Knockdown of PREX2a inhibits the malignant phenotype of glioma cells

Retraction in: /10.3892/mmr.2021.12184

  • Authors:
    • Liang Liu
    • Zhixiong Liu
    • Hao Wang
    • Long Chen
    • Fuqiang Ruan
    • Jihui Zhang
    • Yi Hu
    • Hengshan Luo
    • Shuai Wen
  • View Affiliations

  • Published online on: January 19, 2016     https://doi.org/10.3892/mmr.2016.4799
  • Pages: 2301-2307
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioma is the most common type of malignant brain tumor. Phosphatidylinositol‑3,4,5‑trisphosphate‑dependent Rac exchange factor 2a (PREX2a), which is a regulator of the small guanosine triphosphatase Rac, has previously been identified as an oncoprotein that inhibits phosphatase and tensin homolog deleted on chromosome 10 (PTEN) activity. However, the role of PREX2a in the regulation of the malignant phenotype of glioma has yet to be reported. The present study demonstrated that the mRNA and protein expression levels of PREX2a were significantly increased in glioma tissue, as compared with in normal brain tissue. Furthermore, the expression levels of PREX2a were positively correlated with tumor grade. PREX2a‑specific small interfering RNA‑mediated knockdown significantly inhibited proliferation and induced apoptosis of SWO‑38 glioma cells. Furthermore, inhibition of PREX2a expression significantly suppressed cell cycle progression in glioma cells, as detected by cell cycle arrest at G1 phase. In addition, knockdown of PREX2a inhibited the invasion of SWO‑38 glioma cells. The present study also investigated the molecular mechanisms underlying the effects of PREX2a knockdown, and demonstrated that phosphoinositide 3‑kinase signaling was significantly downregulated, which may be due to the upregulation of PTEN activity. In conclusion, the present study is the first, to the best of our knowledge, to suggest that knockdown of PREX2a may effectively inhibit the malignant phenotype of glioma in vitro; therefore, PREX2a may be considered a potential molecular target for the treatment of glioma.

Introduction

Glioma, which is the most common type of brain tumor, accounts for ~30% of central nervous system tumors and 80% of all malignant brain tumors (1). Despite the notable development of therapies for the treatment of various types of cancer, the median survival rate of glioma has not markedly improved over the past few decades, which is predominantly due to its resistance to radiotherapy, chemotherapy and adjuvant therapies (25). Since aberrant expression of oncogenes and tumor suppressors has been reported to be involved in the development of glioma, identification of novel oncogenes may aid the development of therapeutic strategies for the treatment of glioma.

The phosphoinositide 3-kinase (PI3K) signaling pathway has previously been reported to have a crucial role in the development and progression of human cancer (6,7). As one of the most frequently mutated tumor suppressors in human cancer, phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is able to antagonize PI3K signaling, and the regulation of PTEN/PI3K signaling is of potential clinical importance (8). Phosphatidylinositol 3,4,5-trisphosphate-dependent Rac exchange factor 2a(PREX2a), which is a regulator of the small guanosine triphosphatase Rac, contains an N-terminal Dbl homology and pleckstrin homology (DHPH) domain, which confers guanine nuclear exchange factor (GEF) activity; pairs of PDZ and Dishevelled, Egl-10 and Pleckstrin domains; and a C-terminus, which exhibits weak similarity to inositol 4-polyphosphate phosphatase (9,10). PREX2a has been shown to bind directly to PTEN via its GEF activity (DHPH domain), inhibit PTEN activity, and activate downstream PI3K-dependent signaling (11,12). Therefore, as a direct inhibitor of PTEN activity, PREX2a is considered to have an oncogenic role. Guo et al (13) reported that knockdown of PREX2a was able to suppress gastric cancer cell proliferation and clonogenicity, and induce cell apoptosis and cell cycle arrest at G1-S phase. In addition, an investigation into the underlying molecular mechanism demonstrated that silencing PREX2a expression led to activation of PTEN and a decline in Akt phosphorylation in gastric cancer cells (13). However, the exact role of PREX2a in the regulation of glioma cells, as well as the underlying molecular mechanism, has yet to be elucidated.

The present study aimed to explore the role of PREX2a in the regulation of glioma cell proliferation, apoptosis, cell cycle progression and invasion. In addition, the underlying molecular mechanisms of PREX2a were investigated.

Materials and methods

Reagents

Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), Lipofectamine® 2000 and TRIzol® reagent were purchased from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). RevertAid First Strand cDNA Synthesis kit was purchased from Fermentas (Thermo Fisher Scientific, Inc., Pittsburgh, PA, USA). SYBR Green quantitative polymerase chain reaction (qPCR) Assay kit was purchased from TOYOBO (Shanghai) Co., Ltd. (Shanghai, China). 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was purchased from Biosharp (Hefei, China). Annexin V-Fluorescein Isothiocyanate (FITC) Apoptosis Detection kit was purchased from BD Pharmingen (San Diego, CA, USA). Transwell chamber was obtained from Corning Inc. (Corning, NY, USA). Rabbit anti-PREX2a (1:200; ab121462), phosphorylated (p)-PTEN (1:100; ab76431), PTEN (1:100; ab79156), p-Akt (1:50; ab81283), Akt (1:100; ab32505), p-mammalian target of rapamycin (mTOR; 1:100; ab109268), mTOR (1:100; ab2732) and glyceraldehyde 3-phosphate dehydrogenase (GAPDH; 1:50; ab181602) monoclonal antibodies, and goat anti-rabbit horseradish peroxidase-conjugated secondary antibody (1:5,000; ab6721) were purchased from Abcam (Cambridge, MA, USA). Enhanced chemiluminescence (ECL) kit was purchased from Pierce Biotechnology, Inc. (Rockford, IL, USA).

Tissue specimen collection

The present study was approved by the Ethics Committee of Central South University (Changsha, China). Written informed consent was obtained from all of the patients. A total of 24 primary glioma tissues and six normal brain specimens were collected from the Department of Neurosurgery, Xiangya Hospital of Central South University. The glioma patients included 10 females and 14 males who ranged in age from 28 to 71 years, with a mean age of 49.5 years. None of the patients had received radiation therapy or chemotherapy prior to surgical resection. The 24 glioma samples were classified according to the World Health Organization (WHO) grading system (14), and included five pilocytic astrocytomas (WHO I), six diffuse astrocytomas (WHO II), seven anaplasia astrocytomas (WHO III), and six primary glioblastomas (WHO IV). The tissues were collected under surgical resection and the histomorphology of all of the samples was confirmed by the Department of Pathology, Xiangya Hospital of Central South University. Tissues were immediately snap-frozen in liquid nitrogen following surgical removal.

Cells culture

The SWO-38 human glioma cell line was purchased from the Cell Bank of Central South University. The cells were cultured in DMEM supplemented with 10% FBS at 37°C in a humidified incubator containing 5% CO2.

Transfection

Lipofectamine® 2000 was used to perform transfection, according to the manufacturer's protocol. Briefly, the cells were cultured to 70% confluence and resuspended in serum-free medium. Non-specific (negative control) and PREX2a-specific small interfering (si)RNA (GeneChem Co., Ltd., Shanghai, China) and Lipofectamine® 2000 were diluted with serum-free medium separately. The diluted Lipofectamine® 2000 was added to the diluted siRNA, and incubated for 20 min at room temperature, prior to being added to the cell suspension. Following a 6 h incubation at 37°C in an atmosphere containing 5% CO2, the medium was replaced with normal serum-containing medium. The cells were subsequently cultured for 24 h prior to further experimentation.

Reverse transcription-qPCR (RT-qPCR) analysis

The tissue samples were homogenized in liquid nitrogen by grinding with a grinding rod. Subsequently, total RNA was extracted from the tissues or cells using TRIzol® reagent, according to the manufacturer's protocol. Total RNA was reverse transcribed into cDNA using the RevertAid First Strand cDNA Synthesis kit, according to the manufacturer's protocol. Briefly, 1 µl total RNA was mixed with 1 µl of 100 mm dNTP, 1 µl reverse transcriptase, 10 µl of 10X reverse transcription buffer, 1 µl RNase inhibitor and 1 µl primer. Nuclease-free H2O was added to obtain a final volume of 20 µl. Reverse transcription was performed at 16°C for 30 min, followed by an incubation step at 42°C for 30 min and enzyme inactivation at 85°C for 5 min. PCR was performed on 10 ng cDNA, using the SYBR Green qPCR Assay kit and the Applied Biosystems 7500 Real-Time PCR System (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocols. The specific primer pairs used were as follows: PREX2a, sense 5′-TGG GAG GGG TCC AAC ATCA-3′, anti-sense 5′-TCT TCA ACC GTC TGT GTT TTCTT-3′; GAPDH, sense 5′-CTC CTC CTG TTC GAC AGT CAGC-3′, and anti-sense 5′-CCC AAT ACG ACC AAA TCC GTT-3′ (Sangon Biotech Co., Ltd., Shanghai, China). GAPDH was used as an internal control. The PCR cycling conditions were as follows: 95°C for 10 min, followed by 40 cycles of denaturation at 95°C for 15 sec and annealing/elongation at 60°C for 1 min. Independent experiments were repeated three times. The relative mRNA expression levels were analyzed using the 2−∆∆Cq method (15).

Western blotting

Total protein was extracted from the tissues or cells using cold radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Shanghai, China). The protein concentrations were quantified using the Bicinchoninic Acid Protein Assay kit (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Proteins (50 µg) were then separated by 12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred onto a polyvinylidene difluoride (PVDF) membrane. The PVDF membrane was incubated with 5% milk in Tris-buffered saline containing 0.1% Tween at room temperature for 3 h, and then incubated with rabbit anti-PREX2a, p-Akt, Akt, p-mTOR, mTOR and GAPDH monoclonal antibodies at room temperature for 3 h. Subsequently, the membrane was incubated with goat anti-rabbit secondary antibody at room temperature for 40 min. Chemiluminescent detection was performed using an ECL kit. The blots were analyzed using Image-Pro Plus software 6.0 (Media Cybernetics, Inc., Rockville, MD, USA), and the relative protein expression levels were represented as the density ratio vs. GAPDH.

Cell proliferation assay

An MTT assay was used to measure cell proliferation. Cells in each group were cultured in 100 µl fresh serum-free medium/well in a 96-well plate. MTT (0.5 g/l) was added to the cells and incubated at 37°C for 0, 24, 48 and 72 h. Subsequently, the medium was removed by aspiration and 50 µl dimethyl sulfoxide was added to each well and incubated at 37°C for a further 10 min. The absorbance of each sample was measured at 492 nm using a plate reader (AF2200; Eppendorf, Hamburg, Germany).

Apoptosis analysis

Flow cytometry (FACSCalibur; BD Biosciences, Franklin Lakes, NJ, USA) was used to determine the rate of cell apoptosis using the Annexin V-FITC Apoptosis Detection kit. At 24 h post-transfection, the cells were harvested and washed twice with cold phosphate-buffered saline (PBS). Subsequently, 1×106 cells were resuspended in 200 µl binding buffer with 10 µl Annexin V-FITC and 5 µl propidium iodide (PI), and incubated in the dark for 30 min. Finally, 300 µl binding buffer was added to the cells, which where analyzed by flow cytometry. The flow cytometry data was analyzed using the BD Accuri C6 software (BD Biosciences).

Analysis of cell cycle distribution

A total of 1×106 cells from each group were collected in 1X PBS and fixed with 70% ethanol overnight at −20°C. The cells were then centrifuged at 1,000 × g for 5 min, washed in 1X PBS, and centrifuged for a further 5 min at 300 × g. The cells were resuspended in 300 µl PI staining buffer and incubated for 30 min at room temperature. DNA content analyses were performed using the BD Accuri C6 Flow Cytometer (BD Biosciences).

Cell invasion assay

The invasive ability of glioma cells was determined in 24-well Transwell chambers, which were coated with a layer of Matrigel (BD Biosciences). Glioma cells (1.0×105 cells/ml) suspended in serum-free DMEM were seeded in the upper chamber, and DMEM supplemented with 10% FBS was added to the lower chamber. Following a 24 h incubation at 37°C, the non-invading cells and the Matrigel on the interior of the inserts were removed using a cotton-tipped swab. Invasive cells on the lower surface of the membrane were stained with gentian violet, rinsed with water and air-dried. Five fields were randomly selected, and the number of cells was counted under a microscope (CX31; Olympus, Tokyo, Japan).

Statistical analysis

All data are presented as the mean ± standard deviation. One-way analysis of variance was used to statistically analyze the data using SPSS 17 software (SPSS Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

PREX2a is upregulated in glioma tissue

To explore the role of PREX2a in glioma, RT-qPCR and western blotting were performed, in order to determine the mRNA and protein expression levels of PREX2a in glioma tissue. As shown in Fig. 1A and B, the mRNA and protein expression levels of PREX2a were significantly increased in glioma tissues, as compared with in normal brain tissue. In addition, PREX2a expression was positively correlated with the WHO grade of glioma. No significant difference was detected in PREX2a mRNA expression between normal brain tissue and WHO I glioma, or between WHO III and WHO IV glioma (P>0.05). However, differences between any other two groups were statistically significant (P<0.05).

siRNA-induced knockdown of PREX2a inhibits proliferation and induces apoptosis of glioma cells

To investigate the role of PREX2a in the regulation of glioma in vitro, SWO-38 glioma cells were transfected with PREX2a-specific siRNA or non-specific siRNA, which was used as a negative control (NC). Post-transfection of the SWO-38 glioma cells with PREX2a siRNA, the mRNA expression levels of PREX2a were decreased by 78%, and the protein expression levels of PREX2a were decreased by 72%, as compared with the control group (P<0.01; Fig. 2A and B). However, transfection with NC siRNA did not affect the expression levels of PREX2a, as compared with the control group (P>0.05; Fig. 2A and B).

An MTT assay was conducted to determine cell proliferation in each group. Following knockdown of PREX2a expression, the proliferation rate of SWO-38 glioma cells was significantly reduced by 32%, as compared with the control group (P<0.01; Fig. 2C). In addition, the effects of siRNA-mediated knockdown of PREX2a on apoptosis of glioma SWO-38 cells were detected. As shown in Fig. 2D, the rate of cell apoptosis was markedly increased following knockdown of PREX2a expression, as compared with in the control group (P<0.01). The percentage of apoptotic cells in the control, NC and PREX2a siRNA groups was 2.91±0.12, 3.04±0.14 and 11.87±0.46%, respectively. These results suggest that siRNA-mediated knockdown of PREX2a may inhibit proliferation and induce apoptosis of glioma cells.

siRNA-mediated knockdown of PREX2a induces cell cycle arrest in glioma cells

Suppression of cell cycle progression was detected in SWO-38 glioma cells following knockdown of PREX2a expression. As shown in Fig. 3, post-transfection with PREX2a siRNA, the SWO-38 cells exhibited a significant increase in the percentage of cells in G1 phase (control, 36.87±2.31%; NC, 37.65±2.57%; PREX2a siRNA, 67.03±2.92%; P<0.01) and a corresponding reduction in the percentage of cells in S phase (control, 39.36±2.54%; NC, 38.71±2.36%; PREX2a siRNA, 24.63±2.7%; P<0.01) and G2/M phase (control, 23.77±3.86%; NC, 23.64±3.14%; PREX2a siRNA, 8.34±3.83%; P<0.01). These results suggest that siRNA-mediated knockdown of PREX2a may induce cell cycle arrest at G1 phase.

siRNA-mediated knockdown of PREX2a suppresses invasion of glioma cells

The invasive capacity of glioma SWO-38 cells was significantly reduced following knockdown of PREX2a expression (P<0.01; Fig. 4A and B). The percentage of invasive cells in the control, NC and PREX2a siRNA groups was 100±5.27, 99.9±1.83 and 41.9±5.29%, respectively (Fig. 4B). These results suggest that PREX2a has a promoting role in the regulation of glioma cell invasion.

siRNA-mediated knockdown of PREX2a inhibits PI3K signaling activity in glioma cells

The present study further investigated the activity of the PI3K/Akt/mTOR signaling pathway in SWO-38 cells with or without transfection with PREX2a siRNA. Following knockdown of PREX2a expression, the phosphorylation levels of PTEN in the SWO-38 glioma cells were reduced, suggesting that the activity of PTEN was upregulated. Furthermore, the phosphorylation levels of Akt and mTOR were decreased, indicating that activity of the PI3K/Akt/mTOR signaling pathway was downregulated following knockdown of PREX2a expression (Fig. 5). These results indicate that knockdown of PREX2a may inhibit the activity of the PI3K/Akt/mTOR signaling pathway via activation of PTEN.

Discussion

Previous studies regarding the aberrant expression of oncogenes and tumor suppressors show potential for the development of effective therapeutic strategies for the treatment of glioma (1,16,17). The present study is the first, to the best of our knowledge, to demonstrate that PREX2a may have an oncogenic role in the regulation of glioma in vitro, and may be associated with cell proliferation, apoptosis, cell cycle progression and invasion of glioma cells. Furthermore, the molecular mechanism underlying the effects of PREX2a in glioma cells was investigated, and was revealed to involve inhibition of PTEN activity and the promotion of PI3K/Akt/mTOR signaling, which has a key role in the pathological process of malignant tumors (18).

PREX2 is a GEF for the Rac guanosine triphosphatase (GTPase), which exhibits sequence similarity to PREX1 (11). The expression levels of PREX1 are increased in metastatic prostate cancer, and it has been shown to promote metastasis and invasion of prostate cancer cells (19). The GEF activity of PREX1 is critical for Rac-mediated formation of reactive oxygen species in response to PI3K signaling (20). Similarly, PREX2, including PREX2a and PREX2b, is also a regulator of the small GTPase Rac, leading to increased levels of GTP-bound Rac that could be further stimulated by enhancing the activity of PI3K signaling (11,21). Furthermore, PREX2a has been reported to participate in numerous biological processes, including cell growth, apoptosis, cell cycle progression and migration (13,22).

Inactivation of PTEN leads to accumulation of phosphatidylinositol (3,4,5)-trisphosphate (PIP3), resulting in increased Akt activity, which promotes cellular survival, cell cycle progression and growth, thereby contributing to oncogenesis (23,24). PREX2a is able to stimulate cell proliferation via inhibition of PTEN activity and upregulation of the downstream PI3K signaling pathway, thus suggesting that aberrant regulation of PTEN by PREX2a may represent a key tumorigenic mechanism (11). Chen et al (22) demonstrated that knockdown of PREX2a was able to inhibit cell proliferation, migration and invasion of neuroblastoma cells via mediation of the PTEN/PI3K/Akt/mTOR pathway. Similar findings have also been reported in gastric cancer cells (13). Furthermore, overexpression of PREX has been shown to be associated with poor patient outcome in breast cancer (25). A previous whole-genome sequencing study identified PREX2 as a significantly mutated gene in melanoma (12). However, the detailed role of PREX2a in glioma remains unclear. In the present study, the expression levels of PREX2a were significantly increased in glioma tissue, as compared with in normal adjacent tissue. To further determine whether PREX2a participated in the development and progression of glioma, glioma cells were transfected with PREX2a-specific siRNA, in order to suppress its expression. Knockdown of PREX2a significantly suppressed proliferation and promoted apoptosis of glioma cells. Furthermore, silencing PREX2a induced a cell cycle arrest at G1 phase. Accordingly, these results suggested that inhibition of cell proliferation induced by PREX2a knockdown may be due to cell cycle arrest at G1 phase. In addition, it may be hypothesized that PREX2a promotes the regulation of glioma cell invasion.

In some settings, partial loss of PTEN function is sufficient to drive tumor development (2629); therefore, the binding partners of PTEN may act as potential oncogenes or tumor suppressors via mediation of the activity of PTEN. As a binding partner of PTEN, PREX2a is able to inhibit its lipid phosphatase activity, which leads to accumulation of PIP3 and phosphorylation of Akt/mTOR, as a consequence promoting cellular survival, proliferation and cell cycle progression (25). Accordingly, PREX2a is able to contribute to tumorigenesis. The present study demonstrated that knockdown of PREX2a inhibited the malignant phenotype of glioma cells, and further investigated whether PREX2a influenced the PI3K/Akt/mTOR pathway in glioma cells. Knockdown of PREX2a in glioma cells resulted in increased activity of PTEN and decreased expression of p-AKT and p-mTOR, thus indicating that the PI3K/Akt/mTOR signaling pathway was downregulated.

In conclusion, the present study revealed a crucial role for PREX2a in the regulation of proliferation, apoptosis, cell cycle progression and invasion of glioma cells via mediation of PI3K/Akt/mTOR signaling. Based on these findings, PREX2a may be considered a potential target for the treatment of glioma.

References

1 

Goodenberger ML and Jenkins RB: Genetics of adult glioma. Cancer Genet. 205:613–621. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Stewart LA: Chemotherapy in adult high-grade glioma: A systematic review and meta-analysis of individual patient data from 12 randomised trials. Lancet. 359:1011–1018. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Zhu VF, Yang J, Lebrun DG and Li M: Understanding the role of cytokines in glioblastoma multiforme pathogenesis. Cancer Lett. 316:139–150. 2012. View Article : Google Scholar

4 

Sathornsumetee S, Reardon DA, Desjardins A, Quinn JA, Vredenburgh JJ and Rich JN: Molecularly targeted therapy for malignant glioma. Cancer. 110:13–24. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Pulkkanen KJ and Yla-Herttuala S: Gene therapy for malignant glioma: Current clinical status. Mol Ther. 12:585–598. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Danielsen SA, Eide PW, Nesbakken A, Guren T, Leithe E and Lothe RA: Portrait of the PI3K/AKT pathway in colorectal cancer. Biochim Biophys Acta. 1855:104–121. 2015.

7 

Follo MY, Manzoli L, Poli A, McCubrey JA and Cocco L: PLC and PI3K/Akt/mTOR signalling in disease and cancer. Adv Biol Regul. 57:10–16. 2015. View Article : Google Scholar

8 

Carnero A and Paramio JM: The PTEN/PI3K/AKT pathway in vivo, cancer mouse models. Front Oncol. 4:2522014. View Article : Google Scholar : PubMed/NCBI

9 

Fine B, Hodakoski C, Koujak S, Su T, Saal LH, Maurer M, Hopkins B, Keniry M, Sulis ML, Mense S, et al: Activation of the PI3K pathway in cancer through inhibition of PTEN by exchange factor P-REX2a. Science. 325:1261–1265. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Joseph RE and Norris FA: Substrate specificity and recognition is conferred by the pleckstrin homology domain of the Dbl family guanine nucleotide exchange factor P-Rex2. J Biol Chem. 280:27508–27512. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Rosenfeldt H, Vázquez-Prado J and Gutkind JS: P-REX2, a novel PI-3-kinase sensitive Rac exchange factor. FEBS Lett. 572:167–171. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Pandiella A and Montero JC: Molecular pathways: P-Rex in cancer. Clin Cancer Res. 19:4564–4569. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Guo B, Liu L, Yao J, Ma R, Chang D, Li Z, Song T and Huang C: miR-338–3p suppresses gastric cancer progression through a PTEN-AKT axis by targeting P-REX2a. Mol Cancer Res. 12:313–321. 2014. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

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

16 

Auffinger B, Thaci B, Ahmed A, Ulasov I and Lesniak MS: MicroRNA targeting as a therapeutic strategy against glioma. Curr Mol Med. 13:535–542. 2013. View Article : Google Scholar

17 

Marumoto T and Saya H: Molecular biology of glioma. Adv Exp Med Biol. 746:2–11. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Cantley LC: The phosphoinositide 3-kinase pathway. Science. 296:1655–1657. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Qin J, Xie Y, Wang B, Hoshino M, Wolff DW, Zhao J, Scofield MA, Dowd FJ, Lin MF and Tu Y: Upregulation of PIP3-dependent Rac exchanger 1 (P-Rex1) promotes prostate cancer metastasis. Oncogene. 28:1853–1863. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Welch HC, Coadwell WJ, Ellson CD, Ferguson GJ, Andrews SR, Erdjument-Bromage H, Tempst P, Hawkins PT and Stephens LR: P-Rex1, a PtdIns (3,4,5)P3- and Gbetagamma-regulated guanine-nucleotide exchange factor for Rac. Cell. 108:809–821. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Li Z, Paik JH, Wang Z, Hla T and Wu D: Role of guanine nucleotide exchange factor P-Rex-2b in sphingosine 1-phosphate-induced Rac1 activation and cell migration in endothelial cells. Prostaglandins Other Lipid Mediat. 76:95–104. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Chen X, Pan M, Han L, Lu H, Hao X and Dong Q: miR-338-3p suppresses neuroblastoma proliferation, invasion and migration through targeting PREX2a. FEBS Lett. 587:3729–3737. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Leslie NR and Downes CP: PTEN function: How normal cells control it and tumour cells lose it. Biochem J. 382(Pt 1): 1–11. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Yuan TL and Cantley LC: PI3K pathway alterations in cancer: Variations on a theme. Oncogene. 27:5497–5510. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Barrio-Real L and Kazanietz MG: Rho GEFs and cancer: Linking gene expression and metastatic dissemination. Sci Signal. 5:pe432012. View Article : Google Scholar : PubMed/NCBI

26 

Salmena L, Carracedo A and Pandolfi PP: Tenets of PTEN tumor suppression. Cell. 133:403–414. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Kwabi-Addo B, Giri D, Schmidt K, Podsypanina K, Parsons R, Greenberg N and Ittmann M: Haploinsufficiency of the Pten tumor suppressor gene promotes prostate cancer progression. Proc Natl Acad Sci USA. 98:11563–11568. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Kwon CH, Zhao D, Chen J, Alcantara S, Li Y, Burns DK, Mason RP, Lee EY, Wu H and Parada LF: Pten haploinsufficiency accelerates formation of high-grade astrocytomas. Cancer Res. 68:3286–3294. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Trotman LC, Niki M, Dotan ZA, Koutcher JA, Di Cristofano A, Xiao A, Khoo AS, Roy-Burman P, Greenberg NM, Van Dyke T, et al: Pten dose dictates cancer progression in the prostate. PLoS Biol. 1:E592003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 13 Issue 3

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
Liu L, Liu Z, Wang H, Chen L, Ruan F, Zhang J, Hu Y, Luo H and Wen S: Knockdown of PREX2a inhibits the malignant phenotype of glioma cells Retraction in /10.3892/mmr.2021.12184. Mol Med Rep 13: 2301-2307, 2016
APA
Liu, L., Liu, Z., Wang, H., Chen, L., Ruan, F., Zhang, J. ... Wen, S. (2016). Knockdown of PREX2a inhibits the malignant phenotype of glioma cells Retraction in /10.3892/mmr.2021.12184. Molecular Medicine Reports, 13, 2301-2307. https://doi.org/10.3892/mmr.2016.4799
MLA
Liu, L., Liu, Z., Wang, H., Chen, L., Ruan, F., Zhang, J., Hu, Y., Luo, H., Wen, S."Knockdown of PREX2a inhibits the malignant phenotype of glioma cells Retraction in /10.3892/mmr.2021.12184". Molecular Medicine Reports 13.3 (2016): 2301-2307.
Chicago
Liu, L., Liu, Z., Wang, H., Chen, L., Ruan, F., Zhang, J., Hu, Y., Luo, H., Wen, S."Knockdown of PREX2a inhibits the malignant phenotype of glioma cells Retraction in /10.3892/mmr.2021.12184". Molecular Medicine Reports 13, no. 3 (2016): 2301-2307. https://doi.org/10.3892/mmr.2016.4799