miR-142 inhibits the migration and invasion of glioma by targeting Rac1

  • Authors:
    • Wenyi Qin
    • Xiaofeng Rong
    • Jiangchuan Dong
    • Chao Yu
    • Juan Yang
  • View Affiliations

  • Published online on: July 13, 2017     https://doi.org/10.3892/or.2017.5816
  • Pages: 1543-1550
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Increasing evidence has shown that aberrant microRNAs (miRNAs) are implicated in tumorigenesis and tumor progression by regulating oncogenes or tumor suppressors. Dysregulation of miR-142 has been reported in multiple tumors. However, its clinical roles and underlying mechanism in glioma remain to be elucidated. In the present study, we found that the expression of miR-142 was significantly downregulated in both glioma tissues and cell lines by qRT-PCR. Clinical analysis revealed that decreased miR-142 was markedly associated with advanced World Health Organization (WHO) grade. Moreover, we disclosed that miR-142 was a novel independent prognostic marker in the prediction of the 5-year survival of glioma patients. The ectopic overexpression of miR-142 inhibited cell migration, invasion and invasion‑related gene expression. Notably, miR-142 modulated Rac1 by directly binding to its 3'-untranslated (3'-UTR) region, leading to the suppression of the expression of matrix metalloproteinases (MMPs). In glioma clinical samples, miR-142 was inversely correlated with Rac1 expression, and played positive roles in glioma migration and invasion. Alteration of Rac1 expression at least partially abolished the migration, invasion and MMP expression of miR-142 in glioma cells. In the present study, we identified Rac1 as a functional target of miR-142 in glioma. In conclusion, our data indicated that miR-142 inhibited the migration, invasion and MMP expression of glioma by targeting Rac1, and may represent a novel potential therapeutic target and prognostic marker for glioma.

Introduction

Glioma is the most common, aggressive and lethal type of malignant tumor of the central nervous system (1,2). Its relapse and mortality rate are constantly increasing due to the inefficiency of current treatment and this has led to its poor prognosis despite the combination of multidisciplinary therapies including surgical resection, chemotherapy and radiotherapy (3,4). Although diverse therapeutic efforts have been made, the median survival of glioma patients has not changed markedly in the last few years (5,6). The poor prognosis highlights the urgent need to elucidate the detailed molecular mechanism for the development of novel therapeutic tools against glioma and the identification of diagnostic and prognostic markers of glioma (7).

MicroRNAs (miRNAs) are an endogenous group of evolutionarily conserved, non-coding RNAs, which function as negative modulators of gene expression at the post-transcriptional level via binding to the 3′-untranslated (3′-UTR) regions of target mRNAs through complementary binding, causing translational suppression or mRNA degradation (810). Accumulating evidence has confirmed that miRNAs are involved in diverse fundamental biological processes including cell differentiation, apoptosis, proliferation and invasion (1113). An increasing number of studies have demonstrated that aberrant miRNAs play crucial roles in cancer pathogenesis and progression (14). Recently, miR-142 was reported to be dysregulated in multiple cancers (1517). For instance, it was significantly downregulated in cervical cancer (13), osteosarcoma (18), lung (19) and colon cancer (20,21). Previous studies revealed that miR-142 was identified as a regulator of cell proliferation, migration and invasion in different cancers. miR-142 inhibits cell proliferation and invasion of cervical cancer cells by targeting FZD7 (13). miR-142 functions as a potential tumor suppressor in human osteosarcoma by targeting HMGA1 (22). In addition, serum miR-142 is associated with early relapse in operable lung adenocarcinoma patients (23). However, the expression of miR-142 is overexpressed in nasopharyngeal carcinoma (24), testicular (25), and head and neck cancer (26), esophageal squamous cell (27) and renal cell carcinoma (28). miR-142 suppresses SOCS6 expression and promotes cell proliferation in nasopharyngeal carcinoma (24). Oncogenic miR-142 is associated with cellular migration, proliferation and apoptosis in renal cell carcinoma (28). Therefore, it is suggested that the functional significance of miR-142 in tumorigenesis and progression is cancer-type specific. However, the functional role and the underlying molecular mechanism by which miR-142 regulates the initiation and development of glioma have yet to be elucidated.

