker of myocardial injury and MI. Increasing the expression of MCUR1 may provide a new pathway to decrease effects of MI and subsequent dysfunction.

miR-124 has been reported to be involved in various cellular physiological and pathological processes (6264). Accumulating evidence has indicated that the expression of miR-124 facilitates cell death and differentiation under pathological stress (41,46). Increasing evidence has shown that miR-124 is associated with cardiovascular disease (39,65,66). miR-124 modulates cardiomyocyte differentiation into bone marrow-derived stem cells (65). miR-124 overexpression decreased oxidative stress in doxorubicin-induced cardiac injury and was a hopeful therapeutic target in doxorubicin-related cardiomyopathy (47). Previous studies have described the marked increase of miR-124 expression in smokers and patients with acute coronary syndrome, suggesting that miR-124 may be a biomarker of coronary heart disease (42,43). miR-124 has been proposed to be a cell cycle regulator that regulates cell survival and apoptosis (6769), consistent with the finding of the current study that miR-124 regulated cardiomyocyte apoptosis.

MCUR1 is a regulatory protein of the MCU that plays an important role in mitochondrial Ca2+[(Ca2+)m] uptake and maintenance of normal cellular bioenergy (18). MCUR1 knockout in HeLa cells causes a decrease in ATP synthesis, which is dependent on protein kinase activity and leads to autophagy (70). In addition, knockout of MCU and MCUR1 in vascular endothelial cells impaired [Ca2+]m uptake, thus weakened mitochondrial biosynthesis and cell migration, decreases cell proliferation and induces autophagy (71). Disorders in mitochondrial Ca2+ homeostasis are associated with occurrence and development of various types of tumour such as melanoma and MCUR1 plays an important role in mitochondrial Ca2+ homeostasis (72). The role of MCU in I/R has attracted increasing interest (73). In the present study, MCUR1 expression increased, leading to resistance to apoptosis induced by oxidative stress.

miRNAs negatively regulate gene expression primarily by targeting the 3′-UTR of mRNA transcripts in cytoplasm to achieve instability or translation inhibition (7476). Nuclear miRNAs exert gene activation functions. Human miR-373 was the first activator of gene transcription, which induced both E-cadherin (CDH1) and cold-shock domain-containing protein 2 (CSDC2) transcription (77). The analysis of 1302 breast cancer samples indicated that miRNAs and neighbouring genes may be positively associated (78). Thus, miRNAs have the dual functional ability to activate transcription in the nucleus and inhibit transcription in the cytoplasm. A number of studies have shown that microRNAs at enhancer sites exert transcriptional activation functions (48,79,80). miR-24-1 activates enhancer RNA (eRNA) expression and promotes the enrichment of p300 and RNA Pol II at enhancer sites (48). Nuclear miRNAs serve as enhancer triggers by modifying chromatin states that facilitate activation of transcriptional genes (81). miR-24-1 activates gene transcription by targeting enhancers (81). The present study identified a novel mechanism for the miR-124-MCUR1 axis during cardiac injury in cardiomyocytes. The present results revealed the mechanism of MCUR1 activation by miR-124 binding to MCUR1 enhancers and supported the hypothesis that miR-124 alters chromatin status and increases MCUR1 expression, leading to apoptosis resistance and inhibition of MI. However, further investigation is required to confirm these preliminary results. Further studies should investigate the interaction between miR-124 and MCUR1 and how miR-124 affects the chromosomal status of MUCR1 enhancers.

In summary, the present study demonstrated that high expression of miR-124 was induced under oxidative stress conditions. miR-124 enters the nucleus and combines with the enhancer of MCUR1 to activate expression of MCUR1. Mitochondrial Ca2+ environment remodelling significantly promotes resistance of cardiomyocytes to proliferation and apoptosis.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Project of Shandong Province Higher Educational Science and Technology Program (grant nos. J18KA250 and J18KA127), research project of Qingdao University Medical Group (grant no. YLJT20202039), Major Research Program of the National Natural Science Foundation of China (grant no. 91849209), National Natural Science Foundation of China (grant no. 81602353), Natural Science Foundation of Jiangsu Province (grant no. BK20171145), China Postdoctoral Science Foundation (grant nos. 2019M652314 and 2020T130333), Qingdao Applied Basic Research Project (grant no. 19-6-2-39-cg) and Major Science and Technology Project of Wenzhou Institute and University of Chinese Academy of Sciences (grant no. WIUCASQD2021028).

