Open Access

Suppression of microRNA-130b inhibits glioma cell proliferation and invasion, and induces apoptosis by PTEN/AKT signaling

  • Authors:
    • Jian-Jun Gu
    • Kai-Chun Fan
    • Jian-He Zhang
    • Hong-Jie Chen
    • Shou-Sen Wang
  • View Affiliations

  • Published online on: November 2, 2017     https://doi.org/10.3892/ijmm.2017.3233
  • Pages: 284-292
  • Copyright: © Gu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioblastoma is the most common malignant brain tumor in adults and is characterized by extensive proliferation and the diffused invasion of tumor cells. Due to the intricate signaling pathways involved in glioma progression, more effective targeted therapies and prognostic biomarkers in clinical practice are required. The suppression of proto-oncogene function or recovery of tumor suppressor gene function remains one of the primary approaches in gene therapy. The close association between the abnormal expression or mutation of microRNA (miRNA) and the tumorigenesis, progression and staging in glioma have been demonstrated previously. However, the expression pattern and specific role of microRNA‑130b (miR‑130b) in the tumor occurrence and progression of glioma are unclear. In the present study, quantitative polymerase chain reaction was performed to determine the expression level of miR-130b in 30 brain glioma patients and 3 glioma cell lines. An miR‑130b inhibitor was transfected into U87 cells to downregulate the expression of miR-130b, and assessments of cell proliferation, cell cycle, apoptosis, cell invasion and migration in vitro and nude mouse tumorigenicity in vivo were conducted. Western blotting and luciferase reporter gene technology were used to verify the downstream target gene of miR-130b, namely phosphatase and tensin homolog (PTEN). The results demonstrated that miR-130b expression was increased in glioma tissues and cell lines in comparison with non-glioma tissues or cells. The downregulated expression of miR-130b inhibited the proliferation and invasion of glioma cells, induced apoptosis of the cells in vitro and inhibited their tumorigenicity in vivo. Western blotting and luciferase reporter assays demonstrated that the PTEN gene is a direct target of miR‑130b. Western blotting revealed that the miR-130b inhibitor upregulated the expression of PTEN, inhibited AKT pathway activation, upregulated the tumor suppressor gene p27, and suppressed cyclin D1, matrix metalloproteinase 2 and 9 expression. These results suggest that the miR-130b inhibitor suppressed glioma cell proliferation and invasion via the PTEN/AKT pathway. Therefore, miR‑130b is suggested to be an effective therapeutic target for glioma.

Introduction

Glioma is the most common primary brain tumor, accounting for 80% of malignant brain tumors, and has a high rate of recurrence, mortality and morbidity (1). The excessive proliferation, resistance to apoptosis and high invasion rate of glioma cells lead to the characteristic lack of boundaries with the surrounding brain tissues, regardless of the differentiation of glioma cells (2). It is thus challenging to completely eradicate the tumor by surgery. With current standard management by surgical resection and precise radiotherapy and chemotherapy, the median survival time of patients with glioblastoma multiforme is only 12–15 months, and the 5-year survival rate remains <3% (3). Therefore, elucidation of the mechanism underlying the tumorigenesis, tumor development and clonal evolution of glioma is essential for the identification of an effective target for gene therapy.

MicroRNAs (miRNAs) are a class of small endogenous non-coding RNAs that are ~22 nucleotides in length. These regulate gene expression post-transcriptionally, primarily through complete or incomplete integration with the mRNA 3′ untranslated region (3′UTR) of the target gene. A single miRNA molecule may regulate hundreds of target genes by forming a complex regulatory network with the target genes. miRNAs serve broad roles in embryonic development, cell proliferation, differentiation, apoptosis, cell cycle progression, angiogenesis and a variety of other biological processes (4). Studies have shown that the aberrant expression of specific miRNAs is closely associated with uncontrolled proliferation, apoptosis blockage, invasion and migration, as well as resistance to chemotherapy (57). MicroRNA-130b (miR-130b) is an important member of the microRNA-130 family. A previous study demonstrated that in breast tissues, miR-130b downregulates p21Waf1/Cip1, a cell cycle inhibitor, which in turn inhibits mammary epithelial cell apoptosis and promotes tumor growth (8). In addition, Dong et al (9) reported that a mutant p53 gene induced epithelial-mesenchymal transition by regulating the miR-130b-zinc finger E-box binding homeobox 1 axis in endometrial carcinoma, resulting in the initiation of tumor development. They also observed that in comparison with normal tissues, miR-130b expression was significantly lower in endometrial carcinoma tissues, and the long-term follow-up of patients with endometrial carcinoma revealed a longer survival time for patients with high levels of miR-130b expression (9). These studies suggest that miR-130b is capable of functioning as either a tumor suppressor or a tumor promoter, depending on the type of tumor involved. A study conducted by Malzkorn et al (10) concluded that the expression of miR-130b significantly increased in four patients with glioma as they progressed from World Health Organization (WHO) stage II to IV. However, the functions and mechanisms of miR-130b in glioma require elucidation.

