MicroRNA‑218 inhibits the migration, epithelial‑mesenchymal transition and cancer stem cell properties of prostate cancer cells

  • Authors:
    • Bing Guan
    • Lijun Mu
    • Linlin Zhang
    • Ke Wang
    • Juanhua Tian
    • Shan Xu
    • Xinyang Wang
    • Dalin He
    • Yuefeng Du
  • View Affiliations

  • Published online on: June 1, 2018     https://doi.org/10.3892/ol.2018.8877
  • Pages: 1821-1826
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNA (miRNA) is a class of non‑coding single‑stranded RNA, able to regulate tumor‑associated genes via binding the 3'‑UTR of the target gene mRNA. Previous publications have demonstrated that miRNA‑218 (miR‑218) acts as a tumor‑suppressive miRNA in various types of human cancer, including prostate cancer (PCa). However, the role of miR‑218 in regulating PCa cell stemness and epithelial‑mesenchymal transition remains unknown and requires further research. In the present study, it is demonstrated that miR‑218 was downregulated in 2 PCa cell lines and could suppress cell migration, EMT and the exhibition of cancer stem cell‑like properties. The expression of GLI family zinc finger 1 (Gli1) was inhibited by miR‑218 overexpression, suggesting miR‑218‑suppression of Gli1 as a potential mechanism for the tumor‑suppressive effect of miR‑218. Overall, the results indicate that miR‑218 served a critical role in the inhibition of PCa development. This may provide new insight for elucidating the mechanisms of PCa oncogenesis and suggests that miR‑218 may be a novel therapeutic target for PCa.

Introduction

Prostate cancer (PCa) is the most common form of cancer among males in developed countries, with an estimated 648,400 new cases and 136,500 mortalities in 2008 (1). Currently, androgen-deprivation therapy is a key treatment for metastatic PCa. However, many patients develop castration-resistant PCa, which is a major cause of male mortality in developed countries (2). Further research is urgently required to develop more effective therapies for this disease.

MicroRNA (miRNA) is a class of non-coding single-stranded RNA that regulates tumor-associated genes via binding the 3′-UTR of target gene mRNA (35). Previous research has reported that miRNAs (miRs) serve important roles in human cancer biological processes, including initiation, development, migration and metastasis (68). In terms of the development of PCa, miR-34a inhibits prostate cancer stem cell (CSC) features and metastasis by directly repressing CD44 (9). Upregulation of miR-132/212 expression inhibits TGF-β-mediated epithelial-mesenchymal transition (EMT) of PCa cells by targeting SOX4 (10). Tumor-suppressive miR-29 inhibits cancer cell migration and invasion via targeting LAMC1 in PCa (11). Previous studies have also reported that miR-218 serves a tumor-suppressive role in PCa. However, the role of miR-218 in regulating PCa stemness and EMT remains uncharacterized.

In the present study, the expression of miR-218 in PCa cell lines was investigated, and the impact of miR-218 on tumor migration, EMT and CSC properties in PCa was investigated in vitro. The results demonstrate that miR-218 was downregulated in PCa cell lines and was able to suppress PCa cell migration, EMT and cancer stem cell properties. The expression of Gli1 was inhibited by miR-218 overexpression, suggesting a role for this protein in the mechanism of tumor-suppression of miR-218 in PCa. Altogether, the present study indicates that miR-218 served a critical role in inhibiting PCa development, providing new insights into clarifying the potential mechanisms of PCa oncogenesis and revealing that miR-218 may be a novel therapeutic target for PCa.

