Effects of microRNA‑338‑3p on morphine‑induced apoptosis and its underlying mechanisms

  • Authors:
    • Hong‑Liang Weng
    • Ming‑Jing Wang
  • View Affiliations

  • Published online on: July 12, 2016     https://doi.org/10.3892/mmr.2016.5506
  • Pages: 2085-2092
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate the effects of microRNA-338-3p (miR-338-3p) on morphine (MP)-induced apoptosis, and its underlying mechanisms. Freshly‑isolated mouse peritoneal macrophages were cultured in vitro and treated with MP following transfection with miR‑338‑3p mimic, inhibitor or controls. miR‑338‑3p expression levels increased significantly following MP treatment (P<0.01). This increase was enhanced following transfection with miR‑338‑3p mimic (P<0.05) and abrogated following transfection with miR‑338‑3p inhibitor (P<0.05). The apoptotic rate increased significantly in groups treated with MP (P<0.05); however, this increase was abrogated by transfection with miR‑338‑3p inhibitor (P<0.05). Bioinformatics software predicted that sex determining region Y‑box 4 (SOX4) was the target gene of miR‑338‑3p and this was verified using a dual‑luciferase reporter gene system. SOX4 mRNA and protein expression levels decreased significantly following MP treatment (P<0.05); however, this decrease was abrogated following transfection with miR‑338‑3p inhibitor (P<0.05). Caspase‑3 protein expression levels increased markedly following MP treatment (P<0.05); however, this increase was inhibited by transfection with miR‑338‑3p inhibitor (P<0.05). Therefore, decreased expression of miR‑338‑3p may suppress MP‑induced apoptosis, potentially via the upregulation of SOX4 expression and the caspase‑3‑dependent apoptotic signaling pathway.

Introduction

MicroRNA (miR) is a short-segment (length, 20–23 nt) RNA, which binds to the 3′-untranslated region (UTR) of its target DNA by reverse complement, thus preventing the translation of mRNA or causing its degradation (13). Extensively expressed in mammals, miR regulates various physiological processes, cell functions and signaling pathways via the regulation of gene expression (15). Thus far, >2,000 miRs associated with humans have been identified, many of which perform various gene regulatory functions (6,7). However, the precise functions and underlying regulatory mechanisms of miR remain to be fully elucidated. A number of studies have suggested that miR is associated with certain diseases (810). Therefore, investigating the function of a specific miR and its underlying mechanisms may improve our understanding of the regulation of signaling pathways by miR and the subsequent effects, including cell differentiation, proliferation and apoptosis. This may contribute to improvement of human health and disease treatment.

Morphine (MP) is commonly used during the treatment of cancer. In addition to easing pain, it has been demonstrated to increase the susceptibility of cancer cells to apoptosis by promoting the expression of apoptosis-associated proteins, which suppresses the proliferation of cancer cells (1113). However, MP treatment for cancer is complex and is often associated with other factors, including miR (14,15). In addition, as an opioid, MP is addictive and is associated with various side effects (16,17), which may alter miR expression (15,18,19). miR-338-3p is a widely-reported miR that promotes cancer cell death by regulating specific signaling pathways or associated genes during cancer treatment (20,21). Investigating the association between MP and miR-338-3p may contribute to our understanding of the activity of miR-338-3p and the underlying molecular mechanisms of the effects of MP in cancer treatment. In the present study, mouse macrophages were transfected with miR-338-3p and treated with MP to observe the effect on miR-338-3p expression. Furthermore, the interaction between MP and miR-338-3p was investigated and the target gene of miR-338-3p was verified.

Materials and methods

Isolation of mouse peritoneal macrophages

A total of 5 C57BL/6J mice (regardless of gender; weight, 20±2 g), purchased from the laboratory animal center of Xinjiang Medical University (Ürümqi, China), were housed at 21°C in 50% humidity, with a 12 h light/dark cycle, and free access to food and water. A week later, the mice (6–8 weeks old) were sacrificed by cervical dislocation; the whole mouse was soaked in 75% ethanol for 5 min and dried with sterile gauze. A total of 5 ml Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) was injected i.p. A total of 5 min later, 3.5–4 ml liquid was withdrawn from the abdominal cavity via syringe and centrifuged at 134 × g for 10 min at 4°C. The supernatant was discarded and the cell pellet resuspended in Hank's balanced salt solution (Beijing Huamaike Biotechnology Co., Ltd, Beijing, China). DMEM containing 10% fetal bovine serum (Biowest, Nuaillé, France) and double-antibody [penicillin (100 U/ml) and streptomycin (100 mg/ml)] was added to achieve the correct cell concentration (1×109/l), and the cells were cultured in a 25T-flask under 5% CO2 for 48 h at 37°C. At this stage, certain adherent cells exhibited round or oval morphology when observed under a Zeiss Axio Observer A1 microscope (Carl Zeiss MicroImaging, Inc., Thornwood, NY, USA). DMEM was then exchanged for RPMI-1640 medium (HyClone; GE Healthcare Life Sciences, Logan, UT, USA) containing 10% fetal calf serum (Hyclone; GE Healthcare Life Sciences) and the macrophages were cultured under 5% CO2, 37°C and saturated humidity. Culture medium was replaced every day; digestion was performed every 4 days with 0.25% trypsin; when cell confluence reached 90%, the cells were passaged at a ratio of 1:3. The present study was approved by the Experimental Animals Ethics Committee of Linyi Yishui Central Hospital (Linyi, China). All studies performed on the mice were strictly in accordance with the Provisions of Protection and Use of Experimental Animals from the International Association for the Study of Pain (22).