In the present study, we investigated the expression and biological function of miR-142 in glioma progression. We found that miR-142 was significantly decreased in both glioma tissues and cell lines. Its expression level was closely correlated with World Health Organization (WHO) grading and poor survival outcome in patients. miR-142 inhibited glioma cell migration and invasion in vitro as determined by gain- and loss-of-function experiments by directly targeting Rac1 thus leading to the suppression of matrix metalloproteinases (MMPs). Collectively, these data confirmed the underlying mechanism by which miR-142 inhibits migration and invasion of glioma and identified miR-142 as a novel prognostic biomarker for glioma patients.

Materials and methods

Clinical tissues and cell lines

Ninety-seven glioma and 25 human normal brain tissues were obtained from The First Affiliated Hospital of Chongqing Medical University from January 2007 to December 2010. None of the patients had received local or systemic treatment before surgery. Informed consent was obtained from all the patients and the present study was approved by the Ethics Committee of the First Affiliated Hospital of Chongqing Medical University according to the Declaration of Helsinki.

Human glioma cell lines (U87, U251, LN229 and U373) and normal human astrocytes (NHA) were purchased from the Institute of Biochemistry and Cell Biology (Chinese Academy of Sciences, Shanghai, China) and were cultured in Dulbecco's modified Eagle's medium/F12 (DMEM; HyClone, Logan, UT, USA) supplemented with 10% fetal bovine serum (FBS; Gibco, Grand Island, NY, USA) and 1% penicillin/streptomycin (Sigma-Aldrich, St. Louis, MO, USA) at 37°C with 5% CO2.

RNA extraction, quantitative real-time PCR (qRT-PCR)

Total RNA was extracted from tissues and cells by TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions. RNA was reverse-transcribed using a TaqMan Human MiRNA Assay kit (Applied Biosystems, Foster City, CA, USA) and cDNA was then amplified with a SYBR® Premix Ex Taq™ II (Perfect Real-Time) kit (Takara Bio Inc., Shiga, Japan) and analyzed using an Applied Biosystems 7900 Real-Time PCR System. Hsa-miR-142 primer (HmiRQP0185), snRNA U6 qPCR primer (HmiRQP9001) and GAPDH (HQP006940) were purchased from GeneCopoeia (Guangzhou, China). The primer for Rac1 was synthesized by GenePharma (Shanghai, China).

Western blotting

Proteins were isolated in RIPA buffer (Beyotime, Haimen, China) and the protein concentration was determined using a Bicinchoninic Acid (BCA) Protein Assay kit (Thermo Fischer Scientific, Rockford, IL, USA). Protein (40 µg) was separated on 10% SDS-PAGE and transferred to polyvinylidene difluoride (PVDF) membranes (Millipore, Billerica, MA, USA). The membranes were blocked with 5% non-fat milk in Tris-buffered saline with Tween-20 (TBST) and then incubated at 4°C overnight with primary antibodies: Rac1, MMP2, MMP9 and GAPDH (1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA). After being washed with TBST, the membranes were incubated with an appropriate peroxidase-conjugated secondary antibody for 2 h at room temperature (ZSGB-BIO, Beijing, China). Protein bands were visualized using an enhanced chemiluminescence kit (Amersham, Little Chalfont, UK).

Immunohistochemical staining analysis (IHC)

IHC was performed on formalin-fixed paraffin sections. MMP2 and MMP9 (Cell Signaling Technology, Inc.) (1:100) antibodies were used in immunohistochemical staining with the streptavidin peroxidase-conjugated (SP-IHC) method. The percentage of positive cells was expressed as the following grades: 0, <5%; 1, 6–25%; 2, 26–50%; 3, 51–75%; and 4, >75%.

Cell transfection

The cells were seeded into 6-well plates and transfected with vectors using Lipofectamine 2000 reagent (Invitrogen Life Technologies) in accordance with the manufacturer's protocol. miRNA expression vectors, including miR-142 expression vector (HmiR0282), the control vector (CmiR0001-MR04) were obtained from GeneCopoeia. The Rac1 expression plasmid was produced by GenePharma.

Wound-healing migration assay

To confirm the migratory ability of glioma cells, we performed a wound-healing assay in each group. Cells were seeded into 6-well plates and when confluence reached 80–90%, a 200 µl tip was used to create a wound across the cell plates. Subsequently the cells were washed twice with phosphate-buffered saline (PBS) to remove the detached cells. The cells were then cultured in DMEM in a common incubator for 48 h. Finally, the images were captured using phase-contrast microscope.