Materials and methods

Cell lines and cell culture

PCa cell lines LNCaP and C4-2 were obtained from the American Type Culture Collection (Manassas, VA, USA). BPH-1 cells were provided by Dr Jer-Tsong Hsieh (University of Texas Southwestern Medical Center, Dallas, TX, USA). These three cell lines were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with 10% fetal bovine serum (FBS; Hyclone; GE Healthcare Life Sciences, Logan, UT, USA) in a humidified chamber at 37°C in 5% CO2.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

Total RNA was extracted from harvested cells using TRIzol (Thermo Fisher Scientific, Inc.) and reverse transcribed to cDNA using the miScript II RT kit (Qiagen GmbH, Hilden, Germany), according to the manufacturers' protocols. The CFX96 PCR system (Bio-Rad Laboratories, Inc., Hercules, CA, USA) with SYBR-Green PCR Master Mix (Takara Bio, Inc., Otsu, Japan) was used to detect the transcriptional expression of miR-218. The thermocycling conditions were as follows: 95°C for 30 sec, followed by 40 cycles at 95°C for 5 sec, and then 60°C for 30 sec. U6 was used as an internal control, and relative gene expression was calculated using the 2−ΔΔCq method (12). The primer sequences used were as follows: miR-218 forward, 5′-CGAGTGCATTTGTGCTTGATCTA-3′ and reverse, 5′-TAATGGTCGAACGCCTAACGTC-3′; U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-TGGTGTCGTGGAGTCG-3′.

Lentivirus transfection

LNCaP and C4-2 cells were seeded and cultured for 24 h and to 40-50% confluence. The lentiviral vector 3 (LV3)-miR-218, constructed by GenePharma Co., Ltd. (Shanghai, China), was used to transfect cells in an overnight incubation. LV3 scrambled lentiviral vector (LV3-NC; GenePharma Co., Ltd.) was used as a negative control. At 48 h after infection, the stable clones were maintained by puromycin (2-3 µg/ml; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany)-resistant culturing.

Transwell migration assay

Complete growth medium (RPMI-1640 with 10% FBS, 1 ml) was added to each lower chamber as a chemoattractant. Transwell inserts with a pore diameter of 8-µm were used (Millipore; Merck KGaA). Cells, suspended in serum-free medium at 5-8×104 cells/ml, were seeded into the upper chamber at 400 µl/well. After an incubation of 20 h, the upper surface of the insert was wiped and cells that had migrated to the lower surface were fixed using 4% paraformaldehyde for 30 min and stained with 0.1% crystal violet for 20 min at room temperature. Cell number was counted in 6 random fields per well (magnification, ×200).

Western blot assay

Cells were washed 3 times in PBS before the protein was extracted using radioimmunoprecipitation assay buffer [50 mM Tris (pH 8.0), 150 mM NaCl, 0.1% SDS, 1% NP-40 and 0.5% sodium deoxycholate] with protease inhibitors. The concentration of protein was detected by Bradford assay protein quantitation kit (Abcam, Cambridge, UK). Proteins (30 µg) were separated by 12% SDS-PAGE and transferred into nitrocellulose membranes. Following blocking in 5% skim milk at room temperature for 1 h, the membranes were incubated with primary antibodies at 4°C overnight. Primary antibodies used were as follows: GAPDH (1:10,000; cat. no. KC-5G4; Kangchen Bio-tech Co., Ltd., Shanghai, China); E-cadherin (1:1,000; cat. no. sc-8426; Santa Cruz Biotechnology, Inc., Dallas, TX, USA); Vimentin (1:200; cat. no. sc-6260; Santa Cruz Biotechnology, Inc.); CD44 (1:800; cat. no. 3570; Cell Signaling Technology, Inc., Danvers, MA, USA); Oct4 (1:500; cat. no. ab18976; Abcam); Nanog (1:200; cat. no. ab21624; Abcam); and Gli1 (1:1,000; cat. no. 2643; Cell Signaling Technology, Inc.). The membranes were then washed in Tris-buffered saline with 0.1% Tween and incubated with horseradish peroxidase-conjugated secondary antibodies [goat anti-rabbit IgG (1:2,000; cat. no. ZB-2301); and goat anti-mouse IgG (1:2,000; cat. no. ZB-2305) (all from OriGene Technologies, Inc., Beijing, China) for 1 h at room temperature. Protein bands were visualized using a Molecular Imager ChemiDoc XRS System (Bio-Rad Laboratories).