Transfection and grouping of mouse macrophages

When cell confluence reached 80–90%, macrophages were transferred to a 24-well plate (3×104/well). The cultured cells were divided into 6 groups: i) Blank control group; ii) MP group, in which the macrophages were treated with 10 μM MP (Sigma-Aldrich, St. Louis, MO, USA) for 24 h; iii) MP+miR-338-3p mimic group, in which the macrophages were treated with 10 μM MP for 24 h following transfection with a miR-338-3p mimic for 24 h; iv) MP+control mimic group, in which the macrophages were treated with 10 μM MP for 24 h following transfection with a negative control mimic for 24 h; v) MP+miR-338-3p inhibitor group, in which the macrophages were treated with 10 μM MP following transfection with a miR-338-3p inhibitor for 24 h; and vi) MP+control inhibitor group, in which the macrophages were treated with 10 μM MP following transfection with a negative control inhibitor for 24 h. miR-338-3p mimic, mimic control, miR-338-3p inhibitor and inhibitor control were purchased from Shanghai GenePharma Co., Ltd., Shanghai, China, and transfection sequences are listed in Table I. Transfection was performed using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions, immediately when the confluence of the cells in each group reached 50–60% in each well. Each treatment group was performed in triplicate and the experiment was repeated at least three times with similar results.

Table I

Transfection sequences of miR-338-3p mimic, inhibitor and controls.

Table I

Transfection sequences of miR-338-3p mimic, inhibitor and controls.

GroupSequence
miR-338-3p mimicF, 5′-UCCAGCAUCAGUGAUUUUGUUG-3′
R, 3′-UUAGGUCGUAGUCACUAAAACA-5′
Mimic controlF, 5′-UUCUCCGAACGUGUCACGUTT-3′
R, 3′-TTAAGAGGCUUGCACAGUGCA-5′
miR-338-3p inhibitorF, 5′-CAACAAAAUCACUGAUGCUGGA-3′
Inhibitor controlR, 5′-UUGUAAGUUGCGACAGCCACUCA-3′

[i] miR, microRNA; F, forward; R, reverse.

miR-338-3p and sex determining region Y-box 4 (SOX4) mRNA expression levels detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was used to extract the total RNA and diethylpyrocarbonate (DEPC; 50 μl; Sigma-Aldrich) was used to inactivate RNases. RNA concentration was determined on a DU-800 Spectrophotometer (Beckman Coulter, Inc., Brea, CA, USA). RNA purity and integrity were controlled in accordance with the width ratio of 28S and 18S RNA bands following formaldehyde modified gel electrophoresis (5 V for 1 h). The width ratio of 28S and 18S ≥1.5 and A260 nm/A280 nm ≥1.8 indicated that RNA had a good completeness and no degradation. The extracted total RNA showed clear and bright 28S and 18S bands; no degradation was observed. The A260 nm/A280 nm ratio was 2.0, indicating that the extracted total RNA was of high quality. miR-338-3p RT-qPCR was performed with the All-in-One® First-Strand cDNA Synthesis kit and the SYBR Green I based All-in-One® qPCR Mix (GeneCopoeia, Inc., Rockville, MD, USA) according to the manufacturer's instructions. The reverse transcription reaction contained total RNA with a final concentration of 2 μg (2 μl), 2.5 U/μl PolyA polymerase (1 μl), reverse transcriptase RTase Mix (1 μl), 5X reaction buffer (5 μl) and DEPC H2O (16 μl), in a total volume of 25 μl. The reverse transcription reaction conditions were: 37°C for 60 min and 85°C for 5 min. The qPCR reaction system contained cDNA (2 μl), 2X All-in-ONE Q-PCR Mix (10 μl), universal PCR primer (2 μl), 50X Rox Reference Dye (0.4 μl), miR-338-3p or U6 primer (2 μl) and DEPC H2O (3.6 μl). qPCR cycling conditions were as follows: Preheating for 10 min at 95°C, degeneration for 10 sec at 95°C, annealing for 20 sec at 60°C and extension for 34 sec at 72°C. A total of 40 cycles were performed. SOX4 RT-qPCR was performed with Prime Script™ RT reagent kit (Takara Bio, Inc.) and SYBR® Green Real time PCR Master Mix (Toyobo Co., Ltd., Osaka, Japan) according to the manufacturer's instructions. The reverse transcription reaction contained 5X Moloney-murine leukemia virus (M-MLV) buffer (2 μl), deoxynucleotides (2 mM; 2 μl), Random 6 mers (0.5 μl), RTase M-MLV (200 U/μl; 0.25 μl) and RNA (1 μl), and DEPC H2O was added to make up a final volume of 10 μl. Reaction conditions were as follows: 42°C for l0 min, 95°C for 2 min and 4°C for 10 min. The qPCR reaction system contained 2x SYBR Green (10 μl), forward and reverse primers (0.8 μl), cDNA template (1 μl) and ddH2O was added to make up a total volume of 20 μl. U6 and GAPDH served as internal reference genes. All primer sequences are listed in Table II. The relative quantitative expression of the target gene was calculated by the 2−ΔΔCq method (23). The formula was expressed as follows: ΔΔCq= (Cqexperimental target gene − Cqexperimental reference gene) − (Cqcontrol target gene − Cqcontrol reference gene).