Transwell invasion assays

Cell invasion assays were determined by 8-µM pore-sized Transwell inserts (Nalge Nunc International Corp., Naperville, IL, USA). The upper chambers were pre-coated with Matrigel (BD Biosciences, Franklin Lakes, NJ, USA). The cells (2.5×104) in 150 µl of serum-free DMEM were loaded into the top chamber, and 750 µl of DMEM with 10% FBS were placed in the bottom chamber. After 24 h of incubation, the cells were fixed in 4% paraformaldehyde and stained with 0.3% crystal violet. The cells remaining in the top layer were swabbed carefully and the bottom cells were counted under microscope.

Luciferase reporter assay

Luciferase activity was determined using Dual Luciferase Assay (Promega, Madison, WI, USA) according to the manufacturer's protocols. The wild-type 3′-UTR of Rac1 containing the putative miR-142 target site was amplified and cloned into the pGL3-control luciferase reporter vector (Promega). The mutant constructs were generated using a QuickChange site-directed mutagenesis kit (Stratagene, La Jolla, CA, USA). The experiment was performed in duplicate in three independent experiments.

Statistical analysis

Data are presented as the mean ± SD from at least three independent replicates. SPSS software 16.0 (SPSS, Inc., Chicago, IL, USA) and GraphPad Prism 6.0 (GraphPad, San Diego, CA, USA CA, USA) were used for a two-tailed Students t-test, Pearson's correlation analysis, Kaplan-Meier method and the log-rank test to evaluate statistical significance. Differences were defined as P<0.05.

Results

Decreased expression of miR-142 in human glioma tissues and cells

We performed qRT-PCR to investigate the expression level of miR-142 in clinical tissues. As shown in Fig. 1A, the expression of miR-142 was significantly decreased in the glioma tissues compared with the normal brain tissues (Fig. 1A; P<0.05). According to the pathological diagnosis, the expression of miR-142 in the high grade glioma (III and IV) were lower than that in the low grade glioma tissues (I and II), which indicated that miR-142 expression was associated with glioma malignancy (Fig. 1B; P<0.05). Consistently, all glioma cell lines had significantly decreased expression of miR-142 than normal astrocytes (Fig. 1C; P<0.05). These data revealed that miR-142 may act as a tumor suppressor in glioma.

Decreased expression of miR-142 predicts a poor prognosis

To assess the clinical significance of miR-142 in glioma, we determined the median expression value as a cut-off to divide glioma patients into groups of high or low expression levels of miR-142. As shown in Table I, low expression of miR-142 was prominently associated with the histological grade of the glioma (P=0.001). Hence, these findings revealed that decreased expression of miR-142 was involved in the progression of glioma. Moreover, survival analysis revealed that the downregulation of miR-142 was significantly correlated with shorter overall survival (P=0.0002; Fig. 2) in glioma patients. Furthermore, miR-142 expression was an independent prognostic factor for predicting overall survival in glioma patients (P=0.001; 0.001, respectively; Table II). These results revealed that miR-142 may possibly serve as a potential prognostic biomarker in glioma patients.

Table I.

Association of miR-142 expression with clinicopathological characteristics of glioma patients.

Table I.

Association of miR-142 expression with clinicopathological characteristics of glioma patients.

miR-142 expression level

Clinical parametersCases (n)High (n=43)Low (n=54)P-value (aP<0.05)
Age (years) 0.936
  <60652936
  ≥60321418
Sex 0.629
  Male592534
  Female381820
WHO grade 0.001a
  I+II342410
  III+IV631944
Location 0.791
  Supratentorial693039
  Subtentorial281315

{ label (or @symbol) needed for fn[@id='tfn1-or-38-03-1543'] } miR-142, microRNA-142; WHO, World Health Organization

a Statistically significant.

Table II.

Univariate and multivariate analysis of prognostic factors in glioma patients.

Table II.

Univariate and multivariate analysis of prognostic factors in glioma patients.

