Open Access

MicroRNA‑21 promotes migration and invasion of glioma cells via activation of Sox2 and β‑catenin signaling

Retraction in: /10.3892/mmr.2023.12982

  • Authors:
    • Guoxuan Luo
    • Wentao Luo
    • Xiaohui Sun
    • Jinzhi Lin
    • Mo Wang
    • Yang Zhang
    • Weishi Luo
    • Yong Zhang
  • View Affiliations

  • Published online on: November 28, 2016     https://doi.org/10.3892/mmr.2016.5971
  • Pages: 187-193
  • Copyright: © Luo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

The expression of microRNA 21 (miR-21) has been reported to be upregulated in various types of cancer, including malignant gliomas. However, its functions and mechanisms in glioma remain to be fully elucidated. The present study established miRNA‑21 overexpression and knockdown cell lines using SRY‑box 2 (Sox2) small interfering RNA (siRNA) to knockdown expression and Sox2 cDNA was cloned into pcDNA 3.1 mammalian expression vector for ectopic expression. BIO and XAV‑939 were used for β‑catenin signaling activation and knockdown, respectively. Transwell chambers were used to assay the capacity of cells to migrate. The present study determined that increased expression of miR‑21 significantly promoted the migration and invasion of glioma cells, which was accompanied by an upregulated expression of the Sox2 protein. Sox2 overexpression also promoted glioma cell migration and invasion, whereas Sox2 siRNA markedly reduced the miR‑21‑enhanced migration and invasion of glioma cells, indicating Sox2 may act as a crucial mediator of miR‑21 function. Furthermore, miR‑21 also upregulated the protein expression level of β‑catenin, whereas anti‑miR‑21 and Sox2 knockdown significantly reduced β‑catenin expression. BIO, a β‑catenin specific agonist, enhanced migration and invasion of glioma cells. XAV‑939, an inhibitor of β‑catenin signaling, markedly inhibited the migration and invasion of glioma cells, suggesting that β‑catenin may be associated with miR‑21‑ and Sox2‑induced invasion of glioma cells. Notably, BIO restored the migration and invasion potential of glioma cells, which were inhibited by Sox2 siRNA and anti‑miR‑21. These findings indicated that β‑catenin may be an important downstream mediator of miR‑21 and Sox2. Therefore, the present study identified the miR‑21/Sox2/β‑catenin signaling pathway, which may regulate the migration and invasion of human glioma cells.

Introduction

Malignant gliomas are one of the most common primary malignant brain tumors, with an annual incidence in China of 5.26 per 100,000 individuals. These tumors are frequently associated with a poor prognosis and low quality of life in patients (1). Hypoxia is a major feature of the solid tumor microenvironment and has been associated with tumor progression and poor clinical outcome. Previous studies have demonstrated that pseudopalisades around necrotic foci in malignant gliomas are severely hypoxic, and secrete high levels of vascular endothelial growth factor (VEGF) by increasing the transcriptional activity of hypoxia-inducible factors 1 and 2 (HIF-1 and −2) (2,3). VEGF secretion leads to endothelial proliferation and angiogenesis, which is required for the development, progression, growth and metastasis of the tumor.

In addition, hypoxia effects the migration and invasion of glioma cells by modulation of the expression of extracellular gelatinases, such as matrix metalloproteinases and the urokinase-dependent plasminogen-activating cascade (2,3). The invasion of malignant glioma cells into the healthy regions the brain is a critical factor that limits current therapies for astrocytomas. However, the detailed molecular mechanisms underlying glioma cell migration and invasion remain to be elucidated (4,5).

microRNAs (miRNAs) are a class of non-protein-coding small RNAs and have been identified to be important in the coordination of cell differentiation, proliferation, apoptosis, metabolism and tumorigenic transformation (68). Significant effort has been made towards investigating the function and mechanism of microRNAs (9,10); however, the factors affecting the expression of miRNA transcripts remain to be elucidated. A previous study demonstrated that there are functional links between hypoxia and miRNA expression (11). Previous studies have identified that a specific spectrum of miRNAs may be induced in response to low oxygen levels and their overexpression may result in significant inhibition of proapoptotic signaling in a hypoxic environment, indicating the impact of these miRNAs on tumor growth. Notably, certain hypoxia-induced miRNAs have been identified to be overexpressed in a variety of human tumors, including malignant gliomas. Among these identified miRNAs, miRNA-21 (miR-21) was markedly upregulated in various cancer cells, particularly in gliomas. miR-21 has been determined to be upregulated in the majority of the human glioblastoma (GBM) specimens investigated and its expression level was correlated with the glioma grade (1115). Additionally, the downregulation of miR-21 in glioma cells lead to the reduction of their migratory and invasion abilities (16). However, the underlying molecular mechanism of how miRNA-21 affects glioma migration and invasion remains poorly understood.

The present study determined that the miR-21 overexpression significantly enhanced the migration and invasion of glioma cells, accompanied by SRY-box 2 (Sox2) upregulation and the activation of the β-catenin signaling pathway.

Materials and methods

Cell culture

Four human malignant glioma cell lines (U87, A172, T98 and U343) were obtained from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China) and cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with 10% fetal bovine serum (FBS; GE Healthcare Life Sciences, Logan, UT, USA), 100 U penicillin and 100 mg/ml streptomycin. The cells were maintained at 37°C in a humidified atmosphere containing 5% CO2.

Plasmids, miRNA, small interfering RNA (siRNA) and reagents

For miR-21 overexpression, the cells were transfected with a synthetic RNA duplex corresponding to mature miR-21. The sequences of the miR-21 and scrambled miRNA were as follows: miR-negative control (miR-NC), 5′-CATTAATGTCGGACAACTCAAT-3′ and miR-21, 5′-TCAACATCAGTCTGATAAGCTA-3′. For miR-21 inhibition, the cells were transfected with 50 nM anti-miR using Lipofectamine 2000 (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols as previously described (17). The sequences were as follows: Mismatch siRNA, 5′-TCTTCATGAGTCAGATTACCTA-3′ and anti-miR-21, 5′-TCAACATCAGTCTGATAAGCTA-3′. Sox2 knockdown in glioma cells was achieved using siRNA transfection with Lipofectamine 2000 according to the manufacturer's protocols. The sequence of Sox2 siRNA used was as follows: 5′-CCUGUGGUUACCUCUUCCCCCACU-3′ (18). Sox2 ectopic expression was achieved by subcloning Sox2 cDNA into pcDNA 3.1 mammalian expression vector (Invitrogen; Thermo Fisher Scientific, Inc.) as previously described (19). For the β-catenin knockdown using Lipofectamine 2000 according to the manufacturer's protocols, the following β-catenin siRNA sequence was used: 5′-AGCUGAUAUUGAUGGACAG-3′. The 6-bromoindirubin-3′-oxime (BIO), a β-catenin agonist, and XAV-939, an inhibitor of β-catenin were purchased from Sigma-Aldrich (Merck Millipore, Darmstadt, Germany). Cells activated with BIO were incubated with 1 µM BIO for 24 h. To detect β-catenin inhibition, the glioma cells were incubated with 1.0 µM XAV-939 for 8 h. These treatments were tested to validate their efficiency using quantitative polymerase chain reaction or western blotting. The western blotting was conducted as described below. The qPCR was performed using SYBR® Premix Ex Taq™ II on an ABI 7300 qPCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). Thermocycling conditions were as follows: 95°C for 10 min; and 40 cycles of 95°C for 15 sec and 60°C for 1 min. GAPDH served as an internal control. Normalization and fold changes were calculated using the 2−∆∆Cq method (20). All experiments involving transfected cells were conducted after 48 h.

Transwell migration and invasion assays

A Transwell assay was performed using chambers with polycarbonate filters with 8-µm pore size (Merck Millipore) coated with Matrigel on the upper side. The chambers were placed into a 24-well plate and the lower chamber was filled with DMEM containing 20% FBS. Glioma cells with different treatments (each group untreated, treated with miR-NC or miR-21) were harvested and 1.0×104 cells were placed in the upper chamber, then incubated for an additional 24 h. The cells that penetrated and attached to the bottom of the filter were identified using a crystal violet staining at room temperature for 20 min. The number invaded cells in each different treatment group was counted using microscopy (Leica DM IL LED; Leica Microsystems GmbH, Wetzlar, Germany and the average values of 6 randomly selected fields were used.

Immunofluorescence staining

Cells were cultured on glass coverslips until 80% confluent and then fixed with 4% formaldehyde solution. The cells were blocked with 5% bovine serum albumin (BSA; Sigma-Aldrich; Merck Millipore) for 30 min at room temperature. Next, the cells were incubated with rabbit polyclonal anti-Sox2 antibody (1:200; Wanlei Life Science Shenyang, China; cat. no. WL00982) overnight at 4°C and fluorescein isothiocyanate-labeled goat anti-rabbit antibody (1:100; Beyotime Institute of Biotechnology, Inc., Haimen, China; cat. no. A0423) for 1 h at 37°C. The cells were then subjected to laser scanning confocal microscopy using an Olympus FV1000S-SIM/IX81 (Olympus Corporation, Tokyo, Japan) following DAPI staining (1:1,000; Biosharp Biotech, Hefei, China).

Western blot analysis

Following exposure to the different treatments the cells were lysed with Cell Lysis Buffer (Cell Signaling Technology, Danvers, MA, USA) and 20 µg proteins were separated by SDS-PAGE following quantification using the ultraviolet absorption method. Proteins were transferred to PVDF membrane, blocked with 5% non-fat milk for 4 h at room temperature and then incubated with anti-Sox2 primary antibody (1:2,000) and anti-β-actin (1:1,500; Sigma-Aldrich; Merck Millipore; cat. no. C2206) overnight at 4°C. Following a 4 h incubation at room temperature with horseradish peroxidase-labeled secondary antibody (Beyotime Institute of Biotechnology, Inc.; cat. no. A0239) the specific proteins were detected using enhanced chemiluminescence reagent (7 Sea Biotech, Shanghai, China).

Intracellular flow cytometry analysis

Single cell suspensions in phosphate-buffered saline containing 1% BSA were obtaining by trypsinization following the application of the different treatments. The cells were fixed and permeabilized in Intracellular Fixation & Permeabilization Buffer (plus Brefeldin A; eBioscience, Inc., San Diego, CA, USA). For β-catenin expression detection, freshly harvested cells were incubated with a β-catenin antibody (1:200) according to the manufacturer's protocol and analyzed by flow cytometry followed by CellQuest Pro 4.0 software (BD Biosciences, Franklin Lakes, NJ, USA) for β-catenin expression using relative fluorescence intensity in each group compared with the untreated cells. A total of 30,000 cells were counted for each sample.

Statistical analysis

Data are expressed as the mean ± standard error. Statistical analysis of differences among groups was performed using analysis of variance, followed by Dunnett's post-test. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-21 overexpression promotes the migration and invasion of glioma cells

Migration and invasion of cancer cells is a key factor responsible for cancer metastasis (21). In order to investigate the influence of miR-21 on the migration and invasion of glioma cells a Transwell assay was performed using different glioma cell lines (U87, A172, T98 and U343). The cells were transfected with anti-miR-21 for 48 h, and the migration/invasion efficiency was detected. As presented Fig. 1A, the overexpression of miR-21 increased cell migration/invasion compared with the miR-NC group or untreated cells. Cell migration was increased significantly by 121±30% in U-343, 154±20% in T98, 91±21% in A172 and 110±15% in U87 cells, all compared with their respective untreated cells (n=6; P<0.01; Fig. 1B).

Sox2 is a crucial mediator in miR-21-induced migration and invasion in human glioma cells

A previous study identified that Sox2 was required for the proliferation and anchorage-independent growth of cancer cell lines in lung and esophageal squamous carcinoma, highlighting the importance of Sox2 as a lineage-survival oncogene (22). High Sox2 expression has been associated with several types of human solid tumors, including glioma (23) and silencing of Sox2 inhibited the proliferation and tumorigenesis abilities of glioblastoma cells (24). Additionally, a previous study revealed that Sox2 expression was increased in cells under hypoxic conditions (25). Notably, overexpression of Sox2 has been identified to promote migration and invasion ability of cancer cells (17,18). The present study evaluated the association between miR-21 and Sox2 in glioma cells. It was determined that miR-21 overexpression markedly promoted Sox2 expression in all four cell lines and the increased level of Sox2 was primarily concentrated in the nuclei of the glioma cells (Fig. 1C). Western blotting revealed higher levels of Sox2 in the nucleus of miR-21-transfected cells compared with the control cells transfected with miR-NC (Fig. 1D). These findings suggested that the miR-21 overexpression may increase Sox2 expression and its nuclear localization.

To investigate whether an increased level of Sox2 is required for the miR-21-induced migration/invasion of glioma cells miR-21 overexpression and Sox2 silencing with specific Sox2 siRNA were used. The migration/invasion ability of glioma cells was enhanced following miR-21 transfection, whereas Sox2 silencing significantly decreased the miR-21-induced migration/invasion with the inhibition rate of 45±9% in U-343, 54±11% in T98, 42±13% in A172 and 56±10% in U87 cells (n=6; P<0.01; Fig. 2A and B), respectively, compared with cells transfected with miR-21 only. These findings indicated that an increased expression level of Sox2 may be an important mediator of miR-21-induced glioma cell migration/invasion. In order to confirm this glioma cells were transfected with a Sox2-overexpressing plasmid and the migration/invasion potential of the cells was evaluated using a Transwell assay. It was revealed that Sox2 overexpression exhibited a similar effect as miR-21 in the enhancement of the migration/invasion ability of the glioma cells (increased by 110±20% in U-343, 100±20% in T98, 120±25% in A172 and 80±30% in U87 cells compared with controls; P<0.01; Fig. 2C). In addition, Sox2 knockdown significantly reduced the cell migration/invasion compared with control cells with an inhibition rate of 65±10% in U-343, 51±12% in T98, 62±15% in A172 and 72±10% in U87 cells (n=6; P<0.01; Fig. 2C). These findings indicated that Sox2 may act as an essential mediator in the miR-21-enhanced migration/invasion ability of glioma cells.

miR-21/Sox2-induced migration/invasion of glioma cells is involved in the activation of the Wnt/β-catenin signaling

Previous studies have reported that β-catenin is a vital downstream molecule of Sox2 (19,26). In order to determine whether Wnt/β-catenin signaling may be involved in miR-21-induced cell migration/invasion, the present study examined the effect of miR-21 on Wnt/β-catenin signaling using flow cytometry 48 h after the cells were transfected with miR-21 or anti-miR-21. It was determined that the β-catenin level in the miR-21 transfected cells was significantly higher compared with the control groups (increased by 123±28% in U-343, 132±21% in T98, 152±19% in A172 and 124±27% in U87 cells), whereas in the cells treated with anti-miR-21 a >50% reduction in β-catenin expression was observed (Fig. 3A and B), which indicated that β-catenin was associated with miR-21 levels in human glioma cells. In order to investigate whether β-catenin signaling was involved in glioma cell migration/invasion, a Transwell assay was performed and glioma cells were treated with BIO, a specific β-catenin signaling agonist, or XAV-939, a β-catenin signaling inhibitor. As presented in Fig. 3C, BIO treatment significantly promoted the migration/invasion of the four glioma cell lines compared with the control groups (P<0.01). By contrast, XAV-939 treatment significantly inhibited the migration/invasion compared with the control groups (P<0.01; Fig. 3C). Furthermore, β-catenin knockdown also significantly reduced the migration/invasion ability of the glioma cell lines compared with the control groups (P<0.01; Fig. 3D). Notably, β-catenin siRNA treatment significantly inhibited the miR-21-induced migration/invasion of human glioma cells compared with the (P<0.01; Fig. 3D). These findings suggested that β-catenin may act as a functional mediator in miR-21-enhanced glioma cell migration/invasion.

Sox2 mediates miR-21-induced β-catenin signaling

Previous studies have revealed the regulatory importance of Sox2 on Wnt/β-catenin signaling (19). The present study investigated the importance of Sox2 in miR-21-induced β-catenin signaling. As presented in Fig. 4A, the β-catenin expression level in Sox2-overexpressed glioma cells was significantly increased compared with the control group of untreated cells (P<0.01), whereas simultaneous Sox2 knockdown significantly inhibited the β-catenin expression (P<0.01; Fig. 4A). The β-catenin siRNA significantly reduced the Sox2-induced glioma cell migration/invasion compared with Sox2-overexpressed cells (P<0.01; Fig. 4B). Furthermore, BIO treatment significantly restored the Sox2-siRNA-induced migration/invasion inhibition across all cell lines (P<0.01; Fig. 4C). The present findings indicated that the upregulated expression of β-catenin promoted glioma cell migration/invasion and Sox2 may be a regulator of β-catenin. Treatment with BIO or Sox2 overexpression significantly increased anti-miR-21-induced migration/invasion inhibition in glioma cells (P<0.01; Fig. 4D). The current findings indicated that miR-21 overexpression may increase the migration/invasion ability of glioma cells via activation of Wnt/β-catenin signaling and Sox2 may act as a functional mediator of miR-21 and β-catenin signaling (Fig. 5).

Discussion

Hypoxia is a characteristic feature of locally advanced solid tumors and has been considered to be an adverse prognostic factor (5,2729). Tumor cells have adapted to this hypoxic environment by changing the expression patterns of some associated genes and many of these hypoxia-induced genes are mediated by the HIF-1 complex. Among these hypoxia-induced genes, several are closely involved with the migration and invasion functions of tumor cells. miRNAs are a class of evolutionarily conserved small, non-coding RNAs, which interact with the 3′-untranslated region of coding genes to regulate their expression (30). A previous study reported that a specific spectrum of miRNAs was upregulated under hypoxic conditions, and was partially responsible for the migration and invasion of cancer cells (31). miR-21 expression was upregulated in the majority of the human malignant gliomas specimens previously analyzed and it has been identified to promote cell invasion (16). However, the mechanism involved in miR-21 regulation of glioma cell migration and invasion remains to be elucidated (16,32).

The present study demonstrated that miR-21 overexpression may significantly increase the migration and invasion abilities of glioma cells. These findings are consistent with the observation in glioma specimens as miR-21 expression level has been correlated with the glioma grade and malignant gliomas have a higher invasion potential (22). Furthermore, overexpression of miR-21 was accompanied with upregulation of Sox2, and knockdown of Sox2 significantly inhibited miR-21-enhanced glioma cell migration and invasion. Similar to miR-21 overexpression, Sox2 overexpression also promoted migration and invasion of glioma cells. Therefore, the present study revealed that Sox2 may have a functional role in mediating miRNA-21-induced migration and invasion of glioma cells. Sox2 siRNA significantly inhibited the miR-21-induced migration/invasion of human glioma cells, suggesting that Sox2 may be a key mediator of miR-21-associated migration/invasion signaling. Sox2 is a key transcriptional factor and is upregulated in numerous tumors, including glioblastoma (18,33,34). A previous study demonstrated that Sox-2 induced epithelial-mesenchymal transition via activation of the Wnt/β-catenin signaling pathway in laryngeal cancer cells (35). The present study also revealed that Sox2 expression may induce β-catenin expression and in turn, β-catenin activation promoted migration and invasion of glioma cells. β-catenin siRNA or XAV-939, a β-catenin signaling inhibitor, significantly inhibited the miR-21 or Sox2-induced migration/invasion potential of human glioma cells. Conversely, BIO, a specific β-catenin signaling agonist, significantly increased the miR-21 or Sox2-siRNA-induced migration/invasion inhibition. These findings suggested β-catenin may be a major downstream target of miR-21 and Sox2 in the regulation of migration and invasion properties of glioma cells. Therefore, the present study identified a novel miR-21/Sox2/β-catenin signaling pathway that may regulate the migration and invasion of human glioma cells.

In conclusion, miR-21 may be upregulated by HIF-1 under hypoxic conditions, and miR-21 subsequently activate Sox2 and the β-catenin signaling pathway, which may result in the high migration and invasion potential of glioma cells (Fig. 5).

Acknowledgements

The present study was supported by the Guangdong Natural Science Foundation (grant no. 2014A030313758), Doctoral Fund of Ministry of Education of China (grant no. 20120002120020) and Science, Technology & Innovation Commission of Shenzhen Municipality (grant nos. JCYJ20120616213411826 and JCYJ20140417115840285). And Medical Scientific Research Foundation of Guangdong Province (grant no. A2014156).

References

1 

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

2 

Zagzag D, Lukyanov Y, Lan L, Ali MA, Esencay M, Mendez O, Yee H, Voura EB and Newcomb EW: Hypoxia-inducible factor 1 and VEGF upregulate CXCR4 in glioblastoma: Implications for angiogenesis and glioma cell invasion. Lab Invest. 86:1221–1232. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Brat DJ, Castellano-Sanchez AA, Hunter SB, Pecot M, Cohen C, Hammond EH, Devi SN, Kaur B and Van Meir EG: Pseudopalisades in glioblastoma are hypoxic, express extracellular matrix proteases and are formed by an actively migrating cell population. Cancer Res. 64:920–927. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Brat DJ and Van Meir EG: Vaso-occlusive and prothrombotic mechanisms associated with tumor hypoxia, necrosis, and accelerated growth in glioblastoma. Lab Invest. 84:397–405. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Harris AL: Hypoxia-a key regulatory factor in tumour growth. Nat Rev Cancer. 2:38–47. 2002. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Calin GA, Ferracin M, Cimmino A, Di Leva G, Shimizu M, Wojcik SE, Iorio MV, Visone R, Sever NI, Fabbri M, et al: A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N Engl J Med. 353:1793–1801. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Bentwich I, Avniel A, Karov Y, Aharonov R, Gilad S, Barad O, Barzilai A, Einat P, Einav U, Meiri E, et al: Identification of hundreds of conserved and nonconserved human microRNAs. Nature Genet. 37:766–770. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M and Rajewsky N: Combinatorial microRNA target predictions. Nat Genet. 37:495–500. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP and Burge CB: Prediction of mammalian microRNA targets. Cell. 115:787–798. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Kulshreshtha R, Ferracin M, Wojcik SE, Garzon R, Alder H, Agosto-Perez FJ, Davuluri R, Liu CG, Croce CM, Negrini M, et al: A microRNA signature of hypoxia. Mol Cell Biol. 27:1859–1867. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Ali N, Mah N, McLoughlin P and Costello CM: Identification of a hypoxia-responsive microRNA signature in lung endothelial cells. Irish J Med Sci. 181:S4142012.

13 

Qu K, Lin T, Pang Q, Liu T, Wang Z, Tai M, Meng F, Zhang J, Wan Y, Mao P, et al: Extracellular miRNA-21 as a novel biomarker in glioma: Evidence from meta-analysis, clinical validation and experimental investigations. Oncotarget. 7:33994–34010. 2016.PubMed/NCBI

14 

Shi R, Wang PY, Li XY, Chen JX, Li Y, Zhang XZ, Zhang CG, Jiang T, Li WB, Ding W and Cheng SJ: Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget. 6:26971–26981. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Põlajeva J, Swartling FJ, Jiang Y, Singh U, Pietras K, Uhrbom L, Westermark B and Roswall P: miRNA-21 is developmentally regulated in mouse brain and is co-expressed with SOX2 in glioma. BMC Cancer. 12:3782012. View Article : Google Scholar : PubMed/NCBI

16 

Gabriely G, Wurdinger T, Kesari S, Esau CC, Burchard J, Linsley PS and Krichevsky AM: MicroRNA 21 promotes glioma invasion by targeting matrix metalloproteinase regulators. Mol Cell Biol. 28:5369–5380. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Yang N, Hui L, Wang Y, Yang H and Jiang X: SOX2 promotes the migration and invasion of laryngeal cancer cells by induction of MMP-2 via the PI3K/Akt/mTOR pathway. Oncol Rep. 31:2651–2659. 2014.PubMed/NCBI

18 

Yang N, Hui L, Wang Y, Yang H and Jiang X: Overexpression of SOX2 promotes migration, invasion, and epithelial-mesenchymal transition through the Wnt/β-catenin pathway in laryngeal cancer Hep-2 cells. Tumour Biol. 35:7965–7973. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Chen S, Xu Y, Chen Y, Li X, Mou W, Wang L, Liu Y, Reisfeld RA, Xiang R, Lv D and Li N: SOX2 gene regulates the transcriptional network of oncogenes and affects tumorigenesis of human lung cancer cells. PloS One. 7:e363262012. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

Steeg PS: Tumor metastasis: Mechanistic insights and clinical challenges. Nat Med. 12:895–904. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Bass AJ, Watanabe H, Mermel CH, Yu S, Perner S, Verhaak RG, Kim SY, Wardwell L, Tamayo P, Gat-Viks I, et al: SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat Genet. 41:1238–1242. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Annovazzi L, Mellai M, Caldera V, Valente G and Schiffer D: SOX2 expression and amplification in gliomas and glioma cell lines. Cancer Genomics Proteomics. 8:139–147. 2011.PubMed/NCBI

24 

Gangemi RM, Griffero F, Marubbi D, Perera M, Capra MC, Malatesta P, Ravetti GL, Zona GL and Daga A: SOX2 silencing in glioblastoma tumor-initiating cells causes stop of proliferation and loss of tumorigenicity. Stem cells. 27:40–48. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Heddleston JM, Li Z, Lathia JD, Bao S, Hjelmeland AB and Rich JN: Hypoxia inducible factors in cancer stem cells. Br J Cancer. 102:789–795. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Hall CL, Bafico A, Dai J, Aaronson SA and Keller ET: Prostate cancer cells promote osteoblastic bone metastases through Wnts. Cancer Res. 65:7554–7560. 2005.PubMed/NCBI

27 

Osawa T and Shibuya M: Targeting cancer cells resistant to hypoxia and nutrient starvation to improve anti-angiogeneic therapy. Cell Cycle. 12:2519–2520. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Wilson WR and Hay MP: Targeting hypoxia in cancer therapy. Nat Rev Cancer. 11:393–410. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Melillo G: Targeting hypoxia cell signaling for cancer therapy. Cancer Metastasis Rev. 26:341–352. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Ambros V: microRNAs: Tiny regulators with great potential. Cell. 107:823–826. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Wang D, Qiu C, Zhang H, Wang J, Cui Q and Yin Y: Human microRNA oncogenes and tumor suppressors show significantly different biological patterns: From functions to targets. PloS One. 5(pii): e130672010.PubMed/NCBI

32 

Si ML, Zhu S, Wu H, Lu Z, Wu F and Mo YY: miR-21-mediated tumor growth. Oncogene. 26:2799–2803. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A and Weinberg RA: An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 40:499–507. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Berezovsky AD, Poisson LM, Cherba D, Webb CP, Transou AD, Lemke NW, Hong X, Hasselbach LA, Irtenkauf SM, Mikkelsen T and deCarvalho AC: Sox2 promotes malignancy in glioblastoma by regulating plasticity and astrocytic differentiation. Neoplasia. 16:193–206. 206.e119-e125, 2014.

35 

Yang N, Hui L, Wang Y, Yang H and Jiang X: Overexpression of SOX2 promotes migration, invasion, and epithelial-mesenchymal transition through the Wnt/β-catenin pathway in laryngeal cancer Hep-2 cells. Tumour Biol. 35:7965–7673. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2017
Volume 15 Issue 1

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
Luo G, Luo W, Sun X, Lin J, Wang M, Zhang Y, Luo W and Zhang Y: MicroRNA‑21 promotes migration and invasion of glioma cells via activation of Sox2 and β‑catenin signaling Retraction in /10.3892/mmr.2023.12982. Mol Med Rep 15: 187-193, 2017
APA
Luo, G., Luo, W., Sun, X., Lin, J., Wang, M., Zhang, Y. ... Zhang, Y. (2017). MicroRNA‑21 promotes migration and invasion of glioma cells via activation of Sox2 and β‑catenin signaling Retraction in /10.3892/mmr.2023.12982. Molecular Medicine Reports, 15, 187-193. https://doi.org/10.3892/mmr.2016.5971
MLA
Luo, G., Luo, W., Sun, X., Lin, J., Wang, M., Zhang, Y., Luo, W., Zhang, Y."MicroRNA‑21 promotes migration and invasion of glioma cells via activation of Sox2 and β‑catenin signaling Retraction in /10.3892/mmr.2023.12982". Molecular Medicine Reports 15.1 (2017): 187-193.
Chicago
Luo, G., Luo, W., Sun, X., Lin, J., Wang, M., Zhang, Y., Luo, W., Zhang, Y."MicroRNA‑21 promotes migration and invasion of glioma cells via activation of Sox2 and β‑catenin signaling Retraction in /10.3892/mmr.2023.12982". Molecular Medicine Reports 15, no. 1 (2017): 187-193. https://doi.org/10.3892/mmr.2016.5971