Growth-inhibitory and chemosensitizing effects of microRNA-31 in human glioblastoma multiforme cells

  • Authors:
    • Rong-Jing Zhou
    • Xiong-Ying Xu
    • Bu-Xing Liu
    • Wen-Zhen Dai
    • Mei-Qin Cai
    • Chun-Feng Bai
    • Xian-Fei Zhang
    • Li-Min Wang
    • Li Lin
    • Shu-Zhen Jia
    • Wen-Hua Wang
  • View Affiliations

  • Published online on: August 13, 2015     https://doi.org/10.3892/ijmm.2015.2312
  • Pages: 1159-1164
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The constitutive activation of signal transducer and activator of transcription 3 (STAT3) contributes to resistance to temozolomide (TMZ) in glioblastoma multiforme (GBM). The aim of this study was to explore the biological role of microRNA-31 (miR-31) in GBM, particularly its role in the regulation of TMZ chemosensitivity. For this purpose, the human GBM cell lines, U251 and U87, were transfected with a miR-31 precursor (pre-miR-31), and cell proliferation, apoptosis and STAT3 phosphorylation were then assessed. To evaluate the effects of miR-31 on TMZ cytotoxicity, the cells were transfected with pre-miR-31 and exposed to 100 µM TMZ for 72 h prior to cell proliferation and apoptosis analysis. A constitutively active STAT3 mutant was co-transfected with pre-miR-31 into the cells to confirm the mediating role of STAT3 signaling. The enforced expression of miR-31 significantly reduced cell proliferation and induced mitochondrial apoptosis, as manifested by the loss of mitochondrial membrane potential and the increase in caspase-9 and caspase-3 activity. The phosphorylation level of STAT3 was significantly decreased by the overexpression of miR-31. The co-delivery of the constitutively active STAT3 mutant blocked the tumor suppressive effects of miR-31. In addition, miR-31 overexpression significantly enhanced the cytotoxic effects of TMZ on the GBM cells, as evidenced by the accelerated suppression of cell proliferation and the induction of apoptosis. The chemosensitizing effects of miR-31 were significantly impaired by the expression of the constitutively active STAT3 mutant. Taken together, our results indicate that miR-31 triggers mitochondrial apoptosis and potentiates TMZ cytotoxicity in GBM cells largely through the suppression of STAT3 activation. Thus, the restoration of miR-31 expression may be of therapeutic beenefit in the treatment of GBM.

Introduction

Glioblastoma multiforme (GBM) is the most common and lethal primary malignancy of the central nervous system, and patients with GBM have a poor prognosis (1). Current standard therapies for GBM comprise surgical resection, chemotherapy and radiotherapy (2). Temozolomide (TMZ), a DNA methylating agent, is the primary chemotherapeutic drug used in the treatment of malignant gliomas (3). Clinical studies have indicated that TMZ chemotherapy alone or in combination with radiotherapy increases the survival rate of patients with GBM (4,5). Despite advances in treatment strategies, the median survival rate of patients with GBM is only 12–14 months (6).

Chemoresistance is a major obstacle to effective cancer chemotherapy. TMZ induces the formation of O6-methylguanine in DNA, which consequently leads to DNA replication defects and cell death. A well-established mechanism for resistance to TMZ is mediated by the DNA repair protein O6-methylguanine methyltransferase (MGMT), which repairs TMZ-induced DNA lesions through the removal of the O6-methyl group (7). Additionally, the aberrant activation of survival signaling pathways also contributes to the resistance of GBM cells to TMZ (8). Signal transducer and activator of transcription 3 (STAT3) signaling has been implicated in the development and progression of many types of tumor, including GBM (9). The constitutive activation of STAT3 has been shown to contribute to the resistance of GBM cells to TMZ (10), and thus STAT3 represents an important therapeutic target for this disease.