Univariate analysisMultivariate analysis


VariablesHR95% CIP-valueHR95% CIP-value
Age (years)1.2050.563–2.2730.4581.1800.397–1.7090.436
Sex1.5380.558–2.7830.5231.0390.482–1.9270.329
WHO grade4.3271.872–5.6740.001a2.5121.287–3.1650.002a
miR-1423.9651.583–7.3820.001a3.2181.237–6.9620.001a

{ label (or @symbol) needed for fn[@id='tfn3-or-38-03-1543'] } HR, hazard ratio; CI, confidence interval; miR-142, microRNA-142; WHO, World Health Organization

a Statistically significant.

miR-142 inhibits cell migration and invasion in human glioma cells

To explore the biological function of miR-142 in glioma, we transfected miR-142 mimics to ectopically express this miRNA in U87 and U251 cells. The results revealed that the expression of miR-142 was significantly increased by appropriate vectors in both cell lines as determined by qRT-PCR (Fig. 3A; P<0.01). Wound-healing assay demonstrated that the overexpression of miR-142 significantly inhibited the migration ability of U87 and U251 cells (Fig. 3B; P<0.05). Consistently, Transwell assay revealed that the ectopic expression of miR-142 suppressed the invasive number of glioma cells (Fig. 3C; P<0.05). These results demonstrated that miR-142 regulates the migration and invasion of glioma cells.

Overexpression of miR-142 inhibits invasion-related genes

MMP2 and MMP9 are important members of the MMP family, which degrade all essential components of the extracellular matrix and play critical roles in cancer metastasis, including glioma. We disclosed whether miR-142 modulated these invasion-related genes. Our results revealed that miR-142 overexpression significantly decreased the expression of MMP2 and MMP9 in both U87 and U251 cells (P<0.05; Fig. 4A and B). Moreover, we investigated the correlation between miR-142 expression and MMPs in glioma tissues. We found that MMP2 and MMP9 expression in the high miR-142 expression group was significantly lower than that in the low expression miR-142 group (P<0.05; Fig. 4C). In conclusion, these results revealed that miR-142 is critical for glioma cell invasion by regulating the expression of MMPs.

Rac1 is the direct downstream target of miR-142

We searched for candidate target genes of miR-142 by exploring different algorithm programs (TargetScan and miRanda) and predicted that the 3′-UTR of Rac1 contains a putative binding site for miR-142 (Fig. 5A). A luciferase assay was performed to investigate whether miR-142 directly targets Rac1. Our data demonstrated that ectopic expression of miR-142 induced a clear decrease of the luciferase activity of the wild-type (wt) Rac1 3′-UTR, but did not inhibit the activity of Rac1 with a mutant (mt) 3′-UTR (Fig. 5B; P<0.05). In addition, miR-142 overexpression obviously suppressed Rac1 mRNA (Fig. 5C; P<0.05) and protein (Fig. 5D; P<0.05) expression in U87 and U251 cells. These results indicated that miR-142 directly targeted Rac1 in glioma cells through interaction with its 3′-UTR.

Rac1 levels are inversely correlated with miR-142 levels in glioma tissues

To further confirm the aforementioned findings, we detected the expression of Rac1 in human glioma tissues. Our data demonstrated that Rac1 mRNA and protein levels were significantly lower in the high miR-142 expression group than that in the low miR-142 expression group in glioma (P<0.05; Fig. 6A and B). In addition, the results revealed that the mRNA level of Rac1 in glioma tissues was inversely correlated with miR-142 expression (R2=0.7754; P<0.0001; Fig. 6C). In conclusion, these data revealed that Rac1 was a direct downstream target of miR-142 in glioma.

Overexpression of Rac1 expression counteracted the suppressive effects of miR-142 expression

To further confirm whether Rac1 was involved in miR-142-mediated inhibition of migration, invasion and invasion-related genes, we restored Rac1 expression in U87-miR-142 and U251-miR-142 cells by transfecting Rac1 expression plasmid (P<0.05; Fig. 7A). The results revealed that Rac1 overexpression promoted cell migration (P<0.05; Fig. 7B), invasion (P<0.05; Fig. 7C) and invasion-related MMP expression (P<0.05; Fig. 7D). These data demonstrated that Rac1 is a downstream mediator in the function of miR-142 in glioma.