Availability of data and materials

The datasets used and/or analysed in the current study are available from the corresponding author upon reasonable request.

Authors' contributions

LG and HD conceptualized the study. LG and HD confirm the authenticity of all the raw data. CL and CJ designed the methodology. LHHA used software to process the data. LG, HD and YG performed experiments, wrote the manuscript and supervised the study. HD and YG collected data and reviewed and edited the manuscript. YD and CL analyzed and interpreted the data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

De Luca G, Suryapranata H, Ottervanger JP and Antman EM: Time delay to treatment and mortality in primary angioplasty for acute myocardial infarction: Every minute of delay counts. Circulation. 109:1223–1225. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Heusch G: Cardioprotection: Chances and challenges of its translation to the clinic. Lancet. 381:166–175. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Kumfu S, Chattipakorn S, Fucharoen S and Chattipakorn N: Mitochondrial calcium uniporter blocker prevents cardiac mitochondrial dysfunction induced by iron overload in thalassemic mice. Biometals. 25:1167–1175. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Sripetchwandee J, Sanit J, Chattipakorn N and Chattipakorn SC: Mitochondrial calcium uniporter blocker effectively prevents brain mitochondrial dysfunction caused by iron overload. Life Sci. 92:298–304. 2013. View Article : Google Scholar : PubMed/NCBI

5 

De Marchi E, Bonora M, Giorgi C and Pinton P: The mitochondrial permeability transition pore is a dispensable element for mitochondrial calcium efflux. Cell Calcium. 56:1–13. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Giorgi C, Bonora M, Sorrentino G, Missiroli S, Poletti F, Suski JM, Galindo Ramirez F, Rizzuto R, Di Virgilio F, Zito E, et al: p53 at the endoplasmic reticulum regulates apoptosis in a Ca2+-dependent manner. Proc Natl Acad Sci USA. 112:1779–1784. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Cortassa S, Aon MA, Marbán E, Winslow RL and O'Rourke B: An integrated model of cardiac mitochondrial energy metabolism and calcium dynamics. Biophys J. 84:2734–2755. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Vieira HL and Kroemer G: Pathophysiology of mitochondrial cell death control. Cell Mol Life Sci. 56:971–976. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Moreau B, Nelson C and Parekh AB: Biphasic regulation of mitochondrial Ca2+ uptake by cytosolic Ca2+ concentration. Curr Biol. 16:1672–1677. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Marchi S, Patergnani S, Missiroli S, Morciano G, Rimessi A, Wieckowski MR, Giorgi C and Pinton P: Mitochondrial and endoplasmic reticulum calcium homeostasis and cell death. Cell Calcium. 69:62–72. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Granatiero V, De Stefani D and Rizzuto R: Mitochondrial calcium handling in physiology and disease. Adv Exp Med Biol. 982:25–47. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Pan S, Ryu SY and Sheu SS: Distinctive characteristics and functions of multiple mitochondrial Ca2+ influx mechanisms. Sci China Life Sci. 54:763–769. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Drago I, Pizzo P and Pozzan T: After half a century mitochondrial calcium in- and efflux machineries reveal themselves. EMBO J. 30:4119–4125. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Kirichok Y, Krapivinsky G and Clapham DE: The mitochondrial calcium uniporter is a highly selective ion channel. Nature. 427:360–364. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Baughman JM, Perocchi F, Girgis HS, Plovanich M, Belcher-Timme CA, Sancak Y, Bao XR, Strittmatter L, Goldberger O, Bogorad RL, et al: Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter. Nature. 476:341–345. 2011. View Article : Google Scholar : PubMed/NCBI

16 

De Stefani D, Raffaello A, Teardo E, Szabò I and Rizzuto R: A forty-kilodalton protein of the inner membrane is the mitochondrial calcium uniporter. Nature. 476:336–340. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Docampo R and Lukeš J: Trypanosomes and the solution to a 50-year mitochondrial calcium mystery. Trends Parasitol. 28:31–37. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Mallilankaraman K, Doonan P, Cárdenas C, Chandramoorthy HC, Müller M, Miller R, Hoffman NE, Gandhirajan RK, Molgó J, Birnbaum MJ, et al: MICU1 is an essential gatekeeper for MCU-mediated mitochondrial Ca(2+) uptake that regulates cell survival. Cell. 151:630–644. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Mallilankaraman K, Cardenas C, Doonan P, Chandramoorthy H, Irrinki K, Golenar T, Csordas G, Madireddi P, Yang J, Miller R, et al: MCUR1 is an essential component of mitochondrial Ca2+ uptake that regulates cellular metabolism. Biophys J. 104:616a2013. View Article : Google Scholar