MicroRNAs (miRs or miRNAs) are a class of endogenous, non-coding regulatory RNAs of ~22 nucleotides in length. They regulate a large number of target genes and thus affect various biological processes, such as cell proliferation, differentiation, apoptosis, invasion and tumorigenesis (11,12). miR-31 is dysregulated in many human malignancies, such as bladder cancer (13), melanoma (14) and colorectal cancer (15). Compared to normal brain tissue, miR-31 expression is significantly decreased in glioma tissue (16). miR-31 functions as an oncogene or a tumor suppressor in different types of cancer (1415,17). For instance, miR-31 has been shown to enhance the proliferation of colon cancer cells (17), whereas in melanoma cells, miR-31 has been shown to inhibit cell migration and invasion (14). Despite these studies, relatively little is known about the biological role of miR-31 in GBM, particularly in relation to the regulation of chemosensitivity.

In this study, we investigated the effects of miR-31 overexpression on the proliferation, apoptosis and TMZ sensitivity of GBM cells, and we also examined the involvement of STAT3 signaling.

Materials and methods

Cell culture

Normal human astrocytes were purchased from ScienCell Research Laboratories (Carlsbad, CA, USA) and the human GBM cell lines, U251 and U87, were obtained from the American Type Culture Collection (ATCC; Rockville, MD, USA). The cells were maintained in Dulbecco's modified Eagle's medium (DMEM) with 25 mmol/l D-glucose (Invitrogen, Carlsbad, CA, USA) supplemented with 10% heat-inactivated fetal bovine serum, 2 mM glutamine and penicillin (100 U/ml)-streptomycin (100 μg/ml; all from Invitrogen).

Measurement of miR-31 expression

miR-31 expression was measured by reverse transcription-quantitative polymerase chain reaction (RT-qPCR), as previously described (18). In brief, total RNA was extracted from the cells using TRIzol reagent (Invitrogen), and cDNA was synthesized with specific stem-loop reverse transcription primers. miR-31 expression levels were measured using TaqMan microRNA assays (Taqman® MicroRNA Reverse Transcription kit; Applied Biosystems, Foster City, CA, USA) with the following cycling conditions: 95°C for 5 min, followed by 40 cycles of amplification (95°C for 20 sec, 60°C for 20 sec and 72°C for 30 sec). The relative level of miR-31 following normalization to U6 small nuclear RNA was analyzed using the comparative cycle threshold (ΔΔCt) method, as previously described (19).

Plasmids, miRNA oligonucleotides and cell transfection

The STAT3-C plasmid expressing the constitutively active STAT3 mutant was purchased from Addgene (Cambridge, MA, USA), as previously described (20). The pcDNA 3.1(+) control vector was purchased from Invitrogen. Human pre-miR-31 and pre-miR negative control oligonucleotides were purchased from GenePharma Corp. (Shanghai, China). Cell transfection was carried out using Lipofectamine 2000 (Invitrogen) according to the manufacturer's instructions. Untransfected cells were used as the controls. The final concentration of each miRNA oligonucleotide was 50 nM. To determine the mediating role of STAT3, 1 μg of STAT3-C or the control plasmid was co-transfected into the GBM cells. At 24 h post-transfection, the cells were subjected to cell proliferation, apoptosis and gene expression analyses.

Treatment with TMZ

To determine the effects of miR-31 on TMZ cytotoxicity, the cells were transfected with pre-miR-31 or co-transfected with pre-miR-31 and STAT3-C 24 h prior to exposure to 100 μM TMZ, as previously described (21). Following incubation for an additional 72 h, the cells were harvested for cell proliferation and apoptosis assays.

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay

The cells were seeded in 96-well microplates at a density of 4×103 cells/well. Follwoing treatment, the cells were subjected to viability analysis using an MTT assay. MTT solution (5 mg/ml; Sigma, St. Louis, MO, USA) was added to each well followed by incubation at 37°C for 4 h. Dimethyl sulfoxide (DMSO) was added to dissolve the formazan crystals. The absorbance was then measured at 570 nm using a a spectrophotometer (DU-640; Beckman Coulter, Hialeah, FL, USA).

Apoptosis assay

Following treatment, the cells were harvested by trypsinization, and apoptosis was detected using the Annexin V apoptosis kit (Becton-Dickinson Biosciences, San Diego, CA, USA). The cells were stained with fluorescein isothiocyanate-conjugated Annexin V and propidium iodide (PI) solution (20 μg/ml) for 15 min in the dark. Apoptotic cells (Annexin V-positive) were analyzed by flow cytometry (Becton-Dickinson Biosciences).