Discussion

Recently, accumulating evidence has confirmed that aberrant miRNAs are involved in the initiation, development and progression of human malignancies via modulation of their targeting oncogenes or tumor-suppressor genes, which could potentially serve as biomarkers for the prediction and prognosis of diverse cancers, including glioma. Therefore, identification of cancer-specific miRNAs is critical for elucidating their molecular mechanism and vital for developing novel therapeutic targets. In previous studies, miRNA-142 negatively regulated canonical Wnt/β-catenin signaling, which plays pivotal roles in cancer progression (29). Schwickert et al demonstrated that miRNA-142 inhibits breast cancer cell invasiveness by synchronous targeting of WASL, integrin αV and additional cytoskeletal elements (30). miR-142 functions as a potential tumor suppressor directly targeting HMGB1 in non-small cell lung carcinoma (31). However, conversely, miR-142 could promote cell proliferation in nasopharyngeal carcinoma (24). Moreover, miR-142 plays an oncogenic role in cell migration, proliferation and apoptosis in renal cell carcinoma (28). These data indicated that the expression level and biological function of miR-142 was cancer type-dependent.

In the present study, we confirmed that the expression of miR-142 was significantly downregulated in clinical specimens and cell lines. Moreover, our data revealed that decreased expression of miR-142 was associated with advanced WHO grade in glioma. In addition, miR-142 expression was an independent prognostic indicator in the prediction of the 5-year overall survival of glioma patients. Collectively, these data revealed that miR-142 may play a critical role in the development of glioma and the decreased expression of miR-142 confers a worse prognosis and could be identified as a prognostic indicator. Mechanically, overexpression of miR-142 suppressed the cell migration and invasion capacity of glioma U87 and U251 cells. Moreover, miR-142 inhibited the invasion-related MMP gene expression. Furthermore, we demonstrated that Rac1 was a direct target of miR-142 in glioma, which was consistent with studies on hepatocellular carcinoma and osteosarcoma (18,32). We also observed that miR-142 overexpression suppressed the expression of Rac1 mRNA and protein. Furthermore, we found an inverse correlation between miR-142 expression and Rac1 in glioma tissues. Additionally, the effects of miR-142 alteration on cell migration and invasion of glioma cells were abolished by Rac1 modulation. Thus, these results indicated that miR-142 inhibited migration and invasion in glioma cells by directly blocking the Rac1 pathway.

Emerging evidence has revealed that Rac1-dependent signaling is essential for tumor initiation, development and progression (33). The expression of Rac1 has been demonstrated to be much higher in several human cancers, including glioma (34). The high expression of Rac1 induced glioma cell proliferation and inhibited apoptosis. Moreover, previous studies confirmed that Rac1 could promote migration and invasion by inducing epithelial-to-mesenchymal transition (EMT) and MMP expression (35), which have been identified as key regulators in the invasion and metastasis of diverse cancers. In the present study, we found miR-142 inhibited MMP expression by targeting Rac1. Rac1 restoration abrogated the suppressive effect of miR-142. Collectively, these data demonstrated the suppressive effect of miR-142 was mediated by targeting Rac1 to inhibit MMP expression in glioma.

In conclusion, we demonstrated that miR-142 was downregulated in glioma tissues and cell lines, and its decreased expression was associated with advanced clinicopathological features. Furthermore, we confirmed that miR-142 inhibited cell migration, invasion and MMP expression by inhibiting Rac1. These results revealed that miR-142 is a potential metastasis-associated tumor suppressor in glioma. Collectively, decreased miR-142 expression may play an important role in tumor metastasis and may be a novel prognostic factor and potential therapeutic target for glioma.

Acknowledgements

The present study was supported by a grant from the National Natural Scientific Foundation of China (no. 81403243).

References

1 

Vredenburgh JJ, Desjardins A, Reardon DA and Friedman HS: Experience with irinotecan for the treatment of malignant glioma. Neuro Oncol. 11:80–91. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Xie Q, Mittal S and Berens ME: Targeting adaptive glioblastoma: An overview of proliferation and invasion. Neuro Oncol. 16:1575–1584. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Preusser M, Haberler C and Hainfellner JA: Malignant glioma: Neuropathology and neurobiology. Wien Med Wochenschr. 156:332–337. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Vigneswaran K, Neill S and Hadjipanayis CG: Beyond the World Health Organization grading of infiltrating gliomas: Advances in the molecular genetics of glioma classification. Ann Transl Med. 3:952015.PubMed/NCBI

5 

Cohen AL and Colman H: Glioma biology and molecular markers. Cancer Treat Res. 163:15–30. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al: European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Davis FG and McCarthy BJ: Current epidemiological trends and surveillance issues in brain tumors. Expert Rev Anticancer Ther. 1:395–401. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Makeyev EV and Maniatis T: Multilevel regulation of gene expression by microRNAs. Science. 319:1789–1790. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Bartels CL and Tsongalis GJ: MicroRNAs: Novel biomarkers for human cancer. Clin Chem. 55:623–631. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Esquela-Kerscher A and Slack FJ: Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 6:259–269. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Liu Z, Dou C, Yao B, Xu M, Ding L, Wang Y, Jia Y, Li Q, Zhang H, Tu K, et al: Methylation-mediated repression of microRNA-129-2 suppresses cell aggressiveness by inhibiting high mobility group box 1 in human hepatocellular carcinoma. Oncotarget. 7:36909–36923. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Wu J, Cui H, Zhu Z and Wang L: MicroRNA-200b-3p suppresses epithelial-mesenchymal transition and inhibits tumor growth of glioma through down-regulation of ERK5. Biochem Biophys Res Commun. 478:1158–1164. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Deng B, Zhang Y, Zhang S, Wen F, Miao Y and Guo K: MicroRNA-142-3p inhibits cell proliferation and invasion of cervical cancer cells by targeting FZD7. Tumour Biol. 36:8065–8073. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Malzkorn B, Wolter M, Liesenberg F, Grzendowski M, Stühler K, Meyer HE and Reifenberger G: Identification and functional characterization of microRNAs involved in the malignant progression of gliomas. Brain Pathol. 20:539–550. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Cao XC, Yu Y, Hou LK, Sun XH, Ge J, Zhang B and Wang X: miR-142-3p inhibits cancer cell proliferation by targeting CDC25C. Cell Prolif. 49:58–68. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Pan D, Du Y, Ren Z, Chen Y, Li X, Wang J and Hu B: Radiation induces premature chromatid separation via the miR-142-3p/Bod1 pathway in carcinoma cells. Oncotarget. 7:60432–60445. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Ma Z, Liu T, Huang W, Liu H, Zhang HM, Li Q, Chen Z and Guo AY: MicroRNA regulatory pathway analysis identifies miR-142-5p as a negative regulator of TGF-β pathway via targeting SMAD3. Oncotarget. 7:71504–71513. 2016.PubMed/NCBI

18 

Zheng Z, Ding M, Ni J, Song D, Huang J and Wang J: miR-142 acts as a tumor suppressor in osteosarcoma cell lines by targeting Rac1. Oncol Rep. 33:1291–1299. 2015.PubMed/NCBI

19 

Su YH, Zhou Z, Yang KP, Wang XG, Zhu Y and Fa XE: MIR-142-5p and miR-9 may be involved in squamous lung cancer by regulating cell cycle related genes. Eur Rev Med Pharmacol Sci. 17:3213–3220. 2013.PubMed/NCBI

20 

Yin Y, Song M, Gu B, Qi X, Hu Y, Feng Y, Liu H, Zhou L, Bian Z, Zhang J, et al: Systematic analysis of key miRNAs and related signaling pathways in colorectal tumorigenesis. Gene. 578:177–184. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Shen WW, Zeng Z, Zhu WX and Fu GH: MiR-142-3p functions as a tumor suppressor by targeting CD133, ABCG2, and Lgr5 in colon cancer cells. J Mol Med. 91:989–1000. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Xu G, Wang J, Jia Y, Shen F, Han W and Kang Y: MiR-142-3p functions as a potential tumor suppressor in human osteosarcoma by targeting HMGA1. Cell Physiol Biochem. 33:1329–1339. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Kaduthanam S, Gade S, Meister M, Brase JC, Johannes M, Dienemann H, Warth A, Schnabel PA, Herth FJ, Sültmann H, et al: Serum miR-142-3p is associated with early relapse in operable lung adenocarcinoma patients. Lung Cancer. 80:223–227. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Qi X, Li J, Zhou C, Lv C and Tian M: MiR-142-3p suppresses SOCS6 expression and promotes cell proliferation in nasopharyngeal carcinoma. Cell Physiol Biochem. 36:1743–1752. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Pelloni M, Coltrinari G, Paoli D, Pallotti F, Lombardo F, Lenzi A and Gandini L: Differential expression of miRNAs in the seminal plasma and serum of testicular cancer patients. Endocrine. Oct 28–2016.(Epub ahead of print). https://doi.org/10.1007/s12020-016-1150-z View Article : Google Scholar : PubMed/NCBI