20 

Plovanich M, Bogorad RL, Sancak Y, Kamer KJ, Strittmatter L, Li AA, Girgis HS, Kuchimanchi S, De Groot J, Speciner L, et al: MICU2, a paralog of MICU1, resides within the mitochondrial uniporter complex to regulate calcium handling. PLoS One. 8:e557852013. View Article : Google Scholar : PubMed/NCBI

21 

Calderon MR, Verway M, Benslama RO, Birlea M, Bouttier M, Dimitrov V, Mader S and White JH: Ligand-dependent corepressor contributes to transcriptional repression by C2H2 zinc-finger transcription factor ZBRK1 through association with KRAB-associated protein-1. Nucleic Acids Res. 42:7012–7027. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Kastenhuber ER and Lowe SW: Putting p53 in context. Cell. 170:1062–1078. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Tian C, Xing G, Xie P, Lu K, Nie J, Wang J, Li L, Gao M, Zhang L and He F: KRAB-type zinc-finger protein Apak specifically regulates p53-dependent apoptosis. Nat Cell Biol. 11:580–591. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Yuan L, Tian C, Wang H, Song S, Li D, Xing G, Yin Y, He F and Zhang L: Apak competes with p53 for direct binding to intron 1 of p53AIP1 to regulate apoptosis. EMBO Rep. 13:363–370. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Ren T, Wang J, Zhang H, Yuan P, Zhu J, Wu Y, Huang Q, Guo X, Zhang J, Ji L, et al: MCUR1-mediated mitochondrial calcium signaling facilitates cell survival of hepatocellular carcinoma via reactive oxygen species-dependent P53 degradation. Antioxid Redox Signal. 28:1120–1136. 2018. View Article : Google Scholar : PubMed/NCBI

26 

García-Rivas Gde J, Carvajal K, Correa F and Zazueta C: Ru360, a specific mitochondrial calcium uptake inhibitor, improves cardiac post-ischaemic functional recovery in rats in vivo. Br J Pharmacol. 149:829–837. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Joiner MLA, Koval OM, Li J, He BJ, Allamargot C, Gao Z, Luczak ED, Hall DD, Fink BD, Chen B, et al: CaMKII determines mitochondrial stress responses in heart. Nature. 491:269–273. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Liang N, Wang P, Wang S, Li S, Li Y, Wang J and Wang M: Role of mitochondrial calcium uniporter in regulating mitochondrial fission in the cerebral cortexes of living rats. J Neural Transm (Vienna). 121:593–600. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Zhang L, Gao X, Yuan X, Dong H, Zhang Z and Wang S: Mitochondrial calcium uniporter opener spermine attenuates the cerebral protection of diazoxide through apoptosis in rats. J Stroke Cerebrovasc Dis. 23:829–835. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Yan H, Zhang D, Hao S, Li K and Hang CH: Role of mitochondrial calcium uniporter in early brain injury after experimental subarachnoid hemorrhage. Mol Neurobiol. 52:1637–1647. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Liao Y, Hao Y, Chen H, He Q, Yuan Z and Cheng J: Mitochondrial calcium uniporter protein MCU is involved in oxidative stress-induced cell death. Protein Cell. 6:434–442. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Santulli G, Xie W, Reiken SR and Marks AR: Mitochondrial calcium overload is a key determinant in heart failure. Proc Natl Acad Sci USA. 112:11389–11394. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Bell JR, Erickson JR and Delbridge LM: Ca(2+)/calmodulin dependent kinase II: A critical mediator in determining reperfusion outcomes in the heart? Clin Exp Pharmacol Physiol. 41:940–946. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Icli B, Wara AKM, Moslehi J, Sun X, Plovie E, Cahill M, Marchini JF, Schissler A, Padera RF, Shi J, et al: MicroRNA-26a regulates pathological and physiological angiogenesis by targeting BMP/SMAD1 signaling. Circ Res. 113:1231–1241. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Hu S, Huang M, Li Z, Jia F, Ghosh Z, Lijkwan MA, Fasanaro P, Sun N, Wang X, Martelli F, et al: MicroRNA-210 as a novel therapy for treatment of ischemic heart disease. Circulation. 122 (11 Suppl):S124–S131. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Zhang X, Yoon JY, Morley M, McLendon JM, Mapuskar KA, Gutmann R, Mehdi H, Bloom HL, Dudley SC, Ellinor PT, et al: A common variant alters SCN5A-miR-24 interaction and associates with heart failure mortality. J Clin Invest. 128:1154–1163. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Demkes CJ and van Rooij E: MicroRNA-146a as a regulator of cardiac energy metabolism. Circulation. 136:762–764. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Sun Y, Luo ZM, Guo XM, Su DF and Liu X: An updated role of microRNA-124 in central nervous system disorders: A review. Front Cell Neurosci. 9:1932015. View Article : Google Scholar : PubMed/NCBI