Table II

Primer sequences.

Table II

Primer sequences.

GeneSequence
miR-338-3pF, 5′-TGCGGTCCAGCATCAGTGAT-3′
R, 5′-CCAGTGCAGGGTCCGAGGT-3′
U6F, 5′-TGCGGGTGCTCGCTTCGGCAGC-3′
R, 5′-CCAGTGCAGGGTCCGAGGT-3′
SOX4F, 5′-GTGAGCGAGATGATCTCGGG-3′
R, 5′-CAGGTTGGAGATGCTGGACTC-3′
GAPDHF, 5′-AGAAGGCTGGGGCTCATTTG-3′
R, 5′-AGGGGCCATCCACAGTCTTC-3′

[i] miR, microRNA; SOX4, sex determining region Y-box 4; F, forward; R, reverse.

Dual-luciferase reporter gene detection

The target gene of mus musculus (mmu)-miR-338-3p was predicted by TargetScan (version 7.1; targetscan.org), PicTar (pictar.mdc-berlin.de/) and microRNA.org (August 2010 release; microrna.org/microrna/home.do) online bioinformatics prediction software. The 3′-UTR of the SOX4 gene contained sequences that regulated various sites; amplification was performed according to the manufacturer's instructions of the SYBR® Green Real Time PCR Master Mix (Toyobo Co., Ltd., Japan). The primer sequences were as follows: Forward, GAGCTCCTCCGCCTTCTTTTCTAC and reverse, CTCGAGCACGTCTTCTCATTTACACC. The reaction conditions were 42°C for 10 min, 95°C for 2 min and 4°C for 10 min. The reaction system contained 2x SYBR Green (10 μl), primers (0.8 μl), cDNA (1.0 μl) and ddH2O was added to make up a total volume of 20 μl. The PCR amplified SOX4 3′UTR was cloned into the pmirGLO luciferase miR target expression vector (Promega Corporation, Madison, WI, USA). Site-directed mutagenesis of the miR-338-3p binding site in the 3′-UTR of SOX4 were predicted using the previously mentioned bioinformatics software. The primer sequences were as follows: Forward, TGG ACG ACT TTA AAA AAA CAA TTC AG and reverse, CAG ATT TGA GTT GCG TTT GAA TC. The primers were synthetized by Sangon Biotech Co., Lyd., Shanghai, China). The pRL-TK vector (Takara Bio, Inc.) expressing Renilla luciferase served as an internal reference for transfection efficiency. miR-338-3p mimic or miR-338-3p mimic control was co-transfected with the luciferase vector containing the wild-type or mutated SOX4 3′-UTR, and the negative control was added directly to macrophages. Dual-luciferase reporter gene detection was performed according to the manufacturer's instructions (24).