Colony-forming and tumor sphere formation assays

For the colony-forming assay, 2×103 cells were seeded into each well of a 6-well plate and incubated for 10-14 days. Following 3 washes in PBS, cells were fixed using 4% paraformaldehyde for 30 min at room temperature and stained with 0.1% crystal violet for 20 min at room temperature. The tumor sphere formation assay was performed by seeding 1×104 cells to each well of low-adhesion 6-well plate in serum-free Dulbecco's modified Eagle/F12 medium supplemented with 20 ng/ml epidermal growth factor, 10 ng/ml basic fibroblast growth factor and 2% B27 (all Invitrogen; Thermo Fisher Scientific, Inc.). After 2 weeks, plates were analyzed for tumor sphere formation using an inverted microscope (magnification, ×200).

Statistical analysis

All statistical analysis was performed using GraphPad Prism version 6.0 (GraphPad Software, Inc., La Jolla, CA, USA). Differences between 2 groups were compared using the Student's t-test. For comparisons of ≥3 groups, one-way analysis of variance followed by Tukey's post hoc test was used. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-218 expression is downregulated in PCa cells

RT-qPCR was performed to compare the expression of miR-218 in PCa cells with that in healthy prostate epithelial cells. This revealed that the expression of miR-218 was notably downregulated in PCa cell lines, LNCaP and C4-2, compared with the normal prostate epithelial cells (BPH-1; Fig. 1A).

Construction of miR-218-overexpressing PCa cells

In order to reveal the effects of miR-218 on PCa migration, EMT and CSC properties, miR-218-overexpressing LNCaP/C4-2 cells were constructed by transfecting the cells with LV3-miR-218 lentiviral vectors. Subsequently, cells were analyzed by RT-qPCR to confirm miR-218-overexpression (Fig. 1B).

Overexpression of miR-218 inhibits PCa cell migration and EMT

The Transwell migration assays demonstrated that the migration of LNCaP/C4-2 cells was suppressed by miR-218 overexpression (Fig. 2). Western blotting of EMT markers demonstrated that miR-218 overexpression caused a slight increase in the expression of E-cadherin and a significant decrease in the expression of vimentin (Fig. 3A). These results suggest that overexpression of miR-218 inhibits PCa cell migration and EMT.

Overexpression of miR-218 diminishes PCa cell stemness properties

Protein expression profiling of cancer stemness markers was performed by western blotting. The results indicate that overexpression of miR-218 downregulated the expression of CD44, Oct4 and Nanog (Fig. 3B). Colony forming assays were performed to assess the self-renewal capacity of PCa cells. It was demonstrated that the miR-218-overexpressing LNCaP cells formed fewer and smaller colonies than the control cells (Fig. 4A). Similar results were obtained from the clonogenic assay performed in C4-2 cells (Fig. 4B), indicating that miR-218 may serve a critical role in tumor growth inhibition. The tumor sphere formation assay is well established for measuring the self-renewing capability of stem cells. In these experiments, the control cells generated more tumor-spheres than the miR-218-overexpressing cells in both cell lines (Fig. 4C and D). Therefore, these results suggest that overexpression of miR-218 diminished PCa cell stemness properties.

Glil expression is downregulated by miR-218 overexpression

Numerous studies have indicated that the Hedehog-Gli signaling pathway serves a critical role in cancer cell EMT occurrence and CSC generation (1315). In the preset study, western blot analysis indicated that the expression of Gli1 was inhibited by miR-218 overexpression (Fig. 5), indicating that miR-218 suppression of Gli1 may be a mechanism for the anticancer effect of miR-218 in PCa.

Discussion

Advances have been made in PCa diagnosis and treatment in recent years (16). However, management of PCa remains a challenge. It has been widely reported that EMT and CSCs are critical for cancer initiation and development (1721). However, the molecular mechanisms by which EMT and CSCs execute their effects require further investigation.

Previous studies have demonstrated that miRNAs serve important roles in human cancer biological processes, including initiation, development, migration and metastasis (2226). miR-218 can act as a tumor suppressor and is downregulated in various types of human cancer (2731). miR-218 can inhibit cancer cell proliferation, invasion, migration, EMT, lymph node metastasis and self-renewal in glioma, cervical cancer, gastric cancer and bladder cancer, among others (3236). One study indicated that miR-218 expression is decreased in PCa, and impedes IL-6-induced PCa cