MicroRNA-302a targets GAB2 to suppress cell proliferation, migration and invasion of glioma

  • Authors:
    • Jiuhong Ma
    • Jia Yu
    • Jingmei Liu
    • Xing Yang
    • Miao Lou
    • Jinghui Liu
    • Fuqiang Feng
    • Peigang Ji
    • Liang Wang
  • View Affiliations

  • Published online on: December 15, 2016     https://doi.org/10.3892/or.2016.5320
  • Pages: 1159-1167
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioma is the most frequent and aggressive primary tumor of the brain in humans. Over the last few decades, significant progress has been made in early detection and multi-mode treatments, but the prognosis of gliomas is still extremely poor. MicroRNAs are endogenously expressed non-coding, single strand and short RNA molecules. Increasing number of studies demonstrated that microRNAs are dysregulated in a variety of human cancers, and play significant roles in tumorigenesis and tumor development, including glioma. In the present study, we for the first time found that microRNA-302a (miR-302a) was significantly downregulated in both glioma tissues and cell lines. In glioma patients, low miR-302a expression was correlated with KPS score and WHO grade. Restoration of miR-302a expression inhibited cell proliferation, migration and invasion of glioma in vitro. In addition, GAB2 was identified as a direct target of miR-320a. In clinical glioma tissues, GAB2 was upregulated and in­versely correlated with miR-302a expression. GAB2 underexpression had similar biological roles with miR-302a overexpression in glio­ma cells, further confirming that GAB2 was a functional downstream target of miR-302a. Moreover, rescue experiments showed that upregulation of GAB2 effectively reversed the inhibition effects of miR-302a on glioma cells proliferation, migration and invasion. These findings suggested that miR-302a is an important tumor suppressor of glioma progression by directly targeting GAB2, thus providing new insight into the molecular mechanisms underlying glioma occurrence, development and evolution of glioma.

Introduction

Glioma is the most frequent and aggressive primary tumor of the brain in humans, and accounts for approximately 80% of primary malignant brain tumors (1,2). In view of their likely cellular origins, glioma could be classified into four groups, including astrocytomas, oligodendrogliomas, ependymomas and mixed tumors (3). Currently, the standard treatments of glioma include surgery resection followed by radiotherapy and chemotherapy (4). Despite significant progress made in early detection and multi-mode treatments over the last few decades, the prognosis of gliomas is still extremely poor (5). The average 5-year survival rate of glioma is only 4–5% and mean survival time following diagnosis is 12–15 months (6,7). The main cause of poor prognosis is that glioma is characterized by rapid growth, high invasiveness and suppression of cell apoptosis (8,9). Besides, poorly understanding the etiology and progression of glioma hampers the development of treatments and surveillance strategies (10). Therefore, fully understanding the molecular mechanisms underlying occurrence, development and evolution of glioma is urgently needed to investigate novel and effective therapeutic treatments.

microRNAs (miRNAs) are endogenously expressed non-coding, single strand and short RNA molecules that are approximately 18–25 nucleotide in length (11). miRNAs have been reported to participate in the regulation of human physiological and pathological processes, such as cell proliferation, cycle, apoptosis, differentiation, metabolism, migration, invasion and metastasis (1214). miRNAs exert their regulatory functions through interacting with complementary sites that have the 3′-untranslated regions (3′UTRs) of target mRNAs, resulting in translation repression or mRNA degradation, and thus decreasing the protein expression of their target genes (15,16). An increasing number of studies demonstrated that miRNAs are dysregulated in a variety of human cancers, and play significant roles in tumorigenesis and tumor development (1719). These miRNAs may have similar features to oncogenes or tumor suppressor genes mainly depending on the characteristic and roles of their target genes (20). Downregulated miRNAs in cancer may act as tumor suppressors and suppress carcinogenesis and progression by negatively regulating oncogenes. Conversely, upregulated miRNAs in cancer may function as oncogenes through directly targeting tumor suppressors (2124). Therefore, miRNAs may serve as novel therapeutic targets for antitumor therapeutic agents.

Although miR-302a has been reported to be frequently dysregulated in various human cancers, there is no information available concerning miR-302a in glioma. In the present study, we observed that miR-302a expression was downregulated in glioma tissues and cell lines. Moreover, reduced miR-302a expression was significantly correlated with KPS score and WHO grade of glioma patients. In addition, we provide evidence that miR-302a may act as a tumor suppressor in glioma by inhibiting cell proliferation, migration and invasion. Furthermore, we demonstrated that miR-302a exerted its tumor suppressive functions through directly targeting GAB2. These findings provide valuable clues toward fully understanding the molecular mechanisms underlying tumorigenesis and progression of glioma, and may present an opportunity to develop novel therapeutic targets for glioma treatments.

Materials and methods

Clinical specimens

Seventy-nine paired glioma tissues and adjacent normal tissues were obtained from surgical resections of glioma in the Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Shanxi, China. The adjacent normal tissue was defined as 2 cm away from the tumor tissue. All tissues were derived from glioma cases who had not been treated with chemotherapy or radiotherapy before operation. After excision, tissue samples were immediately frozen in liquid nitrogen and stored at −80°C until use. This study was approved by the Ethics Committee of Tangdu Hospital.

Cell culture

Human glioma cell lines (U251, U87, U373, LN229, A172) and normal human glial cell line (HEB) were obtained from American Type Culture Collection (ATCC, Manassas, VA, USA). Human embryonic kidney (HEK) 293T cell line was purchased from Cell Bank of the Chinese Academy of Sciences (Shanghai, China). All cell lines were grown in Dulbecco's modified Eagle's medium (Gibco, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco) and 1% penicillin/streptomycin (Gibco) at 37°C in a humidified atmosphere with 5% CO2.

Oligonucleotides and cell transfection

miR-302a mimics, miRNA mimics negative control (NC), siRNA targeting GAB2 (si-GAB2) and its negative control (si-NC) were chemically synthesized by GenePharma (Shanghai, China). Overexpression plasmid of GAB2 and its negative control (pCDNA3.1-GAB2 and pCDNA3.1) were purchased from RiboBio (Guangzhou, China). Cells were transfected with these oligonucleotides by using Opti-MEM I and Lipofectamine 2000 reagents (Invitrogen, Carlsbad, CA, USA) at approximately 50–70% confluence according to the manufacturer's instructions.

Quantitative reverse transcription polymerase chain reaction (qRT-PCR)

Total RNA from tissues and cells was isolated using TRIzol reagent (Invitrogen). miR-302a expression was detected using TaqMan microRNA assays (Applied Biosystems, Foster City, CA) following the manufacturer's instructions. U6 was used as an internal control. For mRNA expression, single-stranded cDNA was synthesized from 1 µg of total RNA using an M-MLV Reverse Transcription system (Promega Corp., Madison, WI, USA) following the manufacturer's protocol. Real-time quantitative PCR was performed to measure GAB2 mRNA expression by using SYBR Premix Ex Taq (Takara, Dalian, China) and run in triplicate in an ABI Prism 7500 Sequence detection system (Applied Biosystems). GAPDH was used as an internal control. The relative gene expression was analyzed using the 2−∆∆Ct method.

Cell Counting Kit-8 assay

Transfected cells were harvested, seeded into 96-well plates at a density of 2000 cells/well and cultured for 24, 48, 72, and 96 h. At each time point, Cell Counting Kit-8 (CCK8) assay (Dojindo, Japan) was performed according to the manufacturer's instructions. Briefly, 10 µl of CCK8 solution was added into each well and incubated for 2 h at 37°C in a humidified atmosphere with 5% CO2. The absorbance at 450 nm was determined with a microplate reader (Thermo Scientific, Hudson, NH, USA).

Cell migration and invasion assays

Transwell chambers (24-well) with 8 µm pore size polycarbonate membrane (Millipore, Billerica, MA, USA) were used to carry out cell migration and invasion assays. For cell migration assays, cells were collected 48 h post-transfection and suspended in FBS-free culture medium. Cells (5×104) were seeded on the top side of the Transwell chambers, culture medium containing 20% FBS was added to the low side of the Transwell chambers. After 48 h incubation in a humidified atmosphere with 5% CO2 at 37°C, cells remaining on the upper chambers were removed with cottons swabs. The migrated cells were fixed with 100% methanol, stained with 0.5% crystal violet, photographed and counted under an IX71 inverted microscope (Olympus, Tokyo, Japan). The procedures of cell invasion assays were similar with cell migration assays, except that the Transwell chambers were pre-coated with Matrigel (BD, Franklin Lakes, NJ, USA).

Bioinformatics analysis and luciferase reporter assay

The candidate target genes of miR-302a were generated using three publicly available databases: TargetScan (www.targetscan.org/), miRanda(www.microrna.org) and PicTar (http://pictar.mdc-berlin.de/).

The 3′UTR sequence of GAB2 predicted to interact with miR-302a as well as a mutated sequence with the predicted target sites were synthesized and inserted into pGL3 plasmid. HEK293T cells were seeded into 24-well plates and transfected with pGL3-GAB2-3′UTR Wt or pGL3-GAB2-3′UTR Mut, together with miR-302a mimics or NC by using FuGENE HD transfection reagent (Promega, Manheim, Germany). After 48 h of incubation, firefly and renilla luciferase activities were measured with Dual-Luciferase reporter assay system (Promega) following to the manufacturer's protocol. Firefly luciferase activities were used as an internal control for renilla luciferase activities.

Western blot analysis

The primary antibodies used were mouse anti-human monoclonal GAB2 antibody (1:1000 dilution; sc-365590; Santa Cruz Biotechnology, Santa Cruz, CA, USA) and mouse anti-human monoclonal GADPH antibody (1:1000 dilution; sc-137179; Santa Cruz Biotechnology). Cells were collected at 72 h post-transfection, washed with ice-cold PBS (Gibco) and treated with RIPA protein lysis buffer supplemented with protease inhibitor. The homogenates were then centrifuged for 30 min at 4°C, 12,000 rpm, and the supernatants were collected as protein samples. Equal proteins were separated by 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis, transferred onto polyvinylidene fluoride membranes (Millipore) and probed with primary antibodies at 4°C overnight followed by incubation with goat anti-mouse horseradish peroxidase (HRP)-conjugated secondary antibody (1:5000 dilution; Santa Cruz Biotechnology). Finally, the blot was developed with the enhanced chemiluminescence (ECL) kit (Pierce Biotechnology, Rockford, IL, USA), and images were captured using the FluorChem imaging system (Alpha Innotech, San Leandro, CA, USA).

Statistical analysis

Data are expressed as mean ± SD. Statistical analyses were performed with SPSS 19.0 software (SPSS, Chicago, IL, USA) using Student's t-tests or Chi-square tests. P<0.05 was considered to be statistically significant.

Results

miR-302a is greatly downregulated both in glioma tissues and cell lines

To identify the roles of miR-302a in glioma, we first measured its expression in paired glioma tissues and adjacent normal brain tissues using qRT-PCR. Compared with adjacent normal tissues, glioma tissues showed reduced expression levels of miR-302a (Fig. 1A, P<0.05). Similarly, miR-302a expression was lower in glioma cell lines (U251, U87, U373, LN229, A172) compared with normal human glial cell line (HEB) (Fig. 1B, P<0.05). Thus, the above results indicated that the downregulation of miR-302a may be critically involved in glioma occurrence and development.

Correlation between miR-302a expression and clinicopathological features in gliomas

We next investigated the correlation between the miR-302a expression and clinicopathological features in gliomas. As shown in Table I, low miR-302a expression was significantly correlated with KPS score (P=0.008) and WHO grade (P=0.035). However, there was no association between miR-302a expression and gender (P=0.709), age (P=0.593), family history of cancer (P=0.150), tumor size (P=0.442), and tumor location (P=0.427).

Table I.

Correlation between miR-302a expression and clinicopathologic characteristics in human glioma.

Table I.

Correlation between miR-302a expression and clinicopathologic characteristics in human glioma.

miR-302a expression

Clinicopathologic characteristicsCase no.LowHighP-value
Gender
  Male6649170.709
  Female13  9  4
Age (years)
  <604532130.593
  ≥603426  8
Family history of cancer
  No4838100.150
  Yes312011
Tumor size (cm)
  <32822  60.442
  ≥3513615
Tumor location
  Supratentorial6345180.427
  Infratentorial1613  3
KPS score
  <804941  80.008
  ≥80301713
WHO grade
  I–II3018120.035
  III–IV4940  9

[i] WHO, World Health Organization; KPS, Karnofsky performance score.

miR-302a suppresses U251 and U87 cell proliferation, migration and invasion in vitro

miR-302a was downregulated in glioma tissues and cell lines, hence the functions of miR-302a were investigated using miR-302a mimics in U251 and U87 cells. The transfection efficiencies were evaluated by measuring miR-302a expression in U251 and U87 cells after transfection with miR-302a mimics or NC. As shown in Fig. 2A, miR-302a expression levels in U251 and U87 cells transfected with miR-302a mimics were increased markedly compared with NC groups (P<0.05).

To investigate the effect of miR-302a overexpression on proliferation of glioma cells, CCK8 assay was performed. The results showed that restoration of miR-302a expression decreased U251 and U87 cell proliferation abilities (Fig. 2B, P<0.05). To explore whether the upregulation of miR-302a could influence the metastasis of glioma cells in vitro, cell migration and invasion assays were used. As shown in Fig. 2C and D, ectopic of miR-302a expression reduced the number of migrated and invaded cells compared with those of NC groups (P<0.05). These results indicated that miR-302a had tumor-suppressive roles that could suppress glioma cell growth and metastasis.

GAB2 is a direct target of miR-302a

To investigate the molecular mechanism underlying the tumor suppressive roles of miR-302a in glioma, bioinformatic analysis was performed with publicly available databases. As shown in Fig. 3A, a binding site of miR-302a was observed in the 3′UTR of GAB2. Therefore, GAB2 could be a potential target of miR-302a.

To test this hypothesis, luciferase reporter assay was used. As shown in Fig. 3B, upregulation of miR-302a decreased the luciferase activities of pGL3- GAB2-3′UTR Wt in HEK293T cells (P<0.05) without change in luciferase activities of pGL3-GAB2-3′UTR Mut.

Furthermore, qRT-PCR and western blot analysis were performed to investigate whether miR-302a overexpression could downregulate GAB2 expression in glioma cells. We found that miR-302a could decrease GAB2 at mRNA (Fig. 3C, P<0.05) and protein (Fig. 3D, P<0.05) levels in U251 and U87 cells. These findings demonstrate that miR-302a negatively regulated GAB2 expression by directly binding 3′UTR of GAB2.

GAB2 is upregulated and inversely correlated with miR-302a expression in glioma tissues

Further experiments were performed to determine GAB2 mRNA and protein expression in paired glioma tissues and adjacent normal brain tissues. Results of qRT-PCR showed that GAB2 mRNA was significantly upregulated in clinical glioma tissues compared with those in adjacent normal brain tissues (Fig. 4A, P<0.05). In addition, spearman's correlation analysis was adopted to analyze the relationship between miR-302a and GAB2 mRNA expression in glioma tissues and found that the expression level of miR-302a was inversely correlated with GAB2 mRNA in glioma tissues (Fig. 4B, r= −0.5642, P<0.0001). Furthermore, western blot analysis indicated that GAB2 protein expression was increased in glioma tissues in comparison with adjacent normal brain tissues (Fig. 4C, P<0.05).

Knockdown of GAB2 exhibits similar effects with miR-302a overexpression in glioma cells

To investigate the biological roles of GAB2 in glioma, siRNA targeting GAB2 was used to knock down GAB2 expression in U251 and U87 cells. The efficiency of si-GAB2 transfection was assessed by using western blot analysis. As shown in Fig. 5A, GAB2 protein was downregulated in both U251 and U87 cells (P<0.05). The effects of GAB2 underexpression on glioma cell proliferation, migration and invasion were determined using CCK8 assay and cell migration and invasion assay, respectively. As shown in Fig. 5B-C, downregulation of GAB2 significantly inhibited U251 and U87 cells proliferation (P<0.05), migration and invasion (P<0.05) in vitro. These results suggested that the knockdown of GAB2 had similar biological roles with miR302a overexpression in glioma cells, further confirming that GAB2 was a functional downstream target of miR-302a.

Upregulation of GAB2 reverses the inhibitory effects of miR-302a in glioma cells

To explore whether the suppressive roles of miR-302a in glioma cells were mediated by GAB2, U251 and U87 cells were transfected with pCDNA3.1-GAB2. As shown in Fig. 6A, GAB2 protein was upregulated in U251 and U87 cells after transfection with pCDNA3.1-GAB2 (P<0.05). Rescue experiments were performed in U251 and U87 cells co-transfected with miR-302a mimics or NC, and pCDNA3.1-GAB2. As shown in Fig. 6B-D, upregulation of GAB2 effectively reversed the inhibition effects of miR-302a on U251 and U87 cell proliferation (P<0.05), migration and invasion (P<0.05). These findings suggested that miR-302a inhibited growth and metastasis in glioma cells, at least in part, by suppression of GAB2.

Discussion

Fully understanding the mechanisms underlying glioma occurrence and development is essential for providing novel therapeutic strategies for glioma patients to improve prognosis (25). The aim of the present study was to investigate the expression and roles of miR-302a in glioma and the possible molecular mechanisms. We found that expression levels of miR-302a were reduced in both glioma tissues and cell lines. Low miR-302a expression was significantly correlated with adverse clinicopathological characteristics in glioma. miR-302a overexpression inhibited cell proliferation, migration and invasion of glioma. Additional in vitro studies demonstrated that GAB2 was the direct target gene of miR-302a in glioma. Thus, miR-302a could suppress glioma growth and metastasis through directly targeting GAB2, suggesting that miR-302a functions as a tumor suppressor in glioma and play essential roles in the carcinogenesis and progression of glioma.

Previous studies have revealed that miR-302a was abnormally expressed in several human cancers. Guo et al reported that miR-302a was decreased in ovarian cancer tissues and cell lines. miR-302a expression was significantly correlated with TNM stage of ovarian cancer patients (26). Wei et al found that miR-302a expression was lower in colorectal cancer cell lines than that in normal colon epithelium cell line (27). In prostate cancer, miR-302a has been found at low level in tumor tissues. Moreover, expression level of miR-302a was associated with Gleason score (28). These studies suggested that a low expression pattern of miR-302a may be universal regardless of ethic background and be involved in the occurrence and development of human cancer.

Dysfunction of miR-302a has been found to be involved in the development and progression of human cancers. In ovarian cancer, restoration of miR-302a expression inhibited tumor cell proliferation, transition from G1 phase to S phase and induced apoptosis through directly targeting SDC1 (26). In colorectal cancer, miR-302a decreased cell proliferation and invasion and has been confirmed as a tumor suppressor through regulation of MAPK and PI3K/Akt signaling pathways (27). Zhang et al demonstrated that, in prostate cancer, miR-302a was found to improve G1/S cell cycle arrest and reduce cell growth in vitro and in vivo (28). Liang et al reported that miR-302a was downregulated in metastatic breast cancer cells and tumor tissues. Ectopic miR-302a expression suppressed invasion and metastasis of breast cancer cells in vitro and in vivo by negative regulation of CXCR4 (29). Moreover, miR-302a overexpression enhanced the chemosensitivity of breast cancer cells to adriamycin and mitoxantrone through directly targeting MEKK1 and BCRP, respectively (30,31). However, the biological roles of miR-302a in glioma remain unknown. In the present study, we found that the upregulation of miR-302a inhibited glioma cell proliferation, migration and invasion in vitro, suggesting that miR-302a is a novel tumor-suppressive miRNA that plays significant roles in the regulation of glioma growth and metastasis.

Since the potential molecular mechanism of miR-302a as a tumor suppressor in glioma was unknown, we explored the potential mechanism. In the present study, an important molecular association between miR-302a and GAB2 was identified. Firstly, bioinformatics analysis indicated that GAB2 could be a potential target of miR-302a. Subsequently, GAB2 was found to highly expressed in clinical glioma tissues and inversely correlated with miR-302a expression. Importantly, upregulation of miR-302a decreased GAB2 expression in glioma cells. This hypothesis was further supported by the luciferase reporter assay, in which the results demonstrated that miR-302a directly targeted the 3′UTR of GAB2. Moreover, the knockdown of GAB2 had similar biological roles with miR-302a overexpression in glioma cells. Finally, rescue experiments showed that restoration of GAB2 reversed the inhibitory effects of miR-302a in glioma cells. These results strongly demonstrated that GAB2 was a novel direct and functional target of miR-302a in glioma.

GAB2, a member of the DOS/Gab family of scaffolding adapters, contains an N-terminal pleckstrin homology domain and C-terminal portion with multiple tyrosine phosphorylation sites and proline rich motifs (32,33). GAB2 integrate and amplify signals from cytokines, growth factors and antigen receptors, as well as from cell adhesion molecules. In addition, GAB2 diversify signals by channeling the input information from activated receptors into signal pathways with distinct biological functions (34,35). Previous studies showed that GAB2 was highly expressed in various kinds of human cancer, such as hepatocellular carcinoma (36), ovarian cancer (37), colorectal cancer (38) and breast cancer (39). In glioma, GAB2 was obviously upregulated in tumor tissues and cell lines. In addition, upregulation of GAB2 expression was correlated with WHO grade of glioma patients. Gliomas with high GAB2 level had shorter survival time than patients with low GAB2 (40). Functionally, GAB2 underexpression inhibited glioma cell proliferation, migration and invasion (40,41). In the present study, we also found that knockdown of GAB2 significantly suppressed the proliferation, migration and invasion of glioma cells, indicating the oncogenic role of GAB2 in glioma. Therefore, GAB2 downregulation by miR-302a may stress the mechanism by which miR-302a decreased glioma growth and metastasis.

In summary, the present study for the first time, to the best of our knowledge, demonstrated an inhibitory role of miR-302a in glioma by directly targeting GAB2 and suggests that miR-302a is a novel therapeutic target for treatment of glioma.

References

1 

Kuhnt D, Becker A, Ganslandt O, Bauer M, Buchfelder M and Nimsky C: Correlation of the extent of tumor volume resection and patient survival in surgery of glioblastoma multiforme with high-field intraoperative MRI guidance. Neuro Oncol. 13:1339–1348. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Houdek Z, Cendelín J, Kulda V, Babuška V, Cedíková M, Králíčková M, Pacherník J, Stefano GB and Vožeh F: Intracerebellar application of P19-derived neuroprogenitor and naive stem cells to Lurcher mutant and wild type B6CBA mice. Med Sci Monit. 18:BR174–BR180. 2012. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Ehtesham M, Stevenson CB and Thompson RC: Stem cell therapies for malignant glioma. Neurosurg Focus. 19:E52005. View Article : Google Scholar : PubMed/NCBI

5 

Omuro A and DeAngelis LM: Glioblastoma and other malignant gliomas: A clinical review. JAMA. 310:1842–1850. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Schwartzbaum JA, Fisher JL, Aldape KD and Wrensch M: Epidemiology and molecular pathology of glioma. Nat Clin Pract Neurol. 2:494–503. 2006. View Article : Google Scholar : PubMed/NCBI

7 

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

8 

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

9 

Chen R, Nishimura MC, Bumbaca SM, Kharbanda S, Forrest WF, Kasman IM, Greve JM, Soriano RH, Gilmour LL, Rivers CS, et al: A hierarchy of self-renewing tumor-initiating cell types in glioblastoma. Cancer Cell. 17:362–375. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Wibom C, Späth F, Dahlin AM, Langseth H, Hovig E, Rajaraman P, Johannesen TB, Andersson U and Melin B: Investigation of established genetic risk variants for glioma in prediagnostic samples from a population-based nested case-control study. Cancer Epidemiol Biomarkers Prev. 24:810–816. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Kasinski AL and Slack FJ: Epigenetics and genetics. MicroRNAs en route to the clinic: Progress in validating and targeting microRNAs for cancer therapy. Nat Rev Cancer. 11:849–864. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Yang L, Li Q, Wang Q, Jiang Z and Zhang L: Silencing of miRNA-218 promotes migration and invasion of breast cancer via Slit2-Robo1 pathway. Biomed Pharmacother. 66:535–540. 2012. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Kloosterman WP and Plasterk RH: The diverse functions of microRNAs in animal development and disease. Dev Cell. 11:441–450. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Treiber T, Treiber N and Meister G: Regulation of microRNA biogenesis and function. Thromb Haemost. 107:605–610. 2012. View Article : Google Scholar : PubMed/NCBI

16 

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 : PubMed/NCBI

17 

Wang K, Wang X, Zou J, Zhang A, Wan Y, Pu P, Song Z, Qian C, Chen Y, Yang S, et al: miR-92b controls glioma proliferation and invasion through regulating Wnt/beta-catenin signaling via Nemo-like kinase. Neuro Oncol. 15:578–588. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Hu X, Chen D, Cui Y, Li Z and Huang J: Targeting microRNA-23a to inhibit glioma cell invasion via HOXD10. Sci Rep. 3:34232013.PubMed/NCBI

19 

Wu D, Zhou Y, Pan H, Zhou J, Fan Y and Qu P: microRNA-99a inhibiting cell proliferation, migration and invasion by targeting fibroblast growth factor receptor 3 in bladder cancer. Oncol Lett. 7:1219–1224. 2014.PubMed/NCBI

20 

Chen Y, Gao DY and Huang L: In vivo delivery of miRNAs for cancer therapy: Challenges and strategies. Adv Drug Deliv Rev. 81:128–141. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Medina PP, Nolde M and Slack FJ: OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature. 467:86–90. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Obad S, dos Santos CO, Petri A, Heidenblad M, Broom O, Ruse C, Fu C, Lindow M, Stenvang J, Straarup EM, et al: Silencing of microRNA families by seed-targeting tiny LNAs. Nat Genet. 43:371–378. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Saito Y, Liang G, Egger G, Friedman JM, Chuang JC, Coetzee GA and Jones PA: Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell. 9:435–443. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Liu N and Tu Y: Systematic review of microRNAs and its therapeutic potential in glioma. Cancer Transl Med. 1:50–66. 2015. View Article : Google Scholar

25 

Liu K, Liu S, Zhang W, Jia B, Tan L, Jin Z and Liu Y: miR-494 promotes cell proliferation, migration and invasion, and increased sorafenib resistance in hepatocellular carcinoma by targeting PTEN. Oncol Rep. 34:1003–1010. 2015.PubMed/NCBI

26 

Guo T, Yu W, Lv S, Zhang C and Tian Y: MiR-302a inhibits the tumorigenicity of ovarian cancer cells by suppression of SDC1. Int J Clin Exp Pathol. 8:4869–4880. 2015.PubMed/NCBI

27 

Wei ZJ, Tao ML, Zhang W, Han GD, Zhu ZC, Miao ZG, Li JY and Qiao ZB: Up-regulation of microRNA-302a inhibited the proliferation and invasion of colorectal cancer cells by regulation of the MAPK and PI3K/Akt signaling pathways. Int J Clin Exp Pathol. 8:4481–4491. 2015.PubMed/NCBI

28 

Zhang GM, Bao CY, Wan FN, Cao DL, Qin XJ, Zhang HL, Zhu Y, Dai B, Shi GH and Ye DW: MicroRNA-302a suppresses tumor cell proliferation by inhibiting AKT in prostate cancer. PLoS One. 10:e01244102015. View Article : Google Scholar : PubMed/NCBI

29 

Liang Z, Bian X and Shim H: Inhibition of breast cancer metastasis with microRNA-302a by downregulation of CXCR4 expression. Breast Cancer Res Treat. 146:535–542. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Zhao L, Wang Y, Jiang L, He M, Bai X, Yu L and Wei M: MiR-302a/b/c/d cooperatively sensitizes breast cancer cells to adriamycin via suppressing P-glycoprotein (P-gp) by targeting MAP/ERK kinase kinase 1 (MEKK1). J Exp Clin Cancer Res. 35:252016. View Article : Google Scholar : PubMed/NCBI

31 

Wang Y, Zhao L, Xiao Q, Jiang L, He M, Bai X, Ma M, Jiao X and Wei M: miR-302a/b/c/d cooperatively inhibit BCRP expression to increase drug sensitivity in breast cancer cells. Gynecol Oncol. 141:592–601. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Nishida K and Hirano T: The role of Gab family scaffolding adapter proteins in the signal transduction of cytokine and growth factor receptors. Cancer Sci. 94:1029–1033. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Hibi M and Hirano T: Gab-family adapter molecules in signal transduction of cytokine and growth factor receptors, and T and B cell antigen receptors. Leuk Lymphoma. 37:299–307. 2000.PubMed/NCBI

34 

Wöhrle FU, Daly RJ and Brummer T: Function, regulation and pathological roles of the Gab/DOS docking proteins. Cell Commun Signal. 7:222009. View Article : Google Scholar : PubMed/NCBI

35 

Ding CB, Yu WN, Feng JH and Luo JM: Structure and function of Gab2 and its role in cancer (Review). Mol Med Rep. 12:4007–4014. 2015.PubMed/NCBI

36 

Chen Y, Liu Q, Wu M, Li M, Ding H, Shan X, Liu J, Tao T, Ni R and Chen X: GAB2 promotes cell proliferation by activating the ERK signaling pathway in hepatocellular carcinoma. Tumour Biol. 37:11763–11773. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Duckworth C, Zhang L, Carroll SL, Ethier SP and Cheung HW: Overexpression of GAB2 in ovarian cancer cells promotes tumor growth and angiogenesis by upregulating chemokine expression. Oncogene. 35:4036–4047. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Ding C, Luo J, Yu W, Gao S, Yang L, Chen C and Feng J: Gab2 is a novel prognostic factor for colorectal cancer patients. Int J Clin Exp Pathol. 8:2779–2786. 2015.PubMed/NCBI

39 

Fleuren ED, O'Toole S, Millar EK, McNeil C, Lopez-Knowles E, Boulghourjian A, Croucher DR, Schramek D, Brummer T, Penninger JM, et al: Overexpression of the oncogenic signal transducer Gab2 occurs early in breast cancer development. Int J Cancer. 127:1486–1492. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Shi L, Sun X, Zhang J, Zhao C, Li H, Liu Z, Fang C, Wang X, Zhao C, Zhang X, et al: Gab2 expression in glioma and its implications for tumor invasion. Acta Oncol. 52:1739–1750. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Tian LQ, Liu EQ, Zhu XD, Wang XG, Li J and Xu GM: MicroRNA-197 inhibits cell proliferation by targeting GAB2 in glioblastoma. Mol Med Rep. 13:4279–4288. 2016.PubMed/NCBI

Related Articles

Journal Cover

February-2017
Volume 37 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma J, Yu J, Liu J, Yang X, Lou M, Liu J, Feng F, Ji P and Wang L: MicroRNA-302a targets GAB2 to suppress cell proliferation, migration and invasion of glioma. Oncol Rep 37: 1159-1167, 2017
APA
Ma, J., Yu, J., Liu, J., Yang, X., Lou, M., Liu, J. ... Wang, L. (2017). MicroRNA-302a targets GAB2 to suppress cell proliferation, migration and invasion of glioma. Oncology Reports, 37, 1159-1167. https://doi.org/10.3892/or.2016.5320
MLA
Ma, J., Yu, J., Liu, J., Yang, X., Lou, M., Liu, J., Feng, F., Ji, P., Wang, L."MicroRNA-302a targets GAB2 to suppress cell proliferation, migration and invasion of glioma". Oncology Reports 37.2 (2017): 1159-1167.
Chicago
Ma, J., Yu, J., Liu, J., Yang, X., Lou, M., Liu, J., Feng, F., Ji, P., Wang, L."MicroRNA-302a targets GAB2 to suppress cell proliferation, migration and invasion of glioma". Oncology Reports 37, no. 2 (2017): 1159-1167. https://doi.org/10.3892/or.2016.5320