39 

Bao Q, Chen L, Li J, Zhao M, Wu S, Wu W and Liu X: Role of microRNA-124 in cardiomyocyte hypertrophy induced by angiotensin II. Cell Mol Biol (Noisy-le-grand). 63:23–27. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Zhang L, Chen Q, An W, Yang F, Maguire EM, Chen D, Zhang C, Wen G, Yang M, Dai B, et al: Novel pathological role of hnRNPA1 (heterogeneous nuclear Ribonucleoprotein A1) in vascular smooth muscle cell function and Neointima hyperplasia. Arterioscler Thromb Vasc Biol. 37:2182–2194. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Han F, Chen Q, Su J, Zheng A, Chen K, Sun S, Wu H, Jiang L, Xu X, Yang M, et al: MicroRNA-124 regulates cardiomyocyte apoptosis and myocardial infarction through targeting Dhcr24. J Mol Cell Cardiol. 132:178–188. 2019. View Article : Google Scholar : PubMed/NCBI

42 

de Ronde MWJ, Kok MGM, Moerland PD, Van den Bossche J, Neele AE, Halliani A, van der Made I, de Winther MPJ, Meijers JCM, Creemers EE and Pinto-Sietsma SJ: High miR-124-3p expression identifies smoking individuals susceptible to atherosclerosis. Atherosclerosis. 263:377–384. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Devaux Y, Dankiewicz J, Salgado-Somoza A, Stammet P, Collignon O, Gilje P, Gidlöf O, Zhang L, Vausort M, Hassager C, et al: Association of circulating MicroRNA-124-3p levels with outcomes after out-of-hospital cardiac arrest: A substudy of a randomized clinical trial. JAMA Cardiol. 1:305–313. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Gilje P, Gidlöf O, Rundgren M, Cronberg T, Al-Mashat M, Olde B, Friberg H and Erlinge D: The brain-enriched microRNA miR-124 in plasma predicts neurological outcome after cardiac arrest. Crit Care. 18:R402014. View Article : Google Scholar : PubMed/NCBI

45 

Gacoń J, Kabłak-Ziembicka A, Stępień E, Enguita FJ, Karch I, Derlaga B, Żmudka K and Przewłocki T: Decision-making microRNAs (miR-124, −133a/b, −34a and −134) in patients with occluded target vessel in acute coronary syndrome. Kardiol Pol. 74:280–288. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Ghafouri-Fard S, Shoorei H, Bahroudi Z, Abak A, Majidpoor J and Taheri M: An update on the role of miR-124 in the pathogenesis of human disorders. Biomed Pharmacother. 135:1111982021. View Article : Google Scholar : PubMed/NCBI

47 

Liu Y, Li Y, Ni J, Shu Y, Wang H and Hu T: MiR-124 attenuates doxorubicin-induced cardiac injury via inhibiting p66Shc-mediated oxidative stress. Biochem Biophys Res Commun. 521:420–426. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Zhu P, Li H, Zhang A, Li Z, Zhang Y, Ren M, Zhang Y and Hou Y: MicroRNAs sequencing of plasma exosomes derived from patients with atrial fibrillation: miR-124-3p promotes cardiac fibroblast activation and proliferation by regulating AXIN1. J Physiol Biochem. 78:85–98. 2022. View Article : Google Scholar : PubMed/NCBI

49 

Hescheler J, Meyer R, Plant S, Krautwurst D, Rosenthal W and Schultz G: Morphological, biochemical, and electrophysiological characterization of a clonal cell (H9c2) line from rat heart. Circ Res. 69:1476–1486. 1991. View Article : Google Scholar : PubMed/NCBI

50 

Shimada Y, Fischman DA and Moscona AA: The fine structure of embryonic chick skeletal muscle cells differentiated in vitro. J Cell Biol. 35:445–453. 1967. View Article : Google Scholar : PubMed/NCBI

51 

Forero DA, González-Giraldo Y, Castro-Vega LJ and Barreto GE: qPCR-based methods for expression analysis of miRNAs. Biotechniques. 67:192–199. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Shin S, Jung Y, Uhm H, Song M, Son S, Goo J, Jeong C, Song JJ, Kim VN and Hohng S: Quantification of purified endogenous miRNAs with high sensitivity and specificity. Nat Commun. 11:60332020. View Article : Google Scholar : PubMed/NCBI

53 

John B, Enright AJ, Aravin A, Tuschl T, Sander C and Marks DS: Human MicroRNA targets. PLoS Biol. 2:e3632004. View Article : Google Scholar : PubMed/NCBI

54 

Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP and Burge CB: Prediction of mammalian microRNA targets. Cell. 115:787–798. 2003. View Article : Google Scholar : PubMed/NCBI

55 

Lee Y and Gustafsson AB: Role of apoptosis in cardiovascular disease. Apoptosis. 14:536–548. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Bialik S, Geenen DL, Sasson IE, Cheng R, Horner JW, Evans SM, Lord EM, Koch CJ and Kitsis RN: Myocyte apoptosis during acute myocardial infarction in the mouse localizes to hypoxic regions but occurs independently of p53. J Clin Invest. 100:1363–1372. 1997. View Article : Google Scholar : PubMed/NCBI

57 

Xiao M, Li J, Li W, Wang Y, Wu F, Xi Y, Zhang L, Ding C, Luo H, Li Y, et al: MicroRNAs activate gene transcription epigenetically as an enhancer trigger. RNA Biol. 14:1326–1334. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Lu L, Zhou L, Chen EZ, Sun K, Jiang P, Wang L, Su X, Sun H and Wang H: A novel YY1-miR-1 regulatory circuit in skeletal myogenesis revealed by genome-wide prediction of YY1-miRNA network. PLoS One. 7:e275962012. View Article : Google Scholar : PubMed/NCBI

59 

Lee BK, Bhinge AA and Iyer VR: Wide-ranging functions of E2F4 in transcriptional activation and repression revealed by genome-wide analysis. Nucleic Acids Res. 39:3558–3573. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Guimbellot JS, Erickson SW, Mehta T, Wen H, Page GP, Sorscher EJ and Hong JS: Correlation of microRNA levels during hypoxia with predicted target mRNAs through genome-wide microarray analysis. BMC Med Genomics. 2:152009. View Article : Google Scholar : PubMed/NCBI

61 

Lewis BP, Burge CB and Bartel DP: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 120:15–20. 2005. View Article : Google Scholar : PubMed/NCBI

62 

Xu J, Zheng Y, Wang L, Liu Y, Wang X, Li Y and Chi G: miR-124: A promising therapeutic target for central nervous system injuries and diseases. Cell Mol Neurobiol. 42:2031–2053. 2022. View Article : Google Scholar : PubMed/NCBI

63 

Qin Z, Wang PY, Su DF and Liu X: miRNA-124 in immune system and immune disorders. Front Immunol. 7:4062016. View Article : Google Scholar : PubMed/NCBI

64 

Yang J, Zhang X, Chen X, Wang L and Yang G: Exosome mediated delivery of miR-124 promotes neurogenesis after ischemia. Mol Ther Nucleic Acids. 7:278–287. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Cai B, Li J, Wang J, Luo X, Ai J, Liu Y, Wang N, Liang H, Zhang M, Chen N, et al: microRNA-124 regulates cardiomyocyte differentiation of bone marrow-derived mesenchymal stem cells via targeting STAT3 signaling. Stem Cells. 30:1746–1755. 2012. View Article : Google Scholar : PubMed/NCBI

66 

Devaux Y and Stammet P: Cardiolinc network: What's new in prognostication after cardiac arrest: microRNAs? Intensive Care Med. 44:897–899. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Das E, Jana NR and Bhattacharyya NP: MicroRNA-124 targets CCNA2 and regulates cell cycle in STHdh(Q111)/Hdh(Q111) cells. Biochem Biophys Res Commun. 437:217–224. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Taniguchi K, Sugito N, Kumazaki M, Shinohara H, Yamada N, Nakagawa Y, Ito Y, Otsuki Y, Uno B, Uchiyama K and Akao Y: MicroRNA-124 inhibits cancer cell growth through PTB1/PKM1/PKM2 feedback cascade in colorectal cancer. Cancer Lett. 363:17–27. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Liang YN, Tang YL, Ke ZY, Chen YQ, Luo XQ, Zhang H and Huang LB: MiR-124 contributes to glucocorticoid resistance in acute lymphoblastic leukemia by promoting proliferation, inhibiting apoptosis and targeting the glucocorticoid receptor. J Steroid Biochem Mol Biol. 172:62–68. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Mallilankaraman K, Cárdenas C, Doonan PJ, Chandramoorthy HC, Irrinki KM, Golenár T, Csordás G, Madireddi P, Yang J, Müller M, et al: MCUR1 is an essential component of mitochondrial Ca2+ uptake that regulates cellular metabolism. Nat Cell Biol. 14:1336–1343. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Tomar D, Dong Z, Shanmughapriya S, Koch DA, Thomas T, Hoffman NE, Timbalia SA, Goldman SJ, Breves SL, Corbally DP, et al: MCUR1 is a scaffold factor for the MCU complex function and promotes mitochondrial bioenergetics. Cell Rep. 15:1673–1685. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Romero-Garcia S and Prado-Garcia H: Mitochondrial calcium: Transport and modulation of cellular processes in homeostasis and cancer (review). Int J Oncol. 54:1155–1167. 2019.PubMed/NCBI

73 

Kwong JQ: The mitochondrial calcium uniporter in the heart: Energetics and beyond. J Physiol. 595:3743–3751. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Fabian MR, Sonenberg N and Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 79:351–379. 2010. View Article : Google Scholar : PubMed/NCBI

75 

Pasquinelli AE: MicroRNAs and their targets: Recognition, regulation and an emerging reciprocal relationship. Nat Rev Genet. 13:271–282. 2012. View Article : Google Scholar : PubMed/NCBI

76 

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

77 

Place RF, Li LC, Pookot D, Noonan EJ and Dahiya R: MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci USA. 105:1608–1613. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Dvinge H, Git A, Gräf S, Salmon-Divon M, Curtis C, Sottoriva A, Zhao Y, Hirst M, Armisen J, Miska EA, et al: The shaping and functional consequences of the microRNA landscape in breast cancer. Nature. 497:378–382. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Zou Q, Liang Y, Luo H and Yu W: miRNA-mediated RNAa by targeting enhancers. Adv Exp Med Biol. 983:113–125. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Liu H, Lei C, He Q, Pan Z, Xiao D and Tao Y: Nuclear functions of mammalian MicroRNAs in gene regulation, immunity and cancer. Mol Cancer. 17:642018. View Article : Google Scholar : PubMed/NCBI

81 

Vaschetto LM: miRNA activation is an endogenous gene expression pathway. RNA Biol. 15:826–828. 2018.PubMed/NCBI

Related Articles

Journal Cover

August-2023
Volume 28 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
Guo L, Liu C, Jiang C, Dong Y, Htet Htet Aung L, Ding H and Gao Y: miR‑124 inhibits cardiomyocyte apoptosis in myocardial ischaemia‑reperfusion injury by activating mitochondrial calcium uniporter regulator 1. Mol Med Rep 28: 144, 2023.
APA
Guo, L., Liu, C., Jiang, C., Dong, Y., Htet Htet Aung, L., Ding, H., & Gao, Y. (2023). miR‑124 inhibits cardiomyocyte apoptosis in myocardial ischaemia‑reperfusion injury by activating mitochondrial calcium uniporter regulator 1. Molecular Medicine Reports, 28, 144. https://doi.org/10.3892/mmr.2023.13031
MLA
Guo, L., Liu, C., Jiang, C., Dong, Y., Htet Htet Aung, L., Ding, H., Gao, Y."miR‑124 inhibits cardiomyocyte apoptosis in myocardial ischaemia‑reperfusion injury by activating mitochondrial calcium uniporter regulator 1". Molecular Medicine Reports 28.2 (2023): 144.
Chicago
Guo, L., Liu, C., Jiang, C., Dong, Y., Htet Htet Aung, L., Ding, H., Gao, Y."miR‑124 inhibits cardiomyocyte apoptosis in myocardial ischaemia‑reperfusion injury by activating mitochondrial calcium uniporter regulator 1". Molecular Medicine Reports 28, no. 2 (2023): 144. https://doi.org/10.3892/mmr.2023.13031