Downregulation of miR-95-3p inhibits proliferation, and invasion promoting apoptosis of glioma cells by targeting CELF2

  • Authors:
    • Bo Fan
    • Bao-Hua Jiao
    • Feng-Shi Fan
    • Sheng-Kui Lu
    • Jian Song
    • Cheng-Yong Guo
    • Jian-Kai Yang
    • Liang Yang
  • View Affiliations

  • Published online on: July 10, 2015     https://doi.org/10.3892/ijo.2015.3080
  • Pages: 1025-1033
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gliomas are the most common and aggressive types of tumors in human brain, of which the prognosis remains dismal because of their biological behavior. The involvement of miRNAs in tumorigenesis of various kinds of cancers drives us to explore new miRNAs related to gliomas. We measured expression level of miR‑95‑3p by qRT-PCR in human glioma and non-neoplasm brain tissues and found that higher level of miR‑95‑3p in glioma tissues of higher grade. Biological functions of miR‑95‑3p on glioma cells were investigated by MTT assay, flow cytometry and transwell assay. We discovered the cell lines transfected with miR‑95‑3p ASO (antisense oligonucleotide) had retarded proliferation and invasion but enhanced apoptosis ability. We searched on-line tool Targetscan and selected CELF (CUGBP- and ETR-3-like family 2) as a putative target. Luciferase reporter was employed to confirm the binding sites in 3'UTR region of CELF2 for miR‑95‑3p. The correlation between expression of CELF2 and miR‑95‑3p was determined by western blotting and qRT-PCR both in cell lines and human samples. Results showed CELF2 was a direct target of miR‑95‑3p and expression levels of CELF2 and miR‑95‑3p were negatively correlated. Finally, CELF2 largely abrogated the effects of miR‑95‑3p on proliferation, invasion and apoptosis of glioma cells in rescue experiments, which verified the role of CELF2 in miR‑95‑3p regulating glioma biological behavior. In conclusion, our data suggest the expression level of miR‑95‑3p is positively related to glioma grade and downregulation of miR‑95‑3p affects proliferation, invasion and apoptosis of glioma cells by targeting CELF2. We identified miR‑95‑3p as a putative therapeutic target and CELF2 as a potential tumor suppressor.

Introduction

Gliomas are the most common and aggressive types of tumors in central nervous system. It has been proved that dozens of molecules and signal pathways are involved in the genetic alterations and chromosome aberrations during tumorigenesis. The abnormal abilities of invasion, proliferation, migration and apoptosis of glioma cells are the main obstacles to better prognosis (13). Despite the achievements in conventional treatments, the median survival period of patients suffering from glioblastoma multiforme (GBM), a type of grade IV glioma, remains at 12 months over the past few decades (4). Hence, discovering new therapeutic targets and developing mechanism-based approaches is an urgent need.

MicroRNAs (miRNA or miR) are a group of endogenous non-coding small RNAs which are expressed in the vast majority of eukaryotes (5). They cause gene silencing by targeting specifically 3′-untranslated regions (3′UTR) of mRNAs, leading to the degradation or repression of mRNA translation (6,7). Some miRNA precursors can form two stably expressing mature miRNAs, which are distinguished by adding ‘−3p' or ‘−5p' to the end of the name according to their procession positions. Emerging evidence has strongly indicated that this post-transcriptional regulation by miRNAs has major implications in many physiological and pathological processes, and is associated with the initiation and progression of human cancer, including glioma (811). Some miRNAs are proved to be crucial oncogenes or tumor suppressors of glioma (1214).

miR-21, miR-10b, miR-93 and miR-155 are upregulated in gliomas and have been shown to be significant contributors in tumor growth (13,1517); while miR-7, miR-124, miR-128 and miR-137 are downregulated with tumor suppressor activities (16,1820). Diverse expression profiles between non-neoplasm and glioma tissues can partly elucidate the functions of miRNAs in tumorigenesis and therefore can be utilized to predict novel therapeutic targets. We noticed that miR-95 was reported to have different expression profiles between glioma tissues and non-neoplasm brain tissues by sequencing analysis (21). The results of functional research on miR-95 seem to be contradictory, as miR-95 promotes the growth of colorectal, pancreatic, prostate and breast cancer (2224), while has anticancer activity in hepatic, brain and neck cancer (25,26). However, miR-95 has not been previously reported in gliomas. So here we recruited miR-95-3p as a candidate to explore its biological function in glioma.

In this study, we first compared the expression level of miR-95-3p in glioma and non-neoplasm brain tissues by qRT-PCR, and evaluated its influence on the proliferation, invasion, cell cycle and apoptosis of glioma in vitro. We discovered the reverse relationship between CELF2 and miR-95-3p expression both in cell lines and human samples, and utilized luciferase reporter assay to confirm that CELF2 was a novel target of miR-95-3p. Rescue experiment showed that downregulation of CELF2 could largely abolish the effect induced by repression of miR-95-3p. Taken together, we conclude that miR-95-3p affects proliferation, apoptosis and invasion of glioma cells by targeting CELF2, and we identified miR-95-3p as a putative therapeutic target and CELF2 as a potential tumor suppressor.

Materials and methods

Clinical sample collection

Human glioma samples and non-neoplasm brain samples were collected from the Neurosurgery Department of the Second Affiliated Hospital of Hebei Medical University in 2014. Fifteen females and 35 males were included, with ages ranging from 30 to 60. Thirty-five tumor samples, including 4 grade I, 5 grade II glioma, 17 grade III and 9 grade IV tissues (GBM), were obtained by surgical resection, and 5 non-neoplasma samples as control were obtained by intracranial decompression for severe traumatic brain injury. Diagnoses were established histologically by two pathologists according to the WHO classification guidelines. All samples were stored at −80°C in liquid nitrogen immediately after resection. Written informed consent was obtained from all patients and the study was approved by the ethics committee of The Second Affiliated Hospital of Hebei Medical University.

Cell culture and transfection

Human glioma cell lines U87 and U251 were obtained from the American Type Culture Collection (Manassas, VA, USA) and were maintained in α-MEM (Gibco BRL, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 μg/ml streptomycin at 37°C with 5% CO2. The miR-95-3p ASO, control ASO, CELF2 siRNA and control siRNA were synthesized and purified by GenePharma, Shanghai, China. The RNA sequences mentioned above were as follows: miR-95-3p ASO: 5′-TGCTCAATAAATACCCGTTGAA-3′; Ctrl ASO: 5′-TTATCGCCATGTCCAATGAGGCT-3′; CUGBP2 siRNA: 5′-GCAAACCUUACUGAUCCUA-3′; Ctrl siRNA: 5′-UAAGGCUAUGAAGAGAUAC-3′.

The control RNAs contain random sequences and are predicted not to have any interactions in cells. Cell transfection was performed using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions. To transfect RNA oligonucleotides, 10 pmol of miRNA antisense oligonucleotides and ctrl ASO were used. For rescue experiments, cells were contransfected with 10 pmol of miRNA ASO and siRNA or control siRNA in a 96-well plate.

RNA extraction and real-time quantitative RT-PCR

Total cell RNA was extracted using TRIzol reagent (Invitrogen) according to the manufacturer's instructions. miRNA level was measured by qRT-PCR. All the primers of miRNAs for TaqMan miRNA assays were purchased from Genewiz (Beijing, China). Human U6 small nuclear RNA was used as an internal normalized reference. The forward and reverse primers of miR-95-3p and U6 are as follows: miR-95-3p F/R, 5′-TGCGGTTCAACGGGTATTTATTG-3′/5′-CCAGTG CAGGGTCCGAGGT-3′; U6 F/R, 5′-TGCGGGTGCTCGCTT CGGCAGC-3′/5′-CCAGTGCAGGGTCCGAGGT-3′. RNA (5 μg) was mixed with 1 μl of MMLV reverse transcriptase (Promega, Madison, WI, USA) to synthesize complementary DNA according to the manufacturer's instructions. The expression of miR-95-3p in human samples was quantified using SYBR® Premix Ex Taq™ (Takara, Tokyo, Japan) in the ABI PRISM® 7500 real-time PCR system (Applied Biosystems, Foster City, CA, USA). Real-time PCR was performed according to the manufacturer's instructions. Melting curves were used to evaluate non-specific amplification. All experiments were performed using biological triplicates and experimental duplicates. The relative expression levels were evaluated via the 2−ΔΔCt method.

