microRNA‑188 acts as a tumour suppressor in glioma by directly targeting the IGF2BP2 gene

Retraction in: /10.3892/mmr.2022.12826

  • Authors:
    • Li Ding
    • Ling Wang
    • Feng Guo
  • View Affiliations

  • Published online on: September 7, 2017     https://doi.org/10.3892/mmr.2017.7433
  • Pages: 7124-7130
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioma is the most common and aggressive human brain tumour and accounts for ~35‑61% of intracranial tumours. Despite considerable advances in treatments for glioma, the prognosis for patients with this disease remains unsatisfactory. MicroRNAs (miRNAs of miRs) are small regulatory RNA molecules that have been identified as being involved in the initiation and progression of human cancers, and represent novel therapeutic targets for anticancer treatments. The dysregulation of miR‑188 has been reported in various kinds of human cancer. However, its expression pattern, biological roles and potential mechanism in glioma remain unknown. Expression levels of miR‑188 in glioma tissues and cell lines were detected through reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). Cell Counting Kit-8 assays and migration and invasion assays were used to explore the effects of miR‑188 on the proliferation, migration and invasion of glioma cells, respectively. Bioinformatics analysis and luciferase reporter assays were performed to examine insulin‑like growth factor 2 mRNA-binding protein 2 (IGF2BP2) as a target gene of miR‑188. RT‑qPCR and Spearman's correlation analysis were then performed to measure IGF2BP2 mRNA expression in clinical glioma tissues and its correlation with miR‑188 expression. The regulatory effect of miR‑188 on IGF2BP2 expression was also investigated through RT‑qPCR and western blotting analysis. Finally, the biological roles of IGF2BP2 in glioma cells were assessed. miR‑188 levels were significantly reduced in glioma tissues and cell lines compared with adjacent normal tissues and normal human astrocytes, respectively. In addition, miR‑188 overexpression suppressed cell proliferation, migration and invasion of glioma. The present study identified IGF2BP2 as a direct target of miR‑188 in glioma, and IGF2BP2 under‑expression served tumour‑suppressive roles in glioma growth and metastasis. Thus, miR‑188 had a similar role in glioma by inhibiting the action of its downstream target, IGF2BP2. Therefore, miR‑188 may be a potential therapeutic target for the prevention and treatment of patients with glioma.

Introduction

Glioma is the most common and aggressive type of human brain tumour, accounting for ~35–61% of intracranial tumours (1). Gliomas can be divided into subtypes according to the type of cell with which they share histological features, for example astrocytomas share features with astrocytes, then further subdivided by appearance/behaviour: The subtypes of astrocytoma are pilocytic astrocytomas, diffuse astrocytomas, anaplastic astrocytomas and glioblastoma multiforme (2). Among these subtypes, glioblastoma multiforme is the most severe and malignant form (3). The pathogenesis of gliomas currently remains elusive, but is strongly associated with factors including tumour origin, genetic factors, biochemical environment, ionising radiation, nitrous compounds, air pollution, bad living habits and infection (4). Despite considerable advancements in glioma treatments, including surgery, adjuvant chemotherapy and radiotherapy, the prognosis for patients with glioma remains unsatisfactory, with a 5-year survival rate of only 9.8% (5). The median survival time of patients with gliomas is ~12 months (6). Evidence indicates that gliomas exhibit progressive overgrowth and diffused invasion, and these characteristics contribute to the poor prognosis of gliomas (7). The underlying molecular mechanisms involved in the occurrence and development of glioma remains unclear (8). Thus, investigation of the possible mechanisms underlying glioma progression and development of novel therapeutic treatments for patients with this disease are necessary.

Differential expression of specific microRNAs (miRNAs or miRs) in gliomas compared with normal tissues has previously been reported (911). miRNAs are single-stranded, highly conserved non-coding RNA molecules (19–25 nucleotides in length) that regulate the expression of >60% of human genes (12). They regulate gene expression at the transcriptional and post-transcriptional levels by targeting the 3′-untranslated regions (3′-UTRs) of their target genes and mediate mRNA cleavage or translation inhibition (13). Due to their repressive action, miRNAs are important in several carcinogenic processes, including cell cycle and proliferation (14), apoptosis (15), gene methylation (16), myeloid differentiation (17), haematopoietic stem cell maintenance (18) and progenitor self-renewal (19). Thus, abnormally expressed miRNAs may serve as a signal of tumourigenesis and tumour development in human malignancies (20). In addition, some reports have indicated both tumour-suppressive and oncogenic roles of miRNAs in human cancers such as glioma (2123). For example, miR-133b expression has been demonstrated to be reduced in glioma tissues compared with normal brain tissues, leading to increased proliferation and invasion of tumour cells through the resulting overexpression of its target, sirtuin 1 (SIRT1) (24). Conversely, miR-130b is highly expressed in glioma tissues compared with normal brain tissues and acts as an oncogene through negative regulation of peroxisome proliferator-activated receptor-γ (25). miRNAs have, therefore, been established as targets for anticancer treatments.

miR-188 has been recognised as a tumour-suppressive miRNA that is downregulated in various types of cancer (2628). However, miR-188 has not previously been recognised as such in gliomas. The present study aimed to investigate the expression pattern, biological roles and potential mechanism by which miR-188 dysregulation is correlated with glioma. miR-188 was found to be significantly downregulated in glioma tissues and cell lines compared with normal tissues and cell lines, and restoration of miR-188 expression suppressed glioma cell proliferation, migration and invasion. In addition, insulin like-growth factor 2 mRNA-binding protein 2 (IGF2BP2) was validated as a direct target of miR-188 in glioma.

Materials and methods

Clinical specimens

Glioma samples and adjacent normal tissues were collected from 19 patients at the Department of Neurosurgery, Linyi People's Hospital (Linyi, China) between June 2013 and December 2015. All patients with glioma were newly diagnosed and were not undergoing adjuvant chemotherapy and radiotherapy. All fresh tissues were immediately frozen in liquid nitrogen and stored at −80°C. The present study was approved by the hospital's Protection of Human Subjects Committee, and informed consent was obtained from all patients.

Cell culture and transfection

Human glioma cell lines (U87, U251, U118, LN229 and LN18) and HEK293T cell line were all obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA). Primary normal human astrocytes (NHA) were purchased from ScienCell Research Laboratories (Carlsbad, CA, USA). All cell lines were propagated in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin G and 100 µg/ml streptomycin (all from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C in a humidified air atmosphere of 5% CO2.

Mature miR-188 mimics, miRNA mimics negative control (miR-NC), siRNA targeting IGF2BP2 (si-IGF2BP2) and negative control (si-NC) were obtained from GenePharma (Shanghai, China). The miR-188 mimics sequence was: 5′-CAUCCCUUGCAUGGUGGAGGG-3′; the miR-NC sequence was: 5′-UUCUCCGAACGUGUCACGUTT-3′; the si-IGF2BP2 sequence was: 5′-GCUGGAGCUUCAAUUAAGATT-3′; and the si-NC sequence was: 5′-UUCUCCGAACGUGUCACGUTT-3′. For functional analysis, cells were transfected with miR-188 mimics, miR-NC, si-IGF2BP2 or si-NC using Lipofectamine 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) in accordance with the manufacturer's instructions.

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from tissues and cell lines using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Total RNA concentration was determined using a NanoDrop ND-1000 spectrophotometer (Thermo Fisher Scientific, Inc., Wilmington, DE, USA). To detect the expression of miR-188 and IGF2BP2 mRNA, the extracted RNA was quantified and then cDNA was synthesised using a Moloney Murine Leukemia Virus Reverse Transcription system (Promega Corporation, Madison, WI, USA). qPCR was performed in an Applied Biosystems 7500 Real-time PCR system (Thermo Fisher Scientific, Inc., Waltham, MA, USA) and a SYBR Premix Ex Taq™ kit (Takara Biotechnology Co., Ltd., Dalian, China) was used according to the manufacturer's instructions. U6 and GADPH were employed as internal controls for the miR-188 and IGF2BP2 mRNA expression, respectively. Each sample was analysed in triplicate. The data were calculated through relative quantification (2−ΔΔCq) (29).

Cell Counting Kit-8 (CCK-8) assay

The proliferation of glioma cells was examined using the CCK-8 (Dojindo Molecular Technologies, Inc., Kumamoto, Japan) assay according to the manufacturer's instructions. The cells were seeded in 96-well culture plates at a density of 4×103 cells per well 24 h after transfection. After 24, 48, 72 or 96 h treatment, 10 µl of CCK-8 solution was added to each well and the cells were incubated at 37°C for a further 2 h. The absorbance at 450 nm was obtained using an ELISA reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA). All the experiments were performed in triplicate.

Migration and invasion assays

For the migration assay, 5×104 transfected cells were prepared in FBS-free DMEM medium. Cells were then seeded in the upper chamber of a transwell chamber insert (Corning Inc., Corning, NY, USA). For the invasion assay, the transwell chamber insert was precoated with Matrigel (BD Biosciences, San Jose, CA, USA). The transfected cells in the FBS-free DMEM medium were placed in the upper chamber. Approximately 500 µl DMEM with 20% FBS was added in the lower chamber of Transwell chamber insert. Following incubation at 37°C in a humidified air atmosphere of 5% CO2, nonmigrated and noninvaded cells were removed thoroughly by using cotton-tipped swabs. The migrated and invaded cells in the lower surface of the membrane were fixed in 4% paraformaldehyde and then stained with 0.5% crystal violet before they were air dried. A total of 5 randomly selected fields were then examined under a light microscope at a magnification of 200×.

Western blot analysis

Cells were lysed with cold radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Haimen, China) containing protease and phosphatase inhibitors. The protein was extracted by centrifugation at 12,000 × g for 20 min at 4°C, and quantified with a bicinchoninic acid quantification kit (Beyotime Institute of Biotechnology). Equal amounts of the proteins (30 µg) were then separated by 10% sodium dodecyl sulphate-polyacrylamide gel electrophoresis and then transferred to polyvinylidene difluoride membranes (PVDF; EMD Millipore, Billerica, MA, USA). The membranes were blocked with 5% non-fat dry milk in Tris-buffered saline containing 0.05% Tween-20 (TBST) for 1 h at room temperature. The membranes were then incubated with mouse anti-human monoclonal IGF2BP2 antibody (cat. no. sc-377014, 1:1,000 dilution; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) and mouse anti-human monoclonal GADPH antibody (cat. no. sc-137179, 1:1,000 dilution; Santa Cruz Biotechnology, Inc.) at 4°C overnight. Following 3 washes in TBST, the membranes were incubated for 1 h with goat anti-mouse immunoglobulin G conjugated to horseradish peroxidase (sc-2005, 1:5,000; Santa Cruz Biotechnology, Inc.) for 1 h at room temperature. The protein bands were visualised using an enhanced chemiluminescence kit (Pierce; Thermo Fisher Scientific, Inc.) and then analysed using Quantity One software version 4.62 (Bio-Rad Laboratories, Inc.).

Bioinformatic analysis

Bioinformatic analysis was performed to predict the putative targets of miR-185 using TargetScan (Release 6.0, November 2011; http://www.targetscan.org/) (30).

Luciferase reporter assay

Luciferase reporter assays were used to determine whether IGF2BP2 is a direct target of miR-188. The reporter plasmids containing the wild-type (WT) 3′UTR (pGL3-IGF2BP2-WT-3′UTR) and mutant (MUT) 3′UTR (pGL3-IGF2BP2-MUT-3′UTR) were synthesised by Shanghai GenePharma Co., Ltd., (Shanghai, China). For the luciferase reporter assay, the HEK293T cells were co-transfected with pGL3-IGF2BP2-WT-3′UTR or pGL3-IGF2BP2-MUT-3′UTR and the miR-188 mimic or miR-NC using Lipofectamine 2000 according to the manufacturer's instructions. After 48 h of transfection, the firefly and Renilla luciferase activities were determined by using a Dual Luciferase Assay kit (Promega Corporation) according to the manufacturer's instructions. Each assay was performed in triplicate and then replicated thrice.

Statistical analysis

Data are presented as the mean ± standard deviation or as box plots using SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA). Statistical analyses of the differences between groups were performed by with Student's t-test (for two groups) or one-way analysis of variance (for multiple comparisons) using SPSS 13.0 software (SPSS, Inc., Chicago, IL, USA). SNK analysis was used as a post hoc test following analysis of variance. Correlation analysis between miR-188 and IGF2BP2 mRNA expression was analyzed by the Spearman's correlation analysis with SPSS software version 17.0 (SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

miR-188 is downregulated in glioma

miR-188 expression was detected in 19 pairs of glioma tissues and adjacent normal tissues. In this study, miR-188 expression was significantly downregulated in the glioma tissues compared with expression in the adjacent normal tissues (P<0.05; Fig. 1A). Expression of miR-188 was also observed to be significantly reduced in all 5 glioma cell lines examined, compared with NHA (P<0.05; Fig. 1B).

miR-188 reduces cell proliferation in glioma cell lines

The effects of exogenous miR-188 on glioma were then investigated in the 2 cell lines with the lowest endogenous miR-188 levels in Fig. 1B, U87 and U251 cells. Cells transfected with miR-188 mimics were demonstrated to exhibit significantly higher levels of miR-188 than cells transfected with miR-NC (P<0.05; Fig. 2A). CCK-8 assays were then conducted to evaluate the effect of miR-188 on glioma cell proliferation. As demonstrated in Fig. 2B, cell proliferation was significantly reduced in cells transfected with miR-188 mimics compared with cells transfected with miR-NC (P<0.05; Fig. 2B). These results indicated that miR-188 may be a negative regulator of glioma cell proliferation.

miR-188 decreases cell migration and invasion in glioma

Migration and invasion assays were used to assess the effects of miR-188 overexpression on glioma cell metastasis. As demonstrated in the migration assay, overexpression of miR-188 resulted in a significant decrease in the number of migrated U87 and U251 cells compared with miR-NC (P<0.05; Fig. 3A). The invasion assay also revealed significantly reduced invasive capabilities in cells transfected with miR-188 mimics compared with cells transfected with miR-NC (P<0.05; Fig. 3B). Thus, miR-188 may serve a role in the inhibition of glioma metastasis.

IGF2BP2 is a direct target gene of miR-188

To identify the target of miR-188, bioinformatics prediction was adopted to analyse its potential target genes. This bioinformatics analysis revealed 224 highly conserved sites and 91 poorly conserved sites. Among these putative targets, cyclin D3 (CCND3) and CCNA2 have previously been identified as direct targets of miR-188 in nasopharyngeal carcinoma (31), as has SIX homeobox 1 (SIX1) in oral squamous cell carcinoma (32) and fibroblast growth factor 5 (FGF5) in hepatocellular carcinoma (28). In the present study, IGF2BP2 was selected for further analysis since it has previously been reported to be upregulated in glioma tissues (33) and is involved in the formation and progression of glioma (33). Luciferase reporter assays were performed using plasmids carrying the WT and MUT sequences demonstrated in Fig. 4A. The data indicated that overexpression of miR-188 significantly inhibited the luciferase activity in cells harbouring the construct containing the WT IGF2BP2 3′UTR, but not that containing the MUT IGF2BP2 3′UTR (Fig. 4B).

IGF2BP2 mRNA levels in the clinical samples were then analysed, revealing that significantly higher expression of IGF2BP2 mRNA was observed in glioma tissues than in the adjacent normal tissues (P<0.05; Fig. 4C). The correlation between miR-188 expression and IGF2BP2 mRNA was then analysed through Spearman's correlation analysis. The results revealed a negative correlation between the miR-188 and IGF2BP2 mRNA levels in glioma tissues (R=−0.6482, P=0.0027; Fig. 4D).

RT-qPCR and western blot analyses were then performed to examine whether mRNA and protein expression levels of IGF2BP2, respectively, were downregulated with increased expression of miR-188. As revealed in Fig. 4E and F, respectively, IGF2BP2 mRNA and protein expression levels were decreased in U87 and U251 cells following transfection with miR-188 mimics, compared with miR-NC. Overall, these results indicated that IGF2BP2 is a direct target gene of miR-188.

Downregulation of IGF2BP2 inhibits cell proliferation, migration and invasion in glioma

To clarify the important roles of miR-188 in the inhibition of cell growth and metastasis in glioma via the negative regulation of IGF2BP2, the biological roles of IGF2BP2 were investigated in U87 and U251 cells. As presented in Fig. 5A, IGF2BP2 protein expression was reduced in cells following transfection with si-IGF2BP2, compared with expression in cells transfected with si-NC. CCK-8 assays were then performed, demonstrating that cell proliferation was significantly reduced in cells transfected with si-IGF2BP2 compared with cells transfected with si-NC (Fig. 5B). Furthermore, migration and invasion assays demonstrated that reduced expression of IGF2BP2 resulted in reduced migration and invasion, with significantly fewer migrated and invaded cells in cells transfected with si-IGF2BP2 compared with cells transfected with si-NC (Fig. 5C and D, respectively). These results suggested that IGF2BP2 serves important tumour-suppressive roles in glioma growth and metastasis similar to the roles of miR-188 in glioma. Thus, IGF2BP2 is indicated to be a direct downstream target of miR-188 in glioma.

Discussion

miRNAs are small regulatory RNA molecules which have been identified as serving roles in the initiation and progression of human cancers and may represent novel therapeutic targets for anticancer treatments (34). In the present study, low miR-188 expression was observed in glioma tissues and cell lines, providing, to the best of our knowledge, the first evidence that under-expression of miR-188 is associated with the carcinogenesis and progression of glioma. The effects of miR-188 on the proliferation, migration and invasion of glioma cells were then explored. The results indicated that miR-188 overexpression suppressed cell proliferation, migration and invasion of glioma. In addition, the present study identified IGF2BP2 as a direct target of miR-188 in glioma. IGF2BP2 under-expression was demonstrated to serve a tumour-suppressive role in glioma growth and metastasis, similar to the effects of miR-188 overexpression in glioma. These results suggested that miR-188 may contribute to glioma occurrence and development and warrants further examination in the context of molecular-targeted therapy. To the best of our knowledge, the present study is the first to investigate the expression pattern, biological roles and potential mechanism of miR-188 in glioma.

Aberrant expression of miR-188 has been reported in numerous types of human cancers. For example, miR-188 is downregulated in normal acute myeloid leukemia. High miR-188 expression correlates with increased overall survival and event-free survival of patients with normal acute myeloid leukemia (26). Zhang et al (27) demonstrated that miR-188 expression is decreased in metastatic prostate cancer, and that its low expression was an independent prognostic factor for poor overall and biochemical recurrence-free survival of patients with metastatic prostate cancer. Fang et al (28) reported that miR-188 levels are reduced in hepatocellular carcinoma, and that low expression of miR-188 is significantly associated with multiple nodules, microvascular invasion, overall and disease-free survival. In oral squamous cell carcinoma, low expression of miR-188 has been observed in tumour tissues compared with matched normal tissues (32). These findings suggest that miR-188 could be a diagnostic and prognostic biomarker for human cancer.

Abnormally expressed miR-188 has also been demonstrated to be involved in the carcinogenesis and progression of human cancers. Wu et al (31) demonstrated that upregulation of miR-188 suppresses cell proliferation, colony formation and G1/S cell cycle transition in human nasopharyngeal carcinoma, and directly target CCN and cyclin dependent kinase (CDK) complexes, including CCND1, CCND3, CCNE1, CCNA2, CDK4 and CDK2 (31). In prostate cancer, increased levels of miR-188 inhibits in vitro cell growth and metastasis and decreases tumour growth and metastasis in vivo through the negative regulation of lysosomal protein transmembrane 4β (27). In hepatocellular carcinoma, the increased levels of miR-188 decreased cell proliferation and metastasis via blockade of FGF5 (28). Wang et al (32) demonstrated that overexpression of miR-188 suppresses cell proliferation, cell cycle progression and invasion of oral squamous cell carcinoma through the downregulation of SIX1.

In addition, the mechanisms by which miR-188 dysregulation is correlated with cell proliferation, migration and invasion in glioma were investigated. Bioinformatics analysis was performed to predict the potential target genes of miR-188 by using TargetScan. IGF2BP2 was identified to be a putative target of miR-188; a highly conserved miR-188 binding site was identified in the 3′UTR. Subsequently, luciferase reporter assay results revealed that upregulation of miR-188 caused a significant reduction in the luciferase activities of the WT IGF2BP2 3′UTR reporter but no effect was observed with the MUT IGF2BP2 3′UTR reporter. In addition, RT-qPCR analysis further revealed that IGF2BP2 was highly expressed in glioma tissues compared with adjacent normal tissues. In clinical glioma tissues, IGF2BP2 expression was inversely correlated with miR-188 expression patterns. Furthermore, the upregulation of miR-188 reduced IGF2BP2 expression at both mRNA and protein levels in glioma cell lines. IGF2BP2 was also demonstrated to serve a tumour suppressive role in glioma growth and metastasis, similarly to the effects of miR-188 in glioma. Thus, IGF2BP2 is a direct downstream target of miR-188 in glioma. These results indicated that miR-188 directly downregulated IGF2BP2 expression by binding to the 3′UTR of the IGF2BP2 gene.

The insulin-like growth factor 2 mRNA-binding protein family includes IGF2BP1, IGF2BP2 and IGF2BP3 (35). IGF2BP2, also known as IMP2, has been reported to be dysregulated in several human cancers, including colorectal cancer (36), colon cancer (37), oesophageal adenocarcinoma (38) and breast cancer (39). Mu et al (33) reported that IGF2BP2 expression is increased in glioblastoma tissues compared with normal brain tissues. Results from the database search also revealed that IGF2BP2 was upregulated in glioma tissues. Functional assays demonstrated that downregulation of IGF2BP2 represses the proliferation, migration and invasion ability and epithelial-mesenchymal transition in glioblastomas cells (33). IGF2BP2 may, therefore, be a useful therapeutic target for the therapy of patients with glioma.

In conclusion, the present study provides the first evidence, to the best of our knowledge, to indicate downregulation of miR-188 in both glioma tissues and cell lines. Ectopic expression of miR-188 suppressed cell growth and metastasis of glioma. In addition, IGF2BP2 was validated as a direct target gene of miR-188. The miR-188/IGF2BP2 axis may provide novel insights into the pathogenesis of glioma and could be investigated as a potential therapeutic target for the treatment of this disease. Future study is needed to investigate the upstream regulation mechanism of miR-188 in glioma, and explore whether the potential of miR-188 may be fully realised in glioma.

References

1 

Di Stefano AL, Enciso-Mora V, Marie Y, Desestret V, Labussière M, Boisselier B, Mokhtari K, Idbaih A, Hoang-Xuan K, Delattre JY, et al: Association between glioma susceptibility loci and tumour pathology defines specific molecular etiologies. Neuro Oncol. 15:542–547. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Zhang C, Bao Z, Zhang W and Jiang T: Progress on molecular biomarkers and classification of malignant gliomas. Front Med. 7:150–156. 2013. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Zhu GY, Shi BZ and Li Y: FoxM1 regulates Sirt1 expression in glioma cells. Eur Rev Med Pharmacol Sci. 18:205–211. 2014.PubMed/NCBI

5 

Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al: Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 10:459–466. 2009. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Johnson DR and Galanis E: Incorporation of prognostic and predictive factors into glioma clinical trials. Curr Oncol Rep. 15:56–63. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Hou SX, Ding BJ, Li HZ, Wang L, Xia F, Du F, Liu LJ, Liu YH, Liu XD, Jia JF, et al: Identification of microRNA-205 as a potential prognostic indicator for human glioma. J Clin Neurosci. 20:933–937. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Yan Y, Peng Y, Ou Y and Jiang Y: MicroRNA-610 is downregulated in glioma cells, and inhibits proliferation and motility by directly targeting MDM2. Mol Med Rep. 14:2657–2664. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Guo H, Nan Y, Zhen Y, Zhang Y, Guo L, Yu K, Huang Q and Zhong Y: miRNA-451 inhibits glioma cell proliferation and invasion by downregulating glucose transporter 1. Tumour Biol. 37:13751–13761. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Wang G, Li Z, Tian N, Han L, Fu Y, Guo Z and Tian Y: miR-148b-3p inhibits malignant biological behaviors of human glioma cells induced by high HOTAIR expression. Oncol Lett. 12:879–886. 2016.PubMed/NCBI

12 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Fabian MR, Sonenberg N and Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 79:351–379. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Pulikkan JA, Dengler V, Peramangalam PS, Zada AA Peer, Müller-Tidow C, Bohlander SK, Tenen DG and Behre G: Cell-cycle regulator E2F1 and microRNA-223 comprise an autoregulatory negative feedback loop in acute myeloid leukemia. Blood. 115:1768–1778. 2010. View Article : Google Scholar : PubMed/NCBI

15 

He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, Xue W, Zender L, Magnus J, Ridzon D, et al: A microRNA component of the p53 tumour suppressor network. Nature. 447:1130–1134. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Garzon R, Liu S, Fabbri M, Liu Z, Heaphy CE, Callegari E, Schwind S, Pang J, Yu J, Muthusamy N, et al: MicroRNA-29b induces global DNA hypomethylation and tumor suppressor gene reexpression in acute myeloid leukemia by targeting directly DNMT3A and 3B and indirectly DNMT1. Blood. 113:6411–6418. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Johnnidis JB, Harris MH, Wheeler RT, Stehling-Sun S, Lam MH, Kirak O, Brummelkamp TR, Fleming MD and Camargo FD: Regulation of progenitor cell proliferation and granulocyte function by microRNA-223. Nature. 451:1125–1129. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Dickstein J, Senyuk V, Premanand K, Laricchia-Robbio L, Xu P, Cattaneo F, Fazzina R and Nucifora G: Methylation and silencing of miRNA-124 by EVI1 and self-renewal exhaustion of hematopoietic stem cells in murine myelodysplastic syndrome. Proc Natl Acad Sci USA. 107:9783–9788. 2010; View Article : Google Scholar : PubMed/NCBI

19 

Guo S, Lu J, Schlanger R, Zhang H, Wang JY, Fox MC, Purton LE, Fleming HH, Cobb B, Merkenschlager M, et al: MicroRNA miR-125a controls hematopoietic stem cell number. Proc Natl Acad Sci USA. 107:14229–14234. 2010; View Article : Google Scholar : PubMed/NCBI

20 

Nana-Sinkam P and Croce CM: MicroRNAs in diagnosis and prognosis in cancer: What does the future hold? Pharmacogenomics. 11:667–669. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Chen Q, Guo W, Zhang Y, Wu Y and Xiang J: miR-19a promotes cell proliferation and invasion by targeting RhoB in human glioma cells. Neurosci Lett. 628:161–166. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Zhu Y, Zhao H, Feng L and Xu S: MicroRNA-217 inhibits cell proliferation and invasion by targeting Runx2 in human glioma. Am J Transl Res. 8:1482–1491. 2016.PubMed/NCBI

23 

Duan J, Zhou K, Tang X, Duan J and Zhao L: MicroRNA-34a inhibits cell proliferation and induces cell apoptosis of glioma cells via targeting of Bcl-2. Mol Med Rep. 14:432–438. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Li C, Liu Z, Yang K, Chen X, Zeng Y, Liu J, Li Z and Liu Y: miR-133b inhibits glioma cell proliferation and invasion by targeting Sirt1. Oncotarget. 7:36247–36254. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Gu JJ, Zhang JH, Chen HJ and Wang SS: MicroRNA-130b promotes cell proliferation and invasion by inhibiting peroxisome proliferator-activated receptor-γ in human glioma cells. Int J Mol Med. 37:1587–1593. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Jinlong S, Lin F, Yonghui L, Li Y and Weidong W: Identification of let-7a-2-3p or/and miR-188-5p as prognostic biomarkers in cytogenetically normal acute myeloid leukemia. PLoS One. 10:e01180992015. View Article : Google Scholar : PubMed/NCBI

27 

Zhang H, Qi S, Zhang T, Wang A, Liu R, Guo J, Wang Y and Xu Y: miR-188-5p inhibits tumour growth and metastasis in prostate cancer by repressing LAPTM4B expression. Oncotarget. 6:6092–6104. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Fang F, Chang RM, Yu L, Lei X, Xiao S, Yang H and Yang LY: MicroRNA-188-5p suppresses tumor cell proliferation and metastasis by directly targeting FGF5 in hepatocellular carcinoma. J Hepatol. 63:874–885. 2015. View Article : Google Scholar : PubMed/NCBI

29 

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

30 

Garcia DM, Baek D, Shin C, Bell GW, Grimson A and Bartel DP: Weak seed-pairing stability and high target-site abundance decrease the proficiency of lsy-6 and other microRNAs. Nat Struct Mol Biol. 18:1139–1146. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Wu J, Lv Q, He J, Zhang H, Mei X, Cui K, Huang N, Xie W, Xu N and Zhang Y: MicroRNA-188 suppresses G1/S transition by targeting multiple cyclin/CDK complexes. Cell Commun Signal. 12:662014. View Article : Google Scholar : PubMed/NCBI

32 

Wang L and Liu H: microRNA-188 is downregulated in oral squamous cell carcinoma and inhibits proliferation and invasion by targeting SIX1. Tumour Biol. 37:4105–4113. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Mu Q, Wang L, Yu F, Gao H, Lei T, Li P, Liu P, Zheng X, Hu X, Chen Y, et al: Imp2 regulates GBM progression by activating IGF2/PI3K/Akt pathway. Cancer Biol Ther. 16:623–633. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Shi L, Wang Z, Sun G, Wan Y, Guo J and Fu X: miR-145 inhibits migration and invasion of glioma stem cells by targeting ABCG2. Neuromolecular Med. 16:517–528. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Kanzaki A, Kudo M, Ansai S, Peng WX, Ishino K, Yamamoto T, Wada R, Fujii T, Teduka K, Kawahara K, et al: Insulin-like growth factor 2 mRNA-binding protein-3 as a marker for distinguishing between cutaneous squamous cell carcinoma and keratoacanthoma. Int J Oncol. 48:1007–1015. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Ye S, Song W, Xu X, Zhao X and Yang L: IGF2BP2 promotes colorectal cancer cell proliferation and survival through interfering with RAF-1 degradation by miR-195. FEBS Lett. 590:1641–1650. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Liu W, Li Z, Xu W, Wang Q and Yang S: Humoral autoimmune response to IGF2 mRNA-binding protein (IMP2/p62) and its tissue-specific expression in colon cancer. Scand J Immunol. 77:255–260. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Barghash A, Golob-Schwarzl N, Helms V, Haybaeck J and Kessler SM: Elevated expression of the IGF2 mRNA binding protein 2 (IGF2BP2/IMP2) is linked to short survival and metastasis in esophageal adenocarcinoma. Oncotarget. 7:49743–49750. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Liu W, Li Y, Wang B, Dai L, Qian W and Zhang JY: Autoimmune response to IGF2 mRNA-binding protein 2 (IMP2/p62) in breast cancer. Scand J Immunol. 81:502–507. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ding L, Wang L and Guo F: microRNA‑188 acts as a tumour suppressor in glioma by directly targeting the IGF2BP2 gene Retraction in /10.3892/mmr.2022.12826. Mol Med Rep 16: 7124-7130, 2017
APA
Ding, L., Wang, L., & Guo, F. (2017). microRNA‑188 acts as a tumour suppressor in glioma by directly targeting the IGF2BP2 gene Retraction in /10.3892/mmr.2022.12826. Molecular Medicine Reports, 16, 7124-7130. https://doi.org/10.3892/mmr.2017.7433
MLA
Ding, L., Wang, L., Guo, F."microRNA‑188 acts as a tumour suppressor in glioma by directly targeting the IGF2BP2 gene Retraction in /10.3892/mmr.2022.12826". Molecular Medicine Reports 16.5 (2017): 7124-7130.
Chicago
Ding, L., Wang, L., Guo, F."microRNA‑188 acts as a tumour suppressor in glioma by directly targeting the IGF2BP2 gene Retraction in /10.3892/mmr.2022.12826". Molecular Medicine Reports 16, no. 5 (2017): 7124-7130. https://doi.org/10.3892/mmr.2017.7433