Apoptosis detection by annexin V/propidium iodide (PI) double-staining

The apoptotic rate was determined by Annexin V/PI double-staining (Nanjing KeyGen Biotech Co., Ltd., Nanjing, China). Macrophages were digested with pancreatin and 1–5×105 macrophages were transferred to an Eppendorf tube and washed twice with 1 ml phosphate-buffered saline, followed by centrifugation at 134 × g for 10 min at 4°C. Cells were resuspended in binding buffer (500 μl; Promega Corporation). A total of 5 μl Annexin V-fluorescein isothiocyanate (FITC) was added to the solution, followed by 5 μl PI. Cells were incubated for 5–15 min in the dark at room temperature; assessment by flow cytometry was performed within 1 h of the conclusion of the incubation. The green fluorescence of Annexin V-FITC was examined by the FL1 channel and the red fluorescence of PI was examined by the FL2 channel. The excitation wavelength was 488 nm, and FITC and PI fluorescence were examined through filters with wavelengths of 515 and >560 nm, respectively. The lower left quadrant (Q4) of the scatter diagram exhibited healthy living cells (FITC/PI); the lower right quadrant (Q3) contained early stage apoptotic cells (FITC+/PI); and the upper right quadrant (Q2) contained necrotic and late stage apoptotic cells (FITC+/PI+). Apoptotic rate=early stage (Q3) percentage+late stage (Q2) percentage.

SOX4 and caspase-3 protein expression levels detected by western blotting

Following lysis of macrophages with protein lysis buffer [50 mM Tris/HCl, 150 mM NaCl, 50 mM NaF, 1 mM Na4P2O7·10 H2O, 0.1% deoxycorticosterone, 1.0% Nonidet P40, 50 μl of Na3VO4 and Halt Protease Inhibitor Cocktail (Pierce; Thermo Fisher Scientific, Inc.)], protein concentration was measured by the bicinchoninic method (25). Protein (50 μg) was loaded onto a 10% SDS-PAGE gel for electrophoresis at 200 V for 1 h. Protein was transferred onto polyvinylidene difluoride membranes. Membranes were blocked with 5% skim milk and probed with primary rabbit anti-SOX4 (1:500; ab80261; Abcam, Cambridge, MA, USA), anti-caspase-3 (1:500; ab4051; Abcam) and anti-β-actin (1:1,000; ab8227, Abcam) overnight on a shaker at 4°C. The membranes were then washed in Tris-buffered saline three times. The membranes were subsequently incubated with the secondary goat anti-rabbit horseradish peroxidase-conjugated antibody (1:2,000; ab6721; Abcam) at room temperature for 1 h and visualized with Enhanced Chemiluminescence Plus reagent (GE Healthcare Life Sciences, Chalfont, UK). The protein bands were scanned by a GeneGenius automated gel imaging system (Syngene, Frederick, MD, USA) and analyzed with GeneTools software (version 4.0; Syngene).

Statistical analysis

The data was analyzed with SPSS software version 19.0 (IBM SPSS, Armonk, NY, USA), using the mean ± standard deviation to denote the measurement data, on which normality tests were conducted. The comparison between two groups or among multiple groups was performed using independent sample t-test or one-way analysis of variance (uniformity test of error variance was performed prior to analysis), respectively. The comparison between any two means was conducted using the least significant difference t-test. P<0.05 was considered to indicated a statistically significant difference.

Results

Variations in miR-338-3p expression levels in MP-treated macrophages

Following treatment with MP, miR-338-3p expression levels in the MP group (P<0.001), MP+mimic control group (P<0.001), MP+inhibitor control group (P<0.001), MP+miR-338-3p mimic group (P<0.001) and MP+miR-338-3p inhibitor group (P=0.006) increased significantly compared with the blank (untreated) group (Table III; Fig. 1). No significant differences were observed in miR-338-3p expression levels between the MP group and the MP+mimic control and MP+inhibitor control groups (P=0.521 and 0.936, respectively). In addition, no significant differences were observed in miR-338-3p expression levels between the MP+mimic control group and MP+inhibitor control group (P=0.574). Compared with the MP+mimic control group, the miR-338-3p expression level in the MP+miR-338-3p mimic group increased significantly (P<0.001); compared with the MP+inhibitor control group, the miR-338-3p expression level in the MP+miR-338-3p inhibitor group decreased significantly (P=0.003).

Table III

Relative miR-338-3p expression levels detected by reverse transcription-quantitative polymerase chain reaction.

Table III

Relative miR-338-3p expression levels detected by reverse transcription-quantitative polymerase chain reaction.

GroupRelative expression of miR-338-3p
Blank 1.00±0.00
MP 1.55±0.07a
MP+mimic control 1.60±0.08a
MP+inhibitor control 1.56±0.10a
MP+miR-338-3p mimic 1.98±0.16ac
MP+miR-338-3p inhibitor 1.26±0.10a,b,d,e

a P<0.05 vs. blank;

b P<0.05 vs. MP;

c P<0.05 vs. MP+mimic control;

d P<0.05 vs. MP+inhibitor control;

e P<0.05 vs. MP+miR-338-3p mimic. Analysis of variance results: F-value, 35.03; P-value, <0.001. MP, morphine; miR, microRNA.

Apoptotic rate in MP-treated macrophages

The apoptotic rate in the blank group was 7.70±1.83%; however, the rate the MP group (P<0.001), MP+mimic control group (P<0.001), MP+inhibitor control group (P<0.001), MP+miR-338-3p mimics group (P<0.001) and MP+miR-338-3p inhibitors group (P=0.013) increased significantly (Fig. 2). The apoptosis rates of the MP+mimic control group (23.95±3.15%) and MP+inhibitor control group (24.05±2.90%) were not significantly different from that of the MP group (23.28±2.43%; P=0.720 and 0.679, respectively). No significant differences were observed in apoptotic rate between the MP+mimic control group and MP+inhibitor control group (P=0.956). Compared with the MP+mimic control group, the apoptotic rate of the MP+miR-338-3p mimic group increased significantly (35.40±5.22%; P<0.001); compared with the MP+inhibitor control group, the apoptotic rate of the MP+miR-338-3p inhibitor group decreased significantly (13.01±1.02%; P<0.001).

SOX4 was verified as the target gene of miR-338-3p using a dual-luciferase reporter gene system

Online bioinformatics prediction software TargetScan, PicTar and microRNA.org predicted that the 3′-UTR in the SOX4 gene mRNA contained a locus matching the mmu-miR-338-3p mature sequence, indicating that SOX4 may be the target gene of miR-338-3p (Fig. 3A). A dual-luciferase reporter gene system was adopted to further verify whether SOX4 was the target gene on which miR-338-3p directly acted. The luciferase signal of miR-338-3p mimic and pmirGLO-SOX4-3′-UTR co-transfection group decreased by 50% compared with the other groups (miR-338-3p mimic control+WT-SOX4, miR-338-3p mimic+Mut-SOX4 and miR-338-3p mimic control+Mut-SOX4 groups; P=0.003, 0.002 and 0.003, respectively). However, when the mutated control pmirGLO-SOX4 mut-3′-UTR was co-transfected, the luciferase signal did not decrease and no significant differences occurred among groups regardless of the expression of miR-338-3p mimic or mimic control (miR-338-3p mimic control+WT-SOX4 group vs. miR-338-3p mimic+Mut-SOX4 group, P=0.887; miR-338-3p mimic control+WT-SOX4 group vs. miR-338-3p mimic control+Mut-SOX4 group, P=0.810; miR-338-3p mimic+Mut-SOX4 group vs. miR-338-3p mimic control+Mut-SOX4 group, P=0.692) (Fig. 3B). The result demonstrated that SOX4 expression was inhibited following miR-338-3p binding to the appropriate site in the 3′-UTR of SOX4. SOX4 may therefore be the target gene that miR-338-3p directly regulates.

Effects of miR-338-3p on SOX4 mRNA and protein expression levels in MP-treated macrophages

SOX4 mRNA (Table IV; Fig. 4) and protein (Table IV; Fig. 5) expression levels decreased significantly in the MP group (P<0.001), MP+mimic control group (P<0.001), MP+inhibitor control group (P<0.001), MP+miR-338-3p mimics group (P<0.001) and MP+miR-338-3p inhibitors group (P=0.040 and P<0.001, for mRNA and protein expression, respectively) compared with the blank group. SOX4 mRNA and protein expression levels showed no significant differences between the MP group and the MP+mimic control and MP+inhibitor control groups (mRNA expression, P=0.616 and 0.361, respectively; protein expression, P=0.681 and 0.584, respectively). Compared with the MP+mimic control group, SOX4 mRNA and protein expression levels in the MP+miR-338-3p mimic group decreased significantly (mRNA expression, P=0.016; protein expression, P=0.005); compared with the MP+inhibitor control group, SOX4 mRNA and protein expression levels in the MP+miR-338-3p inhibitor group increased significantly (mRNA expression, P=0.011; protein expression, P=0.001).

Table IV

Relative mRNA and protein expression levels of SOX4 detected by reverse transcription-quantitative polymerase chain reaction and western blotting.

Table IV

Relative mRNA and protein expression levels of SOX4 detected by reverse transcription-quantitative polymerase chain reaction and western blotting.