Western blotting

U87 or U251 cells were processed with RIPA lysis buffer (Saierbio, Tianjing, China) for 48 h before collecting protein samples. Total protein (32 μl) and 8 μl of loading buffer (5X) was boiled for 3 min and cooled down for 3 min on ice. Heat-denatured protein samples were resolved by 10% SDS-polyacrylamide gel (SDS-PAGE) electrophoresis under 100 V and transferred to hybond-nitrocellulose membranes (Millipore, USA) under 350 mA at 4°C. The membrane was blocked in Blotto solution for 2 h, and then incubated for 12 h at 4°C with primary rabbit anti-human monoclonal CELF2 antibody (1:1,000, Abcam, Cambridge, UK) overnight for combination. The GAPDH expression level was measured using rabbit anti-human monoclonal GAPDH antibody (1:3,000, Saier, Tianjing, China) as a loading control. Then the membrand was incubated for 2 h with HRP marked goat anti-rabbit second antibody (1:5,000, Saier, Tianjing, China). The bound antibody was detected with the use of Western Lightning®-ECL, Enhanced Chemiluminescence Substrate (Perkin-Elmer, NEL100001EA). The band density of CELF2 was quantified after normalization with the density of GAPDH.

MTT assay

U87 (1×104) or U251 cells were seeded into each well of a 96-well plate one day before transfection. Following a 48-h incubation after transfection, 20 μl of MTT reagent (5 mg/ml) was added into each well and the plate was incubated for another 4 h. The supernatant was removed, and 150 μl of DMSO was added into each well to dissolve the precipitate. The absorbance value was recorded at 570 nm using a microplate reader (BioTek, Winooski, VT, USA). All experiments were performed using biological triplicates and experimental duplicates, and the data are presented as mean ± SD.

Flow cytometry

The apoptosis analysis was performed with Annexin V/7-AAD apoptosis detection kit (KeyGen, Nanjing, China) according to the manufacturer's instructions. Briefly, 5×105 cells were planted into a 6-well plate and treated with 0.1% trypsin and 0.02% EDTA, and were washed twice with cold PBS before collection. The cells were mixed with 100 μl of 1X binding buffer and resuspended. Annexin V (10 μl) was added and incubated for 30 min. Then 380 μl of 1X binding buffer and 10 μl of 7-AAD was added sequently and fully mixed with the cells. The fluorescence values of Annexin V and 7-AAD were measured by flow cytometry (Beckman Coulter, CA, USA).

For cell cycle assay, 5×105 cells were planted into a 6-well plate, washed by 1X PBS once and treated with 0.1% trypsin and 0.02% EDTA for digestion. The cells were suspended in 1 ml culture medium and centrifuged under 12,000 × g for 5 min. The supernatant was discarded and 1 ml 1X PBS was added into the cells then centrifuged for 5 min. The cells were stained with PI for 30 min and then investigate by flow cytometry. All the procedures were conducted according to the manufacturer's instructions.

Cell invasion assay

Cell invasion ability was determined by Corning transwell insert chambers (Corning, USA) and BD BioCoat Matrigel (BD Biosciences, Bedford, MA, USA). U87 or U251 cells were harvested and trypsinized in serum-free medium after transfection. Then the cell suspension (200 μl, 1×105 cells) was added into the chamber and incubated 24 h at 37°C. Cells that invaded the Matrigel and passed through the filter were fixed with 20% methanol for 30 min, stained with crystal violet for 15 min, and then imaged under ×100 magnification Nikon 90I light microscope (Nikon, Japan) and counted in 6 randomized fields.

Vector construction

For overexpression of miR-95-3p, the human pre-miR-95 sequence was amplified and cloned into pcDNA3.1-hisA constructs (Invitrogen) to generate pcDNA3.1-miR-95 expression vector. The primers were miR-95-KL-F: CCGGAATTCGAAGGTAGGATTGTGA CAC and miR-95-KL-R: CGCGGATCCGGAGGGATGGAT GAATGAC.

For luciferase reporter assay, the 3′UTR of the human CELF2 gene containing the putative binding site was PCR amplified using the following primers: wt-CELF2-Fw: AAA CTAGCGG CCGCTAGTCTAGGAATGGGCTTTTTTTCACCGTTGAA CCCTAGACCTGT; wt-CELF2-Rv: CTAGACAGGTCTAGG GTTCAACGGTGAAAAAAAGCCCATTCCTAGACTAGC GGCCGCTAGTTT. 3′UTR of CELF2 gene containing the mutant binding site used the following primers: mut-CELF2-Fw: AAACTAGCGGCCGCTAGTCTAGGAATGGGCTTT TTTTCAGGCAACTACCCTAGACCTGT; mut-CELF2-Rv: CTAGACAGGTCTAGGGTAGTTGCCTGAAAAAAAGCC CATTCCTAGACTAGCGGCCGCTAGTTT. Fragments in CELF2 3′UTR containing the binding site (wt-CELF2) or mutated site (mut-CELF2) were, respectively, double-digested with XbaI/EcoRI and cloned downstream of firefly luciferase coding region sites of pmirGLO plasmid (Promega, Madison, WI, USA).

Luciferase reporter assay

The binding site was replaced with its complementary sequence to generate mutation. U251 cells were plated in a 96-well plate and then co-transfected with 0.25 μg of wt-CELF2 or mut-CELF2 expression vector and 10 pmol of miR-95-3p ASO or control ASO using Lipofectamine 2000 reagent. Cells were collected 48 h after transfection and analyzed using the Dual-Luciferase Reporter Assay System (Promega, Madison, WI, USA). Luciferase activity was detected by F200/M200 microplate fluorescence reader (Tecan Infinite). The co-transfected Renilla luciferase control vector was used as an internal control to correct the differences in both transfection and the harvesting efficiency. All the regents and instruments were performed according to the manufacturer's instructions. Transfection experiments were performed in duplicates and repeated at least thrice in independent experiments.

Statistical analysis

All tests and vector graphics were done using PRISM version 5.0 (GraphPad Software Inc., San Diego, CA, USA) statistical software. The experiments were carried out for three times independently. Data are mean ± standard deviation except when indicated otherwise. Spearman's rank correlation test was used for association analysis between miR-95-3p and CELF2 level data. Student's t-test was used to analyze the statistical significance between two groups. One-way ANOVA was used to compare multiple groups. P<0.05 was considered to be significant (shown in figures as: *P<0.05; **P<0.01;***P<0.001).

Results

miR-95-3p is significantly upregulated in human glioma tissues

To determine the role of miR-95-3p in tumorigenesis of human glioma, we compared the expression level of miR-95-3p in glioma samples and non-neoplasma brain samples. In this study, 35 tumor samples were used as experimental group and 5 non-neoplasma samples as control. Case number, age, gender, WHO grade and pathological report of each patient were recorded. The qRT-PCR results showed that miR-95-3p was significantly overexpressed in glioma tissues compared to control group (Fig. 1A). We also analyzed the correlation between expression level and pathological grade. One-way ANOVA test showed that the differences between non-neoplasm tissues, low grade glioma (grade I and II) and high grade glioma (grade III and IV) were significant, while the differences between grade I and II or between grade III and IV were not (Fig. 1B).

Downregulation of miR-95-3p manifests tumor-suppressive functions in glioma cell lines

To understand the function of miR-95-3p in glioma cells, expression of miR-95-3p was downregulated in U87 and U251 cell lines for a loss-of-function behavior. Antisense oligonucleotide (ASO) was introduced into the cells to repress miR-95-3p expression. Control cells were transfected with false ASO. Forty-eight hours later, miR-95-3p expression levels were examined by qRT-PCR. As shown in Fig. 2A, miR-95-3p was downregulated in U87 and U251 cells after transfection of ASO. Then we investigated the nature of the cells with different miR-95-3p levels.

MTT assay was performed to investigate the effect of miR-95-3p on cell proliferation. We found that transfection of U87 cells with miR-95-3p ASO significantly decreased the relative absorbance of MTT, which was consistant with that of U251 cells (Fig. 2B). The data indicated an anti-proliferation effect of downregulation of miR-95-3p on glioma cells. Then, we conducted flow cytometry to analyze the effect of miR-95-3p on the cell cycle and apoptosis. Our experiment failed to prove any difference in the cell cycle. The proportion of U87 or U251 cells in G1, S and G2 phase stayed almost unchanged after transfection with miR-95-3p ASO, compared to control group (Fig. 2C). As to apoptosis, U87 or U251 cells transfected with miR-95-3p ASO had a significant increase in number stained by Annexin V and/or 7-AAD, compared to cells with false ASO (Fig. 2D). The data showed that downregulation of miR-95-3p induces cell apoptosis. Finally, cell invasiveness was measured by transwell assay in order to evaluate metastasis potential. The cells were imaged and counted after 24 h of incubation in the transwell chamber. Our experiments showed less U87 or U251 cells transfected with miR-95-3p ASO invaded through the membrane than those with false ASO, which indicated downregulation of miR-95-3p decreased cell invasiveness (Fig. 2E). Taken together, the results manifested that downregulation of miR-95-3p has therapeutic effects on glioma proliferation, invasion and apoptosis.

CELF2 is a bona fide target of miR-95-3p

To delineate the mechanism by which miR-95-3p regulates the biological behavior of glioma cells, we searched the online program Targetscan for the target. Among all the protein candidates predicted, we finally recruited CELF2 (NM_001025077) as a putative target and identified one potential binding site for miR-95-3p (Fig. 3A and B). To confirm that CELF2 is a bona fide target of miR-95-3p, luciferase reporter assays were performed. Fig. 3C shows the mode pattern of pmirGLO vector. We demonstrated the fact that miR-95-3p ASO could significantly increase luciferase activity, which could be abrogated by the mutation of the binding site (Fig. 3D).

We explored the expression profiles of CELF2 and miR-95-3p in 7 human samples. Each piece of sample used in this experiment for western blot assay came from the same tissue with that for qRT-PTR to make their results comparable (Fig. 4A and B). The results supported a negative correlation between CELF2 and miR-95-3p expression level in human tissues (Fig. 4C). We modified the level of miR-95-3p in U251 cells by tansfecting with ASO or expression vector, to investigate the effect of miR-95-3p on CELF2 in vitro. Compared to control cells, CELF2 was downregulated in cells overexpressing miR-95-3p while upregulated in cells low-expressing miR-95-3p (Fig. 4D). Based on the above, we proved that miR-95-3p suppresses CELF2 gene expression by regulating 3′UTR at the post-transcription level.

CELF2 rescues the effect of miR-95-3p on glioma cells

To determine whether miR-95-3p regulates glioma cell biological behavior by targeting CELF2, CELF2 siRNA was used to perform rescue experiments. The downregulation of CELF2 by siRNA was validated by western blotting (Fig. 5A). The results showed that U251 cells transfected with miR-95-3p ASO plus CELF2 siRNA were increased in cell proliferation and invasion while decreased in apoptosis, compared to control (Fig. 5B–D). The data further suggest that CELF2 is a functional downstream target of miR-95-3p in regulating glioma biological behavior.

Discussion

Given the fact that cancer cells develop extraordinary characteristics which make them refractory to therapies, it is crucial to understand the underlying molecular biological mechanism. Evidence has shown miRNAs are implicated in many pathological and physiological processes, and we focused on miRNAs to explore their interaction with gliomas. The functions of miR-95-3p has been studied previously. Huang et al (22) found miR-95 could promote proliferation in human colorectal carcinoma cells. It has also been pointed out that miR-95 overexpression increased tumor growth and resistance to radiation treatment in prostate cancer cells (23). Our preliminary test showed that the expression difference of miR-95 was significant between glioma and non-neoplasm brain samples. So we picked out miR-95-3p, one of miR-95 mature sequences, for study. We analyzed our data and tried to identify correlations of miR-95-3p expression level with pathological grade and found miR-95-3p level increased with WHO grade.

As miR-95-3p has hardly been investigated in glioma previously, we planned functional experiments on multiple aspects of tumorigenesis, including proliferation, cell cycle, apoptosis and invasion. In order to set up the experimental group, miR-95-3p ASO was transfected into glioma cells to reduce the miR-95-3p level. In MTT assay, we observed downregulation of miR-95-3p repressed glioma cells proliferation. MTT results reflected the change of mitochondria viability, or the amount of living cells, regardless of the cause. To explain how the living cells decreased, we further analyzed the cell cycle and apoptosis by flow cytometry. Our results manifested no significant difference in cell cycle between experimental and control group, which indicated miR-95-3p does not affect glioma cell growth rate. Apoptosis analysis showed a significantly increased number of cell death in the experimental group. So we inferred that miR-95-3p suppresses glioma cell proliferation by inducing apoptosis, rather than by slowing down cell mitosis. As invasiveness is an important factor in tumor metastasis, we also performed transwell test to investigate the function of miR-95-3p on this aspect. Less cells transfected with miR-95-3p invaded through the membrane than control, as we expected.

In the following phase of the study, we aimed to understand the mechanism by which miR-95-3p mediates glioma behavior. We searched the online programs TargetScan, mirWalk and PicTar to identify the target of miR-95-3p and focused on CELF2. CELF (CUGBP- and ETR-3-like family) is a group of RNA-binding proteins encoding various functions in post-transcriptional level, including alternative splicing, RNA editing and mRNA translation (27). These proteins contain 3 RNA recognition motifs (RRMs), which are 80–90 amino acid domains with conserve sequences for proteins to bind RNAs (28). Two of the RRMs are located near the N-terminus and separated from the third one by an intervening bridge segment (29). CELF2 (also known as CUGBP2, NAPOR2, ETR3), one of the CELF family members, is expressed ubiquitously, albeit at higher level in mouse cells (30). In view of the fact that CELF2 was found involved in neuroblastoma cell apoptosis (31) and is encoded by a gene located on chromosome 10, which shows monosomy in ≤60% and partial loss in 80% of high grade gliomas (32,33), we hypothesized CELF2 may be implicated in the development of glioma as well. We employed luciferase reporter assay to verify our theory. The results indicated that miR-95-3p ASO significantly increased luciferase activity, which could be abrogated by mutation on the predicted binding site. In the experiments of western blotting and qRT-PCR, our data implied the levels of CELF2 and miR-95-3p were negatively correlated both in cell lines and human samples. The conclusion that CELF2 is a direct target of miR-95-3p was drawn.

To determine whether miR-95-3p regulates glioma cells biological behavior by CELF2, rescue experiments were performed. From the results we found that the effects of miR-95-3p ASO on glioma cells were rescued by decreased expression of CELF2. Thus, the miR-95-3p-CELF2 regulation mechanism on glioma was established. Furthermore, CELF2 was also reported to promote cell apoptosis by mediating Mcl-1, and downregulation of CELF2 by prostaglandin E2 represses mitotic catastrophe induced by radiation in colon cancer cells (34,35). Subramaniam et al (36) also found that CELF2 plays a role in mitotic catastrophe of pancreatic cancer cells. It can be inferred that CELF2 may play an important role in the mitotic process, although miR-95-3p does not, and there may be other pathways related to CELF2 regulating glioma.

In conclusion, this study demonstrates miR-95-3p is upregulated in glioma samples and downregulation of miR-95-3p inhibits proliferation, invasion and promotes apoptosis of glioma cells by targeting CELF2. Thus, we identified miR-95-3p as a putative therapeutic target and CELF2 as a potential tumor suppressor.

Acknowledgements

We thank Mr. Shuang Chen and Yong Pu (Saierbio, Tianjing, China) for their technical advice.

References

1 

Giese A, Bjerkvig R, Berens ME and Westphal M: Cost of migration: Invasion of malignant gliomas and implications for treatment. J Clin Oncol. 21:1624–1636. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Lefranc F, Brotchi J and Kiss R: Possible future issues in the treatment of glioblastomas: Special emphasis on cell migration and the resistance of migrating glioblastoma cells to apoptosis. J Clin Oncol. 23:2411–2422. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Singh SK, Clarke ID, Hide T and Dirks PB: Cancer stem cells in nervous system tumors. Oncogene. 23:7267–7273. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, Hahn WC, Ligon KL, Louis DN, Brennan C, et al: Malignant astrocytic glioma: Genetics, biology, and paths to treatment. Genes Dev. 21:2683–2710. 2007. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Eulalio A, Huntzinger E and Izaurralde E: Getting to the root of miRNA-mediated gene silencing. Cell. 132:9–14. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Carthew RW: Gene regulation by microRNAs. Curr Opin Genet Dev. 16:203–208. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Zhang B, Pan X, Cobb GP and Anderson TA: microRNAs as oncogenes and tumor suppressors. Dev Biol. 302:1–12. 2007. View Article : Google Scholar

9 

Lujambio A and Lowe SW: The microcosmos of cancer. Nature. 482:347–355. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Bagnoli M, De Cecco L, Granata A, Nicoletti R, Marchesi E, Alberti P, Valeri B, Libra M, Barbareschi M, Raspagliesi F, et al: Identification of a chrXq27.3 microRNA cluster associated with early relapse in advanced stage ovarian cancer patients. Oncotarget. 2:1265–1278. 2011.

11 

Iorio MV and Croce CM: microRNA involvement in human cancer. Carcinogenesis. 33:1126–1133. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Godlewski J, Nowicki MO, Bronisz A, Nuovo G, Palatini J, De Lay M, Van Brocklyn J, Ostrowski MC, Chiocca EA and Lawler SE: MicroRNA-451 regulates LKB1/AMPK signaling and allows adaptation to metabolic stress in glioma cells. Mol Cell. 37:620–632. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Gabriely G, Yi M, Narayan RS, Niers JM, Wurdinger T, Imitola J, Ligon KL, Kesari S, Esau C, Stephens RM, et al: Human glioma growth is controlled by microRNA-10b. Cancer Res. 71:3563–3572. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Li Y, Guessous F, Zhang Y, Dipierro C, Kefas B, Johnson E, Marcinkiewicz L, Jiang J, Yang Y, Schmittgen TD, et al: MicroRNA-34a inhibits glioblastoma growth by targeting multiple oncogenes. Cancer Res. 69:7569–7576. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Chan JA, Krichevsky AM and Kosik KS: MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 65:6029–6033. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Ciafrè SA, Galardi S, Mangiola A, Ferracin M, Liu CG, Sabatino G, Negrini M, Maira G, Croce CM and Farace MG: Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun. 334:1351–1358. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Zhou J, Wang W, Gao Z, Peng X, Chen X, Chen W, Xu W, Xu H, Lin MC and Jiang S: MicroRNA-155 promotes glioma cell proliferation via the regulation of MXI1. PLoS One. 8:e830552013. View Article : Google Scholar :

18 

Godlewski J, Nowicki MO, Bronisz A, Williams S, Otsuki A, Nuovo G, Raychaudhury A, Newton HB, Chiocca EA and Lawler S: Targeting of the Bmi-1 oncogene/stem cell renewal factor by microRNA-128 inhibits glioma proliferation and self-renewal. Cancer Res. 68:9125–9130. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Silber J, Lim DA, Petritsch C, Persson AI, Maunakea AK, Yu M, Vandenberg SR, Ginzinger DG, James CD, Costello JF, et al: miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells. BMC Med. 6:142008. View Article : Google Scholar : PubMed/NCBI

20 

Bier A, Giladi N, Kronfeld N, Lee HK, Cazacu S, Finniss S, Xiang C, Poisson L, deCarvalho AC, Slavin S, et al: MicroRNA-137 is downregulated in glioblastoma and inhibits the stemness of glioma stem cells by targeting RTVP-1. Oncotarget. 4:665–676. 2013.PubMed/NCBI

21 

Skalsky RL and Cullen BR: Reduced expression of brain-enriched microRNAs in glioblastomas permits targeted regulation of a cell death gene. PLoS One. 6:e242482011. View Article : Google Scholar : PubMed/NCBI

22 

Huang Z, Huang S, Wang Q, Liang L, Ni S, Wang L, Sheng W, He X and Du X: MicroRNA-95 promotes cell proliferation and targets sorting Nexin 1 in human colorectal carcinoma. Cancer Res. 71:2582–2589. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Huang X, Taeb S, Jahangiri S, Emmenegger U, Tran E, Bruce J, Mesci A, Korpela E, Vesprini D, Wong CS, et al: miRNA-95 mediates radioresistance in tumors by targeting the sphingolipid phosphatase SGPP1. Cancer Res. 73:6972–6986. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Zhang Y, Li M, Wang H, Fisher WE, Lin PH, Yao Q and Chen C: Profiling of 95 microRNAs in pancreatic cancer cell lines and surgical specimens by real-time PCR analysis. World J Surg. 33:698–709. 2009. View Article : Google Scholar

25 

Xiao Z, Ching Chow S, Han Li C, Chun Tang S, Tsui SK, Lin Z and Chen Y: Role of microRNA-95 in the anticancer activity of Brucein D in hepatocellular carcinoma. Eur J Pharmacol. 728:141–150. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Nurul-Syakima AM, Yoke-Kqueen C, Sabariah AR, Shiran MS, Singh A and Learn-Han L: Differential microRNA expression and identification of putative miRNA targets and pathways in head and neck cancers. Int J Mol Med. 28:327–336. 2011.PubMed/NCBI

27 

Barreau C, Paillard L, Méreau A and Osborne HB: Mammalian CELF/Bruno-like RNA-binding proteins: Molecular characteristics and biological functions. Biochimie. 88:515–525. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Keene JD: RNA recognition by autoantigens and autoantibodies. Mol Biol Rep. 23:173–181. 1996. View Article : Google Scholar : PubMed/NCBI

29 

Ladd AN and Cooper TA: Multiple domains control the subcellular localization and activity of ETR-3, a regulator of nuclear and cytoplasmic RNA processing events. J Cell Sci. 117:3519–3529. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Wang GS, Kearney DL, De Biasi M, Taffet G and Cooper TA: Elevation of RNA-binding protein CUGBP1 is an early event in an inducible heart-specific mouse model of myotonic dystrophy. J Clin Invest. 117:2802–2811. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Choi DK, Ito T, Mitsui Y and Sakaki Y: Fluorescent differential display analysis of gene expression in apoptotic neuroblastoma cells. Gene. 223:21–31. 1998. View Article : Google Scholar : PubMed/NCBI

32 

Pershouse MA, Stubblefield E, Hadi A, Killary AM, Yung WK and Steck PA: Analysis of the functional role of chromosome 10 loss in human glioblastomas. Cancer Res. 53:5043–5050. 1993.PubMed/NCBI

33 

Ransom DT, Ritland SR, Moertel CA, Dahl RJ, O'Fallon JR, Scheithauer BW, Kimmel DW, Kelly PJ, Olopade OI, Diaz MO, et al: Correlation of cytogenetic analysis and loss of heterozygosity studies in human diffuse astrocytomas and mixed oligo-astrocytomas. Genes Chromosomes Cancer. 5:357–374. 1992. View Article : Google Scholar : PubMed/NCBI

34 

Subramaniam D, Natarajan G, Ramalingam S, Ramachandran I, May R, Queimado L, Houchen CW and Anant S: Translation inhibition during cell cycle arrest and apoptosis: Mcl-1 is a novel target for RNA binding protein CUGBP2. Am J Physiol Gastrointest Liver Physiol. 294:G1025–G1032. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Natarajan G, Ramalingam S, Ramachandran I, May R, Queimado L, Houchen CW and Anant S: CUGBP2 down-regulation by prostaglandin E2 protects colon cancer cells from radiation-induced mitotic catastrophe. Am J Physiol Gastrointest Liver Physiol. 294:G1235–G1244. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Subramaniam D, Ramalingam S, Linehan DC, Dieckgraefe BK, Postier RG, Houchen CW, Jensen RA and Anant S: RNA binding protein CUGBP2/CELF2 mediates curcumin-induced mitotic catastrophe of pancreatic cancer cells. PLoS One. 6:e169582011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2015
Volume 47 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fan B, Jiao B, Fan F, Lu S, Song J, Guo C, Yang J and Yang L: Downregulation of miR-95-3p inhibits proliferation, and invasion promoting apoptosis of glioma cells by targeting CELF2. Int J Oncol 47: 1025-1033, 2015.
APA
Fan, B., Jiao, B., Fan, F., Lu, S., Song, J., Guo, C. ... Yang, L. (2015). Downregulation of miR-95-3p inhibits proliferation, and invasion promoting apoptosis of glioma cells by targeting CELF2. International Journal of Oncology, 47, 1025-1033. https://doi.org/10.3892/ijo.2015.3080
MLA
Fan, B., Jiao, B., Fan, F., Lu, S., Song, J., Guo, C., Yang, J., Yang, L."Downregulation of miR-95-3p inhibits proliferation, and invasion promoting apoptosis of glioma cells by targeting CELF2". International Journal of Oncology 47.3 (2015): 1025-1033.
Chicago
Fan, B., Jiao, B., Fan, F., Lu, S., Song, J., Guo, C., Yang, J., Yang, L."Downregulation of miR-95-3p inhibits proliferation, and invasion promoting apoptosis of glioma cells by targeting CELF2". International Journal of Oncology 47, no. 3 (2015): 1025-1033. https://doi.org/10.3892/ijo.2015.3080