In the present study, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to determine the expression pattern of miR-130b in 30 glioma patients and 3 glioma cell lines. U87 cells were transfected with an miR-130b inhibitor to downregulate the expression of miR-130b. The biological characteristics of the proliferation, cell cycle, cell invasion and migration of the transfected U87 cells were then assessed. Western blotting and luciferase reporter gene technology were used to verify the expression of phosphatase and tensin homolog (PTEN), a downstream target gene of miR-130b, and its underlying mechanism.

Materials and methods

Clinical samples and cell lines

Human glioma tumor tissues were obtained from the surgical resection of glioma patients (52–73 years old; 21 males and 9 females) in the Fuzhou General Hospital (Fuzhou, China) between January 2015 and December 2016. A total of 30 glioma samples were thoroughly reviewed by an experienced neuropathologist according to the 2007 WHO classification (11), resulting in 10 glioma samples being classified as grades I and II, 10 as grade III and 10 as grade IV. In addition, 5 non-neoplastic brain specimens were obtained from patients with traumatic brain injury during decompression surgery. All tissue samples were frozen in liquid nitrogen immediately following resection and stored at −80°C. Prior patient consent was obtained for the use of these clinical materials for research purposes. In addition, ethical approval for the study was obtained from the Ethics Committee of Fuzhou General Hospital. The normal human astrocyte (NHA) cell line was purchased from ScienCell Research Laboratories, Inc. (Carlsbad, CA, USA). The glioma LN229, U87 and U251 cell lines used in this study were obtained in 2013 from the Institute of Biochemistry and Cell Biology (Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai, China).

Cell culture and transfection

Cells were grown in Dulbecco's modified Eagle's medium (DMEM; Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco, Grand Island, NY, USA) and 1% penicillin/streptomycin in a 5% CO2 atmosphere at 37°C. U87 cells (1×105) were seeded into 6-well plates and transfected with negative control (NC), miR-130b inhibitor and PTEN-targeted small interfering RNA (siPTEN; sequence, 5′-GACUUGAAGGCGUAUACAGtt-3′) purchased from Shanghai GenePharma Co., Ltd. (Shanghai, China) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) following the manufacturer's protocol. The medium was removed following a 24 h transfection, and the cells were placed in the complete medium and maintained at 37°C in 5% CO2.

RNA extraction and RT-qPCR

Total RNAs were extracted from cultured cells and fresh glioma tissues using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) or total miRNAs using mirVana (Ambion; Thermo Fisher Scientific, Inc.) kits according to the manufacturer's protocols. Reverse transcription was then performed by using a Reverse Transcription kit (Promega Corporation, Madison, WI, USA) in accordance with the instructions of the manufacturer. Gene-specific primers were used to synthesize miR-130b cDNA from total RNA, in accordance with the recommended protocol for the miRNA-specific TaqMan miRNA Assay kit (Applied Biosystems; Thermo Fisher Scientific, Inc.); U6 small nuclear RNA (snRNA) was used as an internal control. The expression level of miR-130b and PTEN was examined by qPCR with a SYBR-Green PCR Master Mix kit in conjunction with an ABI-Prism 7300 system (both Applied Biosystems; Thermo Fisher Scientific, Inc.). The primer sequences were as follows: PTEN forward, 5′-TTTGAAGACCATAACCCACCAC-3′ and reverse, 5′-ATTACACCAGTTCGTCCCTTTC-3′; GAPDH forward, 5′-TCGGAGTCAACGGATTTGG-3′ and reverse, 5′-CATGGGTGGAATCATATTGGA-3′; U6 RT-primer, 5′-CGCTTCACGAATTTGCGTGTCAT-3′, forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′. The conditions for PCR were as follows: stage 1, 95°C for 10 min; stage 2, 40 cycles at 95°C for 15 sec, 57°C for 1 min; stage 3 (dissociation stage), 95°C for 15 sec, 60°C for 15 sec, 95°C for 15 sec. The relative expression levels of mature miR-130b and PTEN mRNA were calculated by the 2−ΔΔCq method (12) and normalized to U6 snRNA and GAPDH mRNA levels, respectively.