GroupRelative SOX4 mRNA expressionRelative SOX4 protein expression
Blank 1.00±0.00 1.00±0.00
MP 0.60±0.12a 0.42±0.10a
MP+mimic control 0.56±0.14a 0.39±0.12a
MP+inhibitor control 0.52±0.09a 0.38±0.08a
MP+miR-338-3p mimic 0.32±0.07a,b 0.18±0.05ac
MP+miR-338-3p inhibitor 0.81±0.13a,c,d,e 0.68±0.11a,c,d,e

a P<0.05 vs. blank;

b P<0.05 vs. MP;

c P<0.05 vs. MP+mimic control;

d P<0.05 vs. MP+inhibitor control;

e P<0.05 vs. MP+miR-338-3p mimic. ANOVA results (SOX4 mRNA expression): F-value, 15.95; P-value, <0.001. ANOVA results (SOX4 protein expression): F-value, 33.09; P-value, <0.001. MP, morphine; miR, microRNA; SOX4, sex determining region Y-box 4; ANOVA, analysis of variance.

Effects of miR-338-3p on the apoptosis-associated factor, caspase-3, protein expression levels in MP-treated macrophages

Caspase-3 protein expression levels (Table V; Fig. 6) in the MP, MP+inhibitor control and MP+mimic control groups increased significantly compared with the blank group (P<0.001). No significant differences were observed between the MP group and the MP+mimic control and MP+inhibitor control groups (P=0.921 and 0.962, respectively), as well as between the MP+mimic control group and MP+inhibitor control group (P=0.962). Compared with the blank group, caspase-3 expression levels in the MP+miR-338-3p inhibitor and MP+miR-338-3p mimic groups increased significantly (P<0.001 and P=0.001, respectively). Compared with the MP+mimics control group, caspase-3 protein expression levels in the MP+miR-338-3p mimic group increased significantly (P<0.001); compared with the MP+inhibitor control group, caspase-3 protein expression levels in the MP+miR-338-3p inhibitor group decreased significantly (P=0.012).

Table V

Relative protein expression levels of caspase-3 detected by western blotting.

Table V

Relative protein expression levels of caspase-3 detected by western blotting.

GroupRelative caspase-3 protein expression
Blank0.99±0.12
MP2.51±0.31a
MP+mimic control2.49±0.35a
MP+inhibitor control2.50±0.25a
MP+miR-338-3p mimic4.55±0.27ac
MP+miR-338-3p inhibitor1.90±0.11a,b,d,e

a P<0.05 vs. blank;

b P<0.05 vs. MP;

c P<0.05 vs. MP+mimic control;

d P<0.05 vs. MP+inhibitor control;

e P<0.05 vs. MP+miR-338-3p mimic. Analysis of variance results: F-value, 66.99; P-value, <0.001. MP, morphine; miR, microRNA.

Discussion

Previous studies have demonstrated that MP promotes apoptosis (11,12). miR-338-3p has been revealed to suppress signaling pathways, including p38, mitogen-activated protein kinase and AKT, which additionally suppress the proliferation and migration of cancer cells (20). Therefore, MP may promote apoptosis by regulating the expression of miR-338-3p. The aims of the present study were to investigate the association between MP treatment and the expression levels of miR-338-3p, define the underlying mechanism of the effects of miR-338-3p, and investigate whether miR-338-3p is a potential target molecule for the diagnosis and treatment of cancer.

In the present study, MP significantly increased the expression levels of miR-338-3p and enhanced apoptosis. The apoptosis rate decreased significantly when miR-338-3p was inhibited. Previous studies have demonstrated that miR-338-3p promotes apoptosis of cancer cells (26,27). In the present study, it was demonstrated that miR-338-3p may target the SOX4 gene and decrease SOX4 mRNA and protein expression levels. According to a previous study, SOX4 expression levels were significantly upregulated during cancer (28), which may inhibit apoptosis (29,30). SOX4 suppresses apoptosis by inhibiting the expression of caspase-3, which may result in the proliferation of cancer cells (31). miR-338-3p may suppress the overexpression of SOX4 and prevent the decrease in caspase-3 expression. Therefore, following MP treatment, the increased expression levels of miR-338-3p would decrease the expression levels of SOX4, resulting in an increase in caspase-3 expression levels and the promotion of apoptosis. In the present study, the suppression of SOX4 expression by miR-338-3p was prevented by miR-338-3p inhibitors, resulting in a significant decrease in protein expression levels of caspase-3. This resulted in a decrease in the apoptosis rate. Consistent with previous studies, MP promoted apoptosis in the present study (1113). Thus, upregulated expression of miR-338-3p may be a primary mediator of the therapeutic effect exerted by MP.

On the other hand, a previous study observed that partial miR expression was easily modulated under the influence of MP (32); in addition, numerous miRs have been demonstrated to exhibit increased expression during addiction (31,33,34). Therefore, the upregulated expression of miR-338-3p following MP-treatment may be associated with the effects of addiction. The detailed underlying mechanism of this effect remains to be elucidated. However, the side effects associated with MP should not be ignored, as overuse of MP may result in abnormal apoptosis of cells, particularly nerve cells (13,35). Strategies to increase miR-338-3p expression in the absence of MP may be of potential benefit in cancer treatment (21).

In conclusion, the results of the present study demonstrate that MP treatment upregulated the expression levels of miR-338-3p and enhanced its suppression of SOX4 expression, which resulted in increased expression of caspase-3 and increased apoptosis. The apoptosis rate decreased when the expression of miR-338-3p was inhibited. miR-338-3p may be important in MP treatment of cancer, as a mediator of MP-induced apoptosis. The upregulation of miR-338-3p expression provides a potential strategy for the treatment of cancer.

References

1 

Farazi TA, Hoell JI, Morozov P and Tuschl T: MicroRNAs in human cancer. Adv Exp Med Biol. 774:1–20. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Yu B, Qian T, Wang Y, Zhou S, Ding G, Ding F and Gu X: miR-182 inhibits Schwann cell proliferation and migration by targeting FGF9 and NTM, respectively at an early stage following sciatic nerve injury. Nucleic Acids Res. 40:10356–10365. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Rahmann S, Martin M, Schulte JH, Köster J, Marschall T and Schramm A: Identifying transcriptional miRNA biomarkers by integrating high-throughput sequencing and real-time PCR data. Methods. 59:154–163. 2013. View Article : Google Scholar

4 

Gurtan AM and Sharp PA: The role of miRNAs in regulating gene expression networks. J Mol Biol. 425:3582–3600. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Kong X, Xu X, Yan Y, Guo F, Li J, Hu Y, Zhou H and Xun Q: Estrogen regulates the tumour suppressor MiRNA-30c and its target gene, MTA-1, in endometrial cancer. PLoS One. 9:e908102014. View Article : Google Scholar : PubMed/NCBI

6 

Fiori E, Babicola L, Andolina D, Coassin A, Pascucci T, Patella L, Han YC, Ventura A and Ventura R: Neurobehavioral alterations in a genetic murine model of Feingold Syndrome 2. Behav Genet. 45:547–559. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ and Pandolfi PP: A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature. 465:1033–1038. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Sayed D and Abdellatif M: MicroRNAs in development and disease. Physiol Rev. 91:827–887. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Takahashi RU, Miyazaki H and Ochiya T: The Roles of MicroRNAs in Breast Cancer. Cancers (Basel). 7:598–616. 2015. View Article : Google Scholar

10 

Dahiya N and Atreya CD: MicroRNAs and Major Blood-borne Infectious Viral Diseases. Microrna. 2:212–218. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Bimonte S, Barbieri A, Rea D, Palma G, Luciano A, Cuomo A, Arra C and Izzo F: Morphine promotes tumor angiogenesis and increases breast cancer progression. Biomed Res Int. 2015:1615082015. View Article : Google Scholar : PubMed/NCBI

12 

Nomura Y, Kawaraguchi Y, Sugimoto H, Furuya H and Kawaguchi M: Effects of morphine and fentanyl on 5-fluorouracil sensitivity in human colon cancer HCT116 cells. J Anesth. 28:298–301. 2014. View Article : Google Scholar

13 

Gonzalez-Nunez V, Noriega-Prieto JA and Rodriguez RE: Morphine modulates cell proliferation through mir133b &mir128 in the neuroblastoma SH-SY5Y cell line. Biochim Biophys Acta. 1842:566–572. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Zheng H, Zeng Y, Chu J, Kam AY, Loh HH and Law PY: Modulations of NeuroD activity contribute to the differential effects of morphine and fentanyl on dendritic spine stability. J Neurosci. 30:8102–8110. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Sanchez-Simon FM, Zhang XX, Loh HH, Law PY and Rodriguez RE: Morphine regulates dopaminergic neuron differentiation via miR-133b. Mol Pharmacol. 78:935–942. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Nadeau SE: Opioids for chronic noncancer pain: To prescribe or not to prescribe-What is the question? Neurology. 85:646–651. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Oosten AW, Oldenmenger WH, Mathijssen RH and van der Rijt CC: A systematic review of prospective studies reporting adverse events of commonly used opioids for Cancer-Related Pain: A call for the use of standardized outcome measures. J Pain. 16:935–946. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Li MC, Yu JH, Yu SS, Chi YY and Xiang YB: MicroRNA-873 Inhibits Morphine-Induced Macrophage Apoptosis by Elevating A20 Expression. Pain Med. 16:1993–1999. 2015. View Article : Google Scholar : PubMed/NCBI

19 

He Y, Yang C, Kirkmire CM and Wang ZJ: Regulation of opioid tolerance by let-7 family microRNA targeting the mu opioid receptor. J Neurosci. 30:10251–10258. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Peng Y, Liu YM, Li LC, Wang LL and Wu XL: MicroRNA-338 inhibits growth, invasion and metastasis of gastric cancer by targeting NRP1 expression. PLoS One. 9:e944222014. View Article : Google Scholar : PubMed/NCBI

21 

Guo B, Liu L, Yao J, Ma R, Chang D, Li Z, Song T and Huang C: miR-338-3p suppresses gastric cancer progression through a PTEN-AKT axis by targeting P-REX2a. Mol Cancer Res. 12:313–321. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Wood MM and Cousins MJ: Iatrogenic neurotoxicity in cancer patients. Pain. 39:1–3. 1989. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

Rogler CE, Levoci L, Ader T, Massimi A, Tchaikovskaya T, Norel R and Rogler LE: MicroRNA-23b cluster microRNAs regulate transforming growth factor-beta/bone morphogenetic protein signaling and liver stem cell differentiation by targeting Smads. Hepatology. 50:575–584. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Osnes T, Sandstad O, Skar V, Osnes M and Kierulf P: Total protein in common duct bile measured by acetonitrile precipitation and a micro bicinchoninic acid (BCA) method. Scand J Clin Lad Invest. 53:757–763. 1993. View Article : Google Scholar

26 

Li P, Chen X, Su L, Li C, Zhi Q, Yu B, Sheng H, Wang J, Feng R, Cai Q, et al: Epigenentic silencing of miR-338-3p contributed to tumorigenicity in gastric cancer by targeting SSX2IP. PLoS One. 8:e667822013. View Article : Google Scholar

27 

Chen X, Pan M, Han L, Lu H, Hao X and Dong Q: miR-338-3p suppresses neuroblastoma proliferation, invasion and migration through targeting PREX2a. FEBS Lett. 587:3729–3737. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Gunes S, Yegin Z, Sullu Y, Buyukalpelli R and Bagci H: SOX4 expression levels in urothelial bladder carcinoma. Pathol Res Pract. 207:423–427. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Zhou Y, Wang X, Huang Y, Chen Y, Zhao G, Yao Q, Jin C, Huang Y, Liu X and Li G: Down-regulated SOX4 expression suppresses cell proliferation, metastasis and induces apoptosis in Xuanwei female lung cancer patients. J Cell Biochem. 116:1007–1018. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Jang SM, Kang EJ, Kim JW, Kim CH, An JH and Choi KH: Transcription factor Sox4 is required for PUMA-mediated apoptosis induced by histone deacetylase inhibitor, TSA. Biochem Biophys Res Commun. 438:445–451. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Liu LW, Lu J, Wang XH, Fu SK, Li Q and Lin FQ: Neuronal apoptosis in morphine addiction and its molecular mechanism. Int J Clin Exp Med. 6:540–545. 2013.PubMed/NCBI

32 

Li MD and van der Vaart AD: MicroRNAs in addiction: Adaptation's middlemen? Mol Psychiatry. 16:1159–1168. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Dreyer JL: New insights into the roles of microRNAs in drug addiction and neuroplasticity. Genome Med. 2:922010. View Article : Google Scholar

34 

Hollander JA, Im HI, Amelio AL, Kocerha J, Bali P, Lu Q, Willoughby D, Wahlestedt C, Conkright MD and Kenny PJ: Striatal microRNA controls cocaine intake through CREB signalling. Nature. 466:197–202. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Rodríguez RE: Morphine and microRNA Activity: Is There a Relation with Addiction? Front Genet. 3:2232012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 14 Issue 3

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
Weng HL and Weng HL: Effects of microRNA‑338‑3p on morphine‑induced apoptosis and its underlying mechanisms. Mol Med Rep 14: 2085-2092, 2016
APA
Weng, H., & Weng, H. (2016). Effects of microRNA‑338‑3p on morphine‑induced apoptosis and its underlying mechanisms. Molecular Medicine Reports, 14, 2085-2092. https://doi.org/10.3892/mmr.2016.5506
MLA
Weng, H., Wang, M."Effects of microRNA‑338‑3p on morphine‑induced apoptosis and its underlying mechanisms". Molecular Medicine Reports 14.3 (2016): 2085-2092.
Chicago
Weng, H., Wang, M."Effects of microRNA‑338‑3p on morphine‑induced apoptosis and its underlying mechanisms". Molecular Medicine Reports 14, no. 3 (2016): 2085-2092. https://doi.org/10.3892/mmr.2016.5506