26 

Summerer I, Unger K, Braselmann H, Schuettrumpf L, Maihoefer C, Baumeister P, Kirchner T, Niyazi M, Sage E, Specht HM, et al: Circulating microRNAs as prognostic therapy biomarkers in head and neck cancer patients. Br J Cancer. 113:76–82. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Lin RJ, Xiao DW, Liao LD, Chen T, Xie ZF, Huang WZ, Wang WS, Jiang TF, Wu BL, Li EM, et al: MiR-142-3p as a potential prognostic biomarker for esophageal squamous cell carcinoma. J Surg Oncol. 105:175–182. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Li Y, Chen D, Jin LU, Liu J, Li Y, Su Z, Qi Z, Shi M, Jiang Z, Yang S, et al: Oncogenic microRNA-142-3p is associated with cellular migration, proliferation and apoptosis in renal cell carcinoma. Oncol Lett. 11:1235–1241. 2016.PubMed/NCBI

29 

Isobe T, Hisamori S, Hogan DJ, Zabala M, Hendrickson DG, Dalerba P, Cai S1, Scheeren F, Kuo AH1, Sikandar SS, et al: miR-142 regulates the tumorigenicity of human breast cancer stem cells through the canonical WNT signaling pathway. Elife. 3:doi: 10.7554/eLife.01977. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Schwickert A, Weghake E, Brüggemann K, Engbers A, Brinkmann BF, Kemper B, Seggewiß J, Stock C, Ebnet K, Kiesel L, et al: microRNA miR-142-3p inhibits breast cancer cell invasiveness by synchronous targeting of WASL, integrin alpha V, and additional cytoskeletal elements. PLoS One. 10:e01439932015. View Article : Google Scholar : PubMed/NCBI

31 

Xiao P and Liu WL: MiR-142-3p functions as a potential tumor suppressor directly targeting HMGB1 in non-small-cell lung carcinoma. Int J Clin Exp Pathol. 8:10800–10807. 2015.PubMed/NCBI

32 

Wu L, Cai C, Wang X, Liu M, Li X and Tang H: MicroRNA-142-3p, a new regulator of RAC1, suppresses the migration and invasion of hepatocellular carcinoma cells. FEBS Lett. 585:1322–1330. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Yoon CH, Hyun KH, Kim RK, Lee H, Lim EJ, Chung HY, An S, Park MJ, Suh Y, Kim MJ, et al: The small GTPase Rac1 is involved in the maintenance of stemness and malignancies in glioma stem-like cells. FEBS Lett. 585:2331–2338. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Zhong J, Bach CT, Shum MS and O'Neill GM: NEDD9 regulates 3D migratory activity independent of the Rac1 morphology switch in glioma and neuroblastoma. Mol Cancer Res. 12:264–273. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Man J, Shoemake J, Zhou W, Fang X, Wu Q, Rizzo A, Prayson R, Bao S, Rich JN and Yu JS: Sema3C promotes the survival and tumorigenicity of glioma stem cells through Rac1 activation. Cell Rep. 9:1812–1826. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2017
Volume 38 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qin W, Rong X, Dong J, Yu C and Yang J: miR-142 inhibits the migration and invasion of glioma by targeting Rac1. Oncol Rep 38: 1543-1550, 2017
APA
Qin, W., Rong, X., Dong, J., Yu, C., & Yang, J. (2017). miR-142 inhibits the migration and invasion of glioma by targeting Rac1. Oncology Reports, 38, 1543-1550. https://doi.org/10.3892/or.2017.5816
MLA
Qin, W., Rong, X., Dong, J., Yu, C., Yang, J."miR-142 inhibits the migration and invasion of glioma by targeting Rac1". Oncology Reports 38.3 (2017): 1543-1550.
Chicago
Qin, W., Rong, X., Dong, J., Yu, C., Yang, J."miR-142 inhibits the migration and invasion of glioma by targeting Rac1". Oncology Reports 38, no. 3 (2017): 1543-1550. https://doi.org/10.3892/or.2017.5816