Western blotting

Total cell lysates from different experiments were obtained by lysing the cells in radioimmunoprecipitation assay buffer (Sigma, St. Louis, MO, USA). Protein concentrations were determined using a BCA protein assay (Pierce; Thermo Fisher Scientific, Inc.). Total protein from each sample (40 μg) was resolved by 10% SDS-PAGE and transferred to PVDF membranes (both from EMD Millipore, Billerica, MA, USA). The membranes were incubated with primary antibodies PTEN (1:1,000 dilution; Abcam, Cambridge, MA, USA), phosphorylated (p)-AKT (1:500 dilution; sc-7985-R), p27 (1:500 dilution; sc-528) and cyc1in D1 (1:500 dilution; sc-70899) (all from Santa Cruz Biotechnology, Inc., Dallas, TX, USA), anti-matrix metalloproteinase (MMP)-2 (1:500 dilution) and anti-MMP-9 (1:500 dilution) (both from Abcam) overnight at 4°C, followed by incubation with a horseradish peroxidase-conjugated donkey anti-rabbit secondary antibody (sc-2317; 1:5,000 dilution; Santa Cruz Biotechnology, Inc.) or HRP-conjugated donkey anti-mouse antibody (sc-2318;1:5,000 dilution; Santa Cruz Biotechnology, Inc.) at room temperature for 30 min. Membranes were stripped and then reprobed with a primary antibody against GAPDH (1:1;000 dilution; Santa Cruz Biotechnology, Inc.). The signal intensity was determined with ImageJ gel analysis software version 1.48u (National Institutes of Health, Bethesda, MD, USA). GAPDH was used as an endogenous protein for normalization. Specific bands were visualized using enhanced chemiluminescence detection (Thermo Fisher Scientific, Inc.).

Luciferase reporter assay

The 3′UTR sequence of PTEN predicted to interact with miR-130b or a mutated sequence with the predicted target sites was synthesized and inserted into the XbaI and FseI sites of a pGL3 control vector (Promega Corporation) by integrating the search results of TargetScan and miRDB programs (13). These constructs were designated pGL3-PTEN-3′-UTR and pGL3-PTEN-3′-UTR-mut, respectively. For the reporter assay, U87 cells were plated onto 24-well plates and transfected with pGL3, pGL3-PTEN-3′-UTR or pGL3-PTEN-3′-UTR-mut vector and miR-130b inhibitor or NC vectors using the FuGENE HD transfection reagent (Promega Corporation). A Renilla luciferase vector pRL-SV50 (Promega Corporation) was co-transfected in order to normalize the differences in transfection efficiency. Subsequent to 48 h transfection, the cells were harvested and assayed using a Dual-Luciferase Reporter Assay system (Promega Corporation), according to the manufacturer's protocol. Transfection was repeated in triplicate in three independent experiments.

Proliferation assays

Cells (5×103/well) were plated in 96-well plates and grown for 24, 48, 72 and 96 h following transfection. Cell proliferation was documented every 24 h for 4 days using a Cell Counting kit-8 (CCK-8) assay (Genview, Carlsbad, CA, USA). The absorbance at a wavelength of 570 nm was detected using a microplate reader.

Colony formation assay

The stably-transfected glioma cells (300 cells/well) were seeded into 6-well plates and cultured in cell culture medium for 2 weeks to allow colony formation. The culture medium was refreshed every third day. The colonies were then fixed in 100% methanol for 30 min and stained with crystal violet solution for 30 min. Subsequently, the number of macroscopically observable colonies was recorded.

Cell cycle and apoptosis assays

The effects of miR-310b on the cell cycle and apoptosis of U87 glioma cells were examined by flow cytometry. In brief, the cells were harvested by trypsinization 48 h following transfection. They were then washed three times with ice-cold phosphate-buffered saline (PBS) and fixed with 70% ethanol overnight at 4°C. The cell cycle and apoptotic rate were measured by flow cytometry (BD Biosciences, San Jose, CA, USA) and the data were analyzed using BD CellQuest™ PRO software (BD Biosciences). Annexin V-APC/7-AAD Apoptosis Detection kit (KaiJi, Nanjing, China) was used.