Measurement of mitochondrial membrane potential (ΔΨm)

Changes in the ΔΨm were measured using the JC-1 mitochondrial membrane potential assay kit (Beyotime, Nantong, China). JC-1 forms monomers that emit green fluorescence and JC-1 aggregates are marked by red fluorescence. The ratio of JC-1 green to red fluorescence is proportional to the strength of ΔΨm. At 72 h post-transfection, cells were collected by trypsinization and pelleted. The cells were resuspended in JC-1 working solution and incubated at 37°C for 15 min. After washing, the stained cells were analyzed by flow cytometry (Becton-Dickinson Biosciences).

Measurement of caspase-3 and caspase-9 activity

The activity of caspase-3 and caspase-9 was measured using the Caspase-3/9 Activity assay kit (Beyotime), according to the manufacturer's instructions. Briefly,the transfected cells were lysed and the lysates were incubated with reaction buffer, which contained the fluorescent substrates, N-acetyl-Asp-Glu-Val-Asp-p-nitroanilide (Ac-DEVD-pNA) and N-acetyl-Leu-Glu-His-Asp-p-nitroanilide (Ac-LEHD-pNA), for caspase-3 and caspase-9, respectively. The fluorescence of the cleaved substrates was determined using an SLM 8000 fluorometer (SLM-Aminco, Urbana, IL, USA) at 405 nm.

Western blot analysis

For western blot analysis, the following primary antibodies were used: rabbit anti-total STAT3 (sc-482), anti-phosphorylated (p-)STAT3 (sc-8001-R), anti-cyclin D1 (sc-753), anti-Mcl-1 (sc-819) and anti-survivin (sc-10811) polyclonal antibodies (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) and mouse anti-β-actin (A5316) monoclonal antibody (Sigma). Horseradish peroxidase (HRP)-conjugated goat anti-rabbit (#31460) or anti-mouse (#31430) IgGs were purchased from Pierce Biotechnology (Rockford, IL, USA). Following treatment, the cells were lysed in RIPA buffer (Sigma) containing 1 mM phenylmethanesulfonyl fluoride and complete protease inhibitors (Roche, Mannheim Germany). The protein concentrations in cellular lysates were measured using a BCA Protein assay kit (Thermo Scientific, Rockford, IL, USA). The protein samples were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred onto polyvinylidene fluoride membranes. The membranes were then blocked with 5% fat-free milk and incubated with the primary antibodies at 4°C overnight, followed by incubation with HRP-conjugated secondary antibodies for 1 h. Immunoreactive bands were visualized using an enhanced chemiluminescence detection system (Amersham Biosciences, Piscataway, NJ, USA) and quantified using Quantity One software (Bio-Rad, Hercules, CA, USA).

Statistical analysis

Data are expressed as the means ± standard deviation (SD). Differences between groups were examined by one-way analysis of variance using the SPSS software package v19.0 (SPSS, Inc., Chicago, IL, USA). A P-value <0.05 was considered to indicate a statistically significantly difference.

Results

Overexpression of miR-31 inhibits the viability of GBM cells

RT-qPCR revealed that miR-31 expression was significantly decreased (P<0.05) in the U87 and U251 cells, compared to normal human astrocytes (Fig. 1A). The effects of restoring miR-31 expression on the viability of GBM cells was then investigated. As shown in Fig. 1B, miR-31-overexpressing U87 and U251 cells had a significantly lower viability (P<0.05) compared to the control (untransfected) or negative miRNA-transfected cells.

Overexpression of miR-31 significantly increases the apoptosis of GBM cells through the mitochondrial pathway

Subsequently, we examined the effects of miR-31 overexpression on the survival of GBM cells. Apoptosis detection by Annexin V/PI staining demonstrated that the enforced expression of miR-31 induced a significant increase (P<0.05) in the apoptosis of the U87 and U251 cells (Fig. 2A). As the depolarization of ΔΨm is an early event in mitochondrial-related apoptosis, we measured the changes in ΔΨm induced by miR-31 overexpression using the JC-1 dye. As shown in Fig. 2B, miR-31 overexpression resulted in a significant decrease (P<0.05) in the red/green fluorescence ratio, which was indicative of the loss of ΔΨm. Enzyme activity assay revealed that there was a 2.4- and 2.8-fold increase in caspase-9 activity and an 8.6- and 12.4-fold increase in caspase-3 activity in miR-31-overexpressing U87 and U251 cells, respectively (Fig. 2C and D).

Restoration of miR-31 interferes with the activation of STAT3 signaling in GBM cells

It is known that the constitutive activation of STAT3 signaling contributes to GBM growth and survival (9). Thus, we examined the effects of restoring miR-31 expression on the activation of STAT3 signaling. As shown in Fig. 3A, in the miR-31-overexpressing U87 and U251 cells, a significant decrease (P<0.05) in the phosphorylation of STAT3 was noted, compared to the controls and negative miRNA-transfected cells. We also examined the effects of restoring miR-31expression on the expression of target genes of STAT3, including cyclin D1, Mcl-1 and survivin. Western blot analysis demonstrated that miR-31 overexpression markedly inhibited the expression of cyclin D1, Mcl-1 and survivin in the U87 and U251 cells (Fig. 3B).

miR-31 enhances the efficacy of TMZ in GBM cells

We then investigated the effects of miR-31 overexpression on the sensitivity of GBM cells to TMZ. MTT assay revealed that TMZ at 100 μM induced ~20% decrease in the viability of the U87 and U251 cells after a 72-h incubation (Fig. 4A). Notably, the delivery of miR-31 significantly enhanced (P<0.05) the cytotoxic effects of TMZ on GBM cells, leading to a 60–70% decrease in cell viability (Fig. 4A). Similarly, the overexpression of miR-31 significantly increased (P<0.05) the apoptosis of the TMZ-treated GBM cells (Fig. 4B).

Chemosensitizing effect of miR-31 is mediated through the inactivation of STAT3

Having concluded that STAT3 signaling was suppressed by miR-31 overexpression, we examined the involvement of STAT3 signaling in miR-31-induced chemosensitization. Co-transfection with a constitutively active STAT3 mutant markedly enhanced the phosphorylation of STAT3 in the miR-31-transfected GBM cells (Fig. 5A). Of note, the constitutive activation of STAT3 significantly reversed (P<0.05) the enhancement of TMZ cytotoxicity which was achieved by miR-31 overexpression (Fig. 5B).

Discussion

Certain miRs are deregulated in GBM and have been implicated in tumor growth and survival (22,23). Yue et al (23) demonstrated that miR-205 was significantly downregulated in human glioblastoma cells and that the restoration of its expression induced apoptosis and cell cycle arrest in glioma cells. Zhang et al (24) reported that miR-195 plays a tumor-suppressor role in human glioblastoma cells, impairing cell cycle progression and cellular invasion. Similarly, our data demonstrated that miR-31 functions as a tumor suppressor in GBM, as evidenced by the observation that the ectopic expression of miR-31 significantly suppressed GBM cell proliferation and induced apoptosis. Our data further demonstrated that miR-31 induced the apoptosis of GBM cells through the mitochondrial cascade, which was manifested by the loss of ΔΨm and the activation of caspase-3 and capsase-9. The tumor-suppressive effects of miR-31 have also been documented in several other types of cancer, such as melanoma (14) and breast cancer (25). However, there is also evidence that miR-31 plays an oncogenic role in certain human tumors. For instance, Mitamura et al (26) reported that miR-31 facilitates the anchorage-independent growth and tumorigenesis of endometrial cancer cells. In non-small cell lung cancer cells, it was demonstrated that miR-31 has the ability to inhibit cisplatin-induced apoptosis (27). Therefore, these data suggest that miR-31 regulates tumor growth and survival in a cancer type-dependent manner.

The constitutive activation of Stat3STAT3 plays an important role in the development and progression of GBM (9). It has been reported that the nuclear factor-κB (NF-κB)-induced STAT3 activation contributes to aggressive phenotypes in GBM and that the inhibition of STAT3 signaling retards the growth of human GBM xenografts (28). The pharmacological inhibition of STAT3 signaling has been found to induce the apoptosis of human GBM cells (29). Of note, our data revealed that the overexpression of miR-31 impaired the activation of STAT3. Moreover, the ectopic expression of miR-31 markedly downregulated multiple STAT3 target genes, including cyclin D1, Mcl-1 and survivin in GBM cells. The downregulation of cyclin D1 is linked to the reduced proliferation and enhanced apoptosis of glioblastoma cells (30). Mcl-1 and survivin are two key pro-survival proteins, and their inhibition contributes to the apoptosis of GBM cells through the mitochondrial death pathway (31). These studies, combined with our findings, suggest that STAT3 signaling is involved in the tumor-suppressive activity of miR-31 in GBM.

Previous research corroborates the importance of STAT3 signaling in the development of resistance to TMZ in GBM (32). Kohsaka et al (10) reported that the inhibition of STAT3 signaling can helpt overcome resistance to TMZ in glioblastoma. Given our knowledge of the regulation of STAT3 activation by miR-31, in this study, we examined the effects of miR-31 on TMZ chemosensitivity in GBM cells. Notably, we found that miR-31 overexpression significantly enhanced TMZ cytotoxicity to GBM cells. Moreover, the enforced expression of miR-31 enhanced the apoptosis of GBM cells in the presence of TMZ. The miR-31-mediated chemosensitization to TMZ was reversed by co-transfection with a constitutively active STAT3 mutant. Taken together, these data suggest that the restoration of miR-31 expression sensitizes GBM cells to TMZ largely through the suppression of STAT3 activation. The chemosensitizing activity of miR-31 has also been described in relation to ovarian cancer cells, where the re-introduction of miR31 was shwn to re-sensitize paclitaxel-resistant cells to this agent (33).

In conclusion, our data confirm that miR-31 functions as a tumor suppressor gene in GBM. The restoration of miR-31 expression induced the apoptosis of and enhanced TMZ cytotoxicity in human GBM cells, which was mediated through the suppression of STAT3 activation. The re-expression of miR-31 may thus represent a promising strategy for improving the efficacy of TMZ in GBM.

References

1 

Wilson TA, Karajannis MA and Harter DH: Glioblastoma multiforme: State of the art and future therapeutics. Surg Neurol Int. 5:642014. View Article : Google Scholar : PubMed/NCBI

2 

Alifieris C and Trafalis DT: Glioblastoma multiforme: Pathogenesis and treatment. Pharmacol Ther. 152:63–82. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Hart MG, Garside R, Rogers G, Stein K and Grant R: Temozolomide for high grade glioma. Cochrane Database Syst Rev. 4:CD0074152013.PubMed/NCBI

4 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Hau P, Koch D, Hundsberger T, Marg E, Bauer B, Rudolph R, Rauch M, Brenner A, Rieckmann P, Schuth J, et al: Safety and feasibility of long-term temozolomide treatment in patients with high-grade glioma. Neurology. 68:688–690. 2007. View Article : Google Scholar : PubMed/NCBI

6 

do Carmo A, Balça-Silva J, Matias D and Lopes MC: PKC signaling in glioblastoma. Cancer Biol Ther. 14:287–294. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, et al: MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 352:997–1003. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Fan QW and Weiss WA: Targeting the RTK-PI3K-mTOR axis in malignant glioma: overcoming resistance. Curr Top Microbiol Immunol. 347:279–296. 2010.PubMed/NCBI

9 

Luwor RB, Stylli SS and Kaye AH: The role of Stat3 in glioblastoma multiforme. J Clin Neurosci. 20:907–911. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Kohsaka S, Wang L, Yachi K, Mahabir R, Narita T, Itoh T, Tanino M, Kimura T, Nishihara H and Tanaka S: STAT3 inhibition overcomes temozolomide resistance in glioblastoma by downregulating MGMT expression. Mol Cancer Ther. 11:1289–1299. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Fabbri M: MicroRNAs and cancer: towards a personalized medicine. Curr Mol Med. 13:751–756. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Williams AE: Functional aspects of animal microRNAs. Cell Mol Life Sci. 65:545–562. 2008. View Article : Google Scholar

13 

Wang S, Li Q, Wang K, Dai Y, Yang J, Xue S, Han F, Zhang Q, Liu J and Wu W: Decreased expression of microRNA-31 associates with aggressive tumor progression and poor prognosis in patients with bladder cancer. Clin Transl Oncol. 15:849–854. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Asangani IA, Harms PW, Dodson L, Pandhi M, Kunju LP, Maher CA, Fullen DR, Johnson TM, Giordano TJ, Palanisamy N and Chinnaiyan AM: Genetic and epigenetic loss of microRNA-31 leads to feed-forward expression of EZH2 in melanoma. Oncotarget. 3:1011–1025. 2012.PubMed/NCBI

15 

Nosho K, Igarashi H, Nojima M, Ito M, Maruyama R, Yoshii S, Naito T, Sukawa Y, Mikami M, Sumioka W, et al: Association of microRNA-31 with BRAF mutation, colorectal cancer survival and serrated pathway. Carcinogenesis. 35:776–783. 2014. View Article : Google Scholar

16 

Wang S, Jiao B, Geng S, Song J, Liang Z and Lu S: Concomitant microRNA-31 downregulation and radixin upregulation predicts advanced tumor progression and unfavorable prognosis in patients with gliomas. J Neurol Sci. 338:71–76. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Li T, Luo W, Liu K, Lv X and Xi T: miR-31 promotes proliferation of colon cancer cells by targeting E2F2. Biotechnol Lett. 37:523–532. 2015. View Article : Google Scholar

18 

Huang S, Wu B, Li D, Zhou W, Deng G, Zhang K and Li Y: Knockdown of astrocyte elevated gene-1 inhibits tumor growth and modifies microRNAs expression profiles in human colorectal cancer cells. Biochem Biophys Res Commun. 444:338–345. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−ΔΔC(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

20 

Pilati C, Amessou M, Bihl MP, Balabaud C, Nhieu JT, Paradis V, Nault JC, Izard T, Bioulac-Sage P, Couchy G, et al: Somatic mutations activating STAT3 in human inflammatory hepatocellular adenomas. J Exp Med. 208:1359–1366. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Filippi-Chiela EC, Thomé MP, Bueno e Silva MM, Pelegrini AL, Ledur PF, Garicochea B, Zamin LL and Lenz G: Resveratrol abrogates the temozolomide-induced G2 arrest leading to mitotic catastrophe and reinforces the temozolomide-induced senescence in glioma cells. BMC Cancer. 13:1472013. View Article : Google Scholar : PubMed/NCBI

22 

Chen L, Wang X, Wang H, Li Y, Yan W, Han L, Zhang K, Zhang J, Wang Y, Feng Y, et al: miR-137 is frequently downregulated in glioblastoma and is a negative regulator of Cox-2. Eur J Cancer. 48:3104–3111. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Yue X, Wang P, Xu J, Zhu Y, Sun G, Pang Q and Tao R: MicroRNA-205 functions as a tumor suppressor in human glioblastoma cells by targeting VEGF-A. Oncol Rep. 27:1200–1206. 2012.

24 

Zhang QQ, Xu H, Huang MB, Ma LM, Huang QJ, Yao Q, Zhou H and Qu LH: MicroRNA-195 plays a tumor-suppressor role in human glioblastoma cells by targeting signaling pathways involved in cellular proliferation and invasion. Neuro Oncol. 14:278–287. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Körner C, Keklikoglou I, Bender C, Wörner A, Münstermann E and Wiemann S: MicroRNA-31 sensitizes human breast cells to apoptosis by direct targeting of protein kinase C epsilon (PKCepsilon). J Biol Chem. 288:8750–8761. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Mitamura T, Watari H, Wang L, Kanno H, Kitagawa M, Hassan MK, Kimura T, Tanino M, Nishihara H, Tanaka S and Sakuragi N: microRNA 31 functions as an endometrial cancer oncogene by suppressing Hippo tumor suppressor pathway. Mol Cancer. 13:972014. View Article : Google Scholar : PubMed/NCBI

27 

Dong Z, Zhong Z, Yang L, Wang S and Gong Z: MicroRNA-31 inhibits cisplatin-induced apoptosis in non-small cell lung cancer cells by regulating the drug transporter ABCB9. Cancer Lett. 343:249–257. 2014. View Article : Google Scholar

28 

McFarland BC, Hong SW, Rajbhandari R, Twitty GB Jr, Gray GK, Yu H, Benveniste EN and Nozell SE: NF-κB-induced IL-6 ensures STAT3 activation and tumor aggressiveness in glioblastoma. PLoS One. 8:e787282013. View Article : Google Scholar

29 

Swiatek-Machado K, Mieczkowski J, Ellert-Miklaszewska A, Swierk P, Fokt I, Szymanski S, Skora S, Szeja W, Grynkiewicz G, Lesyng B, et al: Novel small molecular inhibitors disrupt the JAK/STAT3 and FAK signaling pathways and exhibit a potent antitumor activity in glioma cells. Cancer Biol Ther. 13:657–670. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Wang J, Wang Q, Cui Y, Liu ZY, Zhao W, Wang CL, Dong Y, Hou L, Hu G, Luo C, et al: Knockdown of cyclin D1 inhibits proliferation, induces apoptosis, and attenuates the invasive capacity of human glioblastoma cells. J Neurooncol. 106:473–484. 2012. View Article : Google Scholar

31 

Premkumar DR, Jane EP, Foster KA and Pollack IF: Survivin inhibitor YM-155 sensitizes tumor necrosis factor- related apoptosis-inducing ligand-resistant glioma cells to apoptosis through Mcl-1 downregulation and by engaging the mitochondrial death pathway. J Pharmacol Exp Ther. 346:201–210. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Lo HW, Cao X, Zhu H and Ali-Osman F: Constitutively activated STAT3 frequently coexpresses with epidermal growth factor receptor in high-grade gliomas and targeting STAT3 sensitizes them to Iressa and alkylators. Clin Cancer Res. 14:6042–6054. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Mitamura T, Watari H, Wang L, Kanno H, Hassan MK, Miyazaki M, Katoh Y, Kimura T, Tanino M, Nishihara H, et al: Downregulation of miRNA-31 induces taxane resistance in ovarian cancer cells through increase of receptor tyrosine kinase MET. Oncogenesis. 2:e402013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2015
Volume 36 Issue 4

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
Zhou R, Xu X, Liu B, Dai W, Cai M, Bai C, Zhang X, Wang L, Lin L, Jia S, Jia S, et al: Growth-inhibitory and chemosensitizing effects of microRNA-31 in human glioblastoma multiforme cells. Int J Mol Med 36: 1159-1164, 2015
APA
Zhou, R., Xu, X., Liu, B., Dai, W., Cai, M., Bai, C. ... Wang, W. (2015). Growth-inhibitory and chemosensitizing effects of microRNA-31 in human glioblastoma multiforme cells. International Journal of Molecular Medicine, 36, 1159-1164. https://doi.org/10.3892/ijmm.2015.2312
MLA
Zhou, R., Xu, X., Liu, B., Dai, W., Cai, M., Bai, C., Zhang, X., Wang, L., Lin, L., Jia, S., Wang, W."Growth-inhibitory and chemosensitizing effects of microRNA-31 in human glioblastoma multiforme cells". International Journal of Molecular Medicine 36.4 (2015): 1159-1164.
Chicago
Zhou, R., Xu, X., Liu, B., Dai, W., Cai, M., Bai, C., Zhang, X., Wang, L., Lin, L., Jia, S., Wang, W."Growth-inhibitory and chemosensitizing effects of microRNA-31 in human glioblastoma multiforme cells". International Journal of Molecular Medicine 36, no. 4 (2015): 1159-1164. https://doi.org/10.3892/ijmm.2015.2312