Transwell and scratch-wound assays

Cell invasion was determined using Transwell and scratch-wound assays. For the Transwell assay, U87 cells were transfected with miR-130b inhibitor or NC. Subsequent to incubation for 48 h, 3×104 cells were transferred to the top of a Matrigel-coated invasion chamber (BD Biosciences) in serum-free DMEM. DMEM containing 10% FBS was added to the lower chamber. After 24 h, the non-invading cells were removed, and the invading cells were fixed using 95% ethanol for 30 min, stained with 0.1% crystal violet for 30 min and images were captured under a magnification of ×100. For the scratch-wound assay, miR-130b inhibitor or NC was transfected into the cells in 6-well plates. The cell layers were then scratched using a 20-μl sterile pipette tip to form wound gaps. The wound location in the 6-well plates was marked and the cells were then allowed to migrate into the cell-free wound in serum-free DMEM for 24 h. The tests were repeated in three independent experiments. The wound widths were measured and the relative wound widths were calculated. Data are shown as means ± standard deviation (SD) of 3 independent experiments.

Nude mouse tumorigenicity

U87 cells stably-transfected with miR-130b inhibitor or NC were resuspended in PBS and 1.5×106 cells were injected subcutaneously into the right flanks of male BALB/c athymic (nude) mice (6–8 weeks old; weighing 20 ± 2 g obtained from Shanghai SLAC Experimental Animal Co., Ltd., Shanghai, China). Mice were maintained in a pathogen-free environment. The experiment was conducted using 10 mice (n=5/group). Tumor volumes were measured at different time-points using the formula: Tumor volume = length (mm) × width2 (mm2)/2.

Statistical analysis

All data are shown as mean ± standard deviation, and the experiments were repeated three times. Statistical analyses were performed using a two-tailed Student's t-test with SPSS version 12.0 software (SPSS, Inc., Chicago, IL, USA) and multiple-group comparisons were analyzed using one-way ANOVA for the subsequent individual group comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

Upregulation of miR-130b in glioma tissues and cells

The expression levels of miR-130b in 30 glioma cases, 5 normal brain tissues and 3 glioma cell lines were detected using RT-qPCR. The results demonstrated that miR-130b expression in glioma tissues was significantly higher than that in normal brain tissues (P<0.05; Fig. 1A), and its expression in high level glioma tissues (III and IV) was significantly higher (P<0.05) than in low grade glioma (I/II). Furthermore, miR-130b expression in the 3 glioma cell lines was significantly upregulated compared with that in the NHAs (P<0.05; Fig. 1B).

PTEN is a potential downstream target of miR-130b

Using the TargetScan bioinformatic database, the target site for miR-130b interaction was detected in the 3′UTR of the PTEN gene. Fig. 2A shows that the target sequence was located at nucleotides 412–418 of the 3′UTR region of the PTEN gene. Then, the 3′-UTR of a PTEN fragment containing the miR-130b interaction site was ligated into a vector at a region downstream of the luciferase reporter gene. The luciferase reporter assay revealed that following transfection with the miR-130b inhibitor, a significant increase in wild-type luciferase activity was observed (P<0.05). By contrast, the luciferase activity of the mutant showed no significant change compared with that of the NC (P>0.05; Fig. 2B). The present study thus shows that miR-130b directly binds the 3′UTR of the PTEN gene, thereby inhibiting its expression.

Inhibition of the proliferation and induction of apoptosis of U87 cells by miR-130b

To confirm that U87 glioma cell proliferation is inhibited by miR-130b, U87 cells were transfected with miR-130b inhibitor, and the CCK-8 colorimetric assay and colony formation proliferation experiments were conducted to examine the inhibitory effects of the miR-130b inhibitor on U87 cell proliferation. At ~24 h after transfection, the cells were harvested and seeded at a density of 1×105 cells/well in 6-well plates. The cells were collected at 24, 48, 72 and 96 h after plating, and cell proliferation was examined. The results demonstrated that following transfection of the miR-130b inhibitor, cell proliferation at 48, 72 and 96 h was significantly inhibited, with a lower proliferation rate compared with that of the NC (P<0.05; Fig. 3A). Similarly, the results of the colony formation assay suggested that the downregulation of miR-130b significantly inhibited the colony formation of U87 cells, compared with that of the NC (P<0.05; Fig. 3B and C). The cells transiently transfected with the miR-130b inhibitor were subjected to cell cycle analysis by flow cytometry. Compared with the NC, U87 cells transfected with the miR-130b inhibitor underwent G0/G1 cell cycle arrest (Fig. 3D and E). Furthermore, transient transfection with the miR-130b inhibitor significantly induced apoptosis of the U87 cells (Fig. 3F and G). These in vitro experiments suggest that the reduced expression of miR-130b suppressed glioblastoma cell proliferation via cell cycle arrest and the promotion of cell apoptosis.

Inhibition of nude mouse tumorigenicity by miR-130b

To substantiate the effect of the miR-130b inhibitor in glioma carcinogenesis, U87 cells transfected with miR-130b inhibitor or miR-130b NC were implanted into the right flanks of BALB/c athymic mice by subcutaneous injection. At 28 days post-injection, the mean volumes of tumors generated from the miR-130b cells were significantly smaller than those originating from NC cells (Fig. 4A–C). Western blotting for the glioma xenograft tissues revealed that expression of PTEN was higher in the miR-130b inhibitor group compared with the NC group (Fig. 4D).

Inhibition of the invasion and migration of U87 cells by the miR-130b inhibitor

Following the transfection of U87 cells with the miR-130b inhibitor for 24 h, cell invasion ability was examined using a Transwell assay. U87 cells transfected with the miR-130b inhibitor exhibited a significant reduction in the number of cells that traversed the membrane compared with that of the NC (P<0.05; Fig. 5A and B). The wound healing assay also demonstrated that cell migration was inhibited in the U87 cells transfected with the miR-130b inhibitor compared with the NC (Fig. 5C and D).

Inhibition of the AKT signaling pathway by an miR-130b inhibitor via PTEN upregulation

The miR-130b inhibitor was transiently transfected into U87 cells to reduce miR-130b expression, and then total cellular RNA and protein were extracted following 48 h of transfection. PTEN expression at the RNA and protein levels was measured. The results demonstrated that compared with cells transfected with NC, when miR-130b expression was downregulated via transfection with the miR-130b inhibitor, PTEN was clearly upregulated at the protein level (Fig. 6A), but no significant difference in the level of mRNA expression was observed (Fig. 6B). The phosphorylation of AKT was significantly inhibited, and the tumor suppressor gene p27 was upregulated by the miR-130b inhibitor. In addition, cyclin D1, MMP-2 and MMP-9 expression was decreased.

Knockdown of endogenous PTEN reverses the effect of the miR-130b inhibitor

To confirm that miR-130b and PTEN serve a role in glioma cell proliferation and invasion, U87 cells were co-transfected with the miR-130b inhibitor and siPTEN. At 48 h after transfection, total proteins were extracted and western blotting demonstrated that the protein expression level of PTEN was markedly reduced (Fig. 7). The protein of PTEN was markedly reduced when U87 cells were co-transfected with the miR-130b inhibitor and siPTEN, the phosphorylation of AKT was significantly activated, and the tumor suppressor gene p27 was downregulated. In addition, cyclin D1, MMP-2 and MMP-9 expression was increased. CCK-8 and Transwell assay results indicated that following co-transfection with the miR-130b inhibitor and siPTEN U87, cell proliferation and cell invasion were markedly increased compared with that of the cells transfected with miR-130b inhibitor alone (Fig. 8A–C). Cell cycle analysis demonstrated that U87 cells co-transfected with the miR-130b inhibitor and siPTEN presented a lower percentage of cells in the G1 phase and a higher percentage of cells in the S phase (Fig. 8D). The analysis of apoptosis by flow cytometry revealed that the knockdown of endogenous PTEN attenuated the apoptosis-inducing effect of the miR-130b inhibitor (Fig. 8E and F). These findings confirm that miR-130b inhibits glioblastoma cell proliferation and invasion and induces their apoptosis via the downregulation of PTEN expression.

Discussion

Glioma is the most common malignancy and the most lethal adult brain tumor (3). Due to the abnormal cellular and molecular heterogeneity of tumor cells, the treatment of glioma using surgery combined with radiotherapy and comprehensive chemotherapy with temozolomide has remained unsatisfactory, often resulting in short survival times and poor prognosis. In the past decade, the median survival time of patients with glioblastoma was only 15 months (14,15). Research on the diagnosis and treatment of glioma is now particularly focused at the molecular mechanism underlying glioma tumorigenesis and development, particularly in the evolution of a variety of genes. Therefore, the discovery of the fundamental disease genes associated with glioma may provide an effective treatment for the targeted prevention and control of the disease at the genetic level. A number of studies have shown that miRNAs serve an important role in the initiation and progression of human cancer, acting as either oncogenes or tumor suppressor genes (16,17). It has been confirmed that a single miRNA molecule may act on multiple or even hundreds of target genes, and several different miRNAs may be able to regulate the same gene, so that miRNAs and their targeting proteins form a complex regulatory network that is important in regulating growth, development and tumorigenesis (18).

The miR-130 family includes miR-130a and b. The human miRNA-130b gene consists of 22 nucleotides (5′-CAGUGCA AUGAUGAAAGGGCAU-3′). miR-130a and miR-130b are co-localized on chromosome 22, located 10 kb apart, and can be expressed in cells in tandem, suggesting that they may have similar biological functions (19). The present study detected abnormal miR-130b expression in a variety of gliomas, which was consistent with the increased expression of miR-130b in melanoma (20), gastric cancer (21), bladder cancer (22), colorectal cancer (23), metastatic renal carcinoma (24) and glioma (10), whereas miR-130b has been found to be downregulated in papillary thyroid carcinomas (25), endometrial cancer (9), pituitary adenoma (26) and pancreatic cancer (27). In addition, miR-130b has an association with neurodegenerative diseases such as Alzheimer's and Parkinson's diseases (28), and inhibits lipogenesis (20). Tu et al (29) detected the increased expression of miR-130b in human hepatocellular carcinoma (HCC) and liver cancer cell lines, which was associated with a poor prognosis in liver cancer patients. They also demonstrated that the inhibition of miR-130b expression upregulated its target genes through PPAR-γ, which resulted in inhibition of the invasion and migration of HCC. Furthermore, a study conducted by Zhao et al (27) demonstrated that the expression of miR-130b was reduced in pancreatic cancer, and identified an association of the downregulation of miR-130b with poor prognosis in pancreatic cancer patients. The study also found that the over-expression of miR-130b inhibited the proliferation and invasion of pancreatic cancer cells by downregulating the target gene signal transducer and activator of transcription 3. However, the expression pattern of miR-130b and the specific mechanisms underlying its effects in the pathogenesis of glioma have not been fully elucidated. The purpose of the present study was to investigate the mechanism by which miR-130b acts in glioma cell proliferation and invasion, and to determine whether it regulates the target gene, PTEN. In the present study RT-qPCR detected significantly higher miR-130b expression in human glioma tissues than in normal brain tissues, as well as significantly higher expression in the LN229, U87 and U251 cell lines compared with the NHA cell line. The downregulation of miR-130b in U87 glioma cells inhibited cell proliferation, invasion and migration and induced apoptosis. These findings indicate that miR-130b functions as an oncogene in glioma cells.

The PTEN gene is a tumor suppressor gene that is located on chromosome 10q23.3 and has dual specificity (30). PTEN, as a lipid phosphatase, dephosphorylates the second messenger phosphatidylinositol 3,4,5-triphosphate, blocks the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway, upregulates the tumor suppressor gene p27 and inhibits cyclin D1 accumulation to regulate the cell cycle, which in turn causes cell G0/G1 arrest that prevent cells from entering the S phase. p27 is the key mediator in the repression of the G1 phase by PTEN, as well as a downstream target for PTEN-dependent cell cycle arrest (31,32). PTEN also reduces AKT phosphorylation and inhibits MMP secretion, thereby inhibiting tumor invasion and migration (33). Human malignant glioma (astrocytoma and glioblastoma) has a high level of PTEN gene deletions and mutations, which leads to abnormal activation of the PI3K/Akt pathway, particularly an increase in Akt phosphorylation and activity that promotes tumor cell growth, proliferation, invasion and metastasis (34). The present study demonstrated that the transfection of U87 cells with an miR-130b inhi bitor promoted the expression of the target gene PTEN and reduced the phosphorylation of AKT. Glioma cells transfected with the miR-130b inhibitor also exhibited increased expression of the tumor suppressor gene p27, suppressed cyclin D1 expression and reduced cell proliferation with G0/G1 arrest, which prevented cells from entering the S phase. Finally, the miR-130b inhibitor also reduced Akt phosphorylation levels to inhibit MMP-2 and -9 expression, thereby suppressing U87 invasion and migration. The siRNA silencing of PTEN reversed the inhibition of cell proliferation and invasion and promoted the apoptosis-inducing effect of the miR-130b inhibitor. Thus, the present study demonstrated that miR-130b functions in the tumorigenesis and development of glioma via the PTEN/AKT signaling pathway.

In conclusion, the present study has elucidated the oncogenic functions of miR-130b in glioma, which involved the negatively regulation of the expression of PTEN. These results indicate that miR-130b is important in the proliferation, apoptosis and invasion of glioma, and suggest that the inhibition of miR-130b expression may be an effective treatment approach for human glioma.

References

1 

Ohgaki H and Kleihues P: Epidemiology and etiology of gliomas. Acta Neuropathol. 109:93–108. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Zhou X, Ren Y, Moore L, Mei M, You Y, Xu P, Wang B, Wang G, Jia Z, Pu P, et al: Downregulation of miR-21 inhibits EGFR pathway and suppresses the growth of human glioblastoma cells independent of PTEN status. Lab Invest. 90:144–155. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Wen PY and Kesari S: Malignant gliomas in adults. N Engl J Med. 359:492–507. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Liu Y, Yan W, Zhang W, Chen L, You G, Bao Z, Wang Y, Wang H, Kang C and Jiang T: miR-218 reverses high invasiveness of glioblastoma cells by targeting the oncogenic transcription factor LEF1. Oncol Rep. 28:1013–1021. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Gu JJ, Gao GZ and Zhang SM: miR-218 inhibits the migration and invasion of glioma U87 cells through the Slit2-Robo1 pathway. Oncol Lett. 9:1561–1566. 2015.PubMed/NCBI

7 

Jiang L, Wang C, Lei F, Zhang L, Zhang X, Liu A, Wu G, Zhu J and Song L: miR-93 promotes cell proliferation in gliomas through activation of PI3K/Akt signaling pathway. Oncotarget. 6:8286–8299. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Borgdorff V, Lleonart ME, Bishop CL, Fessart D, Bergin AH, Overhoff MG and Beach DH: Multiple microRNAs rescue from Ras-induced senescence by inhibiting p21(Waf1/Cip1). Oncogene. 29:2262–2271. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Dong P, Karaayvaz M, Jia N, Kaneuchi M, Hamada J, Watari H, Sudo S, Ju J and Sakuragi N: Mutant p53 gain-of-function induces epithelial-mesenchymal transition through modulation of the miR-130b-ZEB1 axis. Oncogene. 32:3286–3295. 2013. View Article : Google Scholar

10 

Malzkorn B, Wolter M, Liesenberg F, Grzendowski M, Stühler K, Meyer HE and Reifenberger G: Identification and functional characterization of microRNAs involved in the malignant progression of gliomas. Brain Pathol. 20:539–550. 2010. View Article : Google Scholar

11 

Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW and Kleihues P: The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114:97–109. 2007. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Yu T, Cao R, Li S, Fu M, Ren L, Chen W, Zhu H, Zhan Q and Shi R: MiR-130b plays an oncogenic role by repressing PTEN expression in esophageal squamous cell carcinoma cells. BMC Cancer. 15:292015. View Article : Google Scholar : PubMed/NCBI

14 

Wang Y and Jiang T: Understanding high grade glioma: molecular mechanism, therapy and comprehensive management. Cancer Lett. 331:139–146. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Carlsson SK, Brothers SP and Wahlestedt C: Emerging treatment strategies for glioblastoma multiforme. EMBO Mol Med. 6:1359–1370. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Li R and Li X, Ning S, Ye J, Han L, Kang C and Li X: Identification of a core miRNA-pathway regulatory network in glioma by therap eutically targeting miR-181d, miR-21, miR-23b, β-catenin, CBP, and STAT3. PLoS One. 9:e1019032014. View Article : Google Scholar

17 

Jun GJ, Zhong GG and Ming ZS: miR-218 inhibits the proliferation of glioma U87 cells through the inactivation of the CDK6/cyclin D1/p21Cip1/Waf1 pathway. Oncol Lett. 9:2743–2749. 2015.PubMed/NCBI

18 

Friedman RC, Farh KK, Burge CB and Bartel DP: Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 19:92–105. 2009. View Article : Google Scholar :

19 

Lee EK, Lee MJ, Abdelmohsen K, Kim W, Kim MM, Srikantan S, Martindale JL, Hutchison ER, Kim HH, Marasa BS, et al: miR-130 suppresses adipogenesis by inhibiting peroxisome proliferator-activated receptor gamma expression. Mol Cell Biol. 31:626–638. 2011. View Article : Google Scholar

20 

Sand M, Skrygan M, Sand D, Georgas D, Gambichler T, Hahn SA, Altmeyer P and Bechara FG: Comparative microarray analysis of microRNA expression profiles in primary cutaneous malignant melanoma, cutaneous malignant melanoma metastases, and benign melanocytic nevi. Cell Tissue Res. 351:85–98. 2013. View Article : Google Scholar

21 

Lai KW, Koh KX, Loh M, Tada K, Subramaniam MM, Lim XY, Vaithilingam A, Salto-Tellez M, Iacopetta B, Ito Y, et al: Singapore Gastric Cancer Consortium: MicroRNA-130b regulates the tumour suppressor RUNX3 in gastric cancer. Eur J Cancer. 46:1456–1463. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Scheffer AR, Holdenrieder S, Kristiansen G, von Ruecker A, Müller SC and Ellinger J: Circulating microRNAs in serum: novel biomarkers for patients with bladder cancer. World J Urol. 32:353–358. 2014. View Article : Google Scholar

23 

Colangelo T, Fucci A, Votino C, Sabatino L, Pancione M, Laudanna C, Binaschi M, Bigioni M, Maggi CA, Parente D, et al: MicroRNA-130b promotes tumor development and is associated with poor prognosis in colorectal cancer. Neoplasia. 15:1218–1231. 2013. View Article : Google Scholar

24 

Wu X, Weng L, Li X, Guo C, Pal SK, Jin JM, Li Y, Nelson RA, Mu B, Onami SH, et al: Identification of a 4-microRNA signature for clear cell renal cell carcinoma metastasis and prognosis. PLoS One. 7:e356612012. View Article : Google Scholar : PubMed/NCBI

25 

Yip L, Kelly L, Shuai Y, Armstrong MJ, Nikiforov YE, Carty SE and Nikiforova MN: MicroRNA signature distinguishes the degree of aggressiveness of papillary thyroid carcinoma. Ann Surg Oncol. 18:2035–2041. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Leone V, Langella C, D'Angelo D, Mussnich P, Wierinckx A, Terracciano L, Raverot G, Lachuer J, Rotondi S, Jaffrain-Rea ML, et al: miR-23b and miR-130b expression is downregulated in pituitary adenomas. Mol Cell Endocrinol. 390:1–7. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Zhao G, Zhang JG, Shi Y, Qin Q, Liu Y, Wang B, Tian K, Deng SC, Li X, Zhu S, et al: miR-130b is a prognostic marker and inhibits cell proliferation and invasion in pancreatic cancer through targeting STAT3. PLoS One. 8:e738032013. View Article : Google Scholar : PubMed/NCBI

28 

Shacka JJ, Roth KA and Zhang J: The autophagy-lysosomal degradation pathway: role in neurodegenerative disease and therapy. Front Biosci. 13:718–736. 2008. View Article : Google Scholar

29 

Tu K, Zheng X, Dou C, Li C, Yang W, Yao Y and Liu Q: MicroRNA-130b promotes cell aggressiveness by inhibiting peroxisome proliferator-activated receptor gamma in human hepatocellular carcinoma. Int J Mol Sci. 15:20486–20499. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Chen Z, Trotman LC, Shaffer D, Lin HK, Dotan ZA, Niki M, Koutcher JA, Scher HI, Ludwig T, Gerald W, et al: Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature. 436:725–730. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Sansal I and Sellers WR: The biology and clinical relevance of the PTEN tumor suppressor pathway. J Clin Oncol. 22:2954–2963. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Chung JH and Eng C: Nuclear-cytoplasmic partitioning of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) differentially regulates the cell cycle and apoptosis. Cancer Res. 65:8096–8100. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Yamada KM and Araki M: Tumor suppressor PTEN: modulator of cell signaling, growth, migration and apoptosis. J Cell Sci. 114:2375–2382. 2001.PubMed/NCBI

34 

Vivanco I and Sawyers CL: The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat Rev Cancer. 2:489–501. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 41 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gu J, Fan K, Zhang J, Chen H and Wang S: Suppression of microRNA-130b inhibits glioma cell proliferation and invasion, and induces apoptosis by PTEN/AKT signaling. Int J Mol Med 41: 284-292, 2018
APA
Gu, J., Fan, K., Zhang, J., Chen, H., & Wang, S. (2018). Suppression of microRNA-130b inhibits glioma cell proliferation and invasion, and induces apoptosis by PTEN/AKT signaling. International Journal of Molecular Medicine, 41, 284-292. https://doi.org/10.3892/ijmm.2017.3233
MLA
Gu, J., Fan, K., Zhang, J., Chen, H., Wang, S."Suppression of microRNA-130b inhibits glioma cell proliferation and invasion, and induces apoptosis by PTEN/AKT signaling". International Journal of Molecular Medicine 41.1 (2018): 284-292.
Chicago
Gu, J., Fan, K., Zhang, J., Chen, H., Wang, S."Suppression of microRNA-130b inhibits glioma cell proliferation and invasion, and induces apoptosis by PTEN/AKT signaling". International Journal of Molecular Medicine 41, no. 1 (2018): 284-292. https://doi.org/10.3892/ijmm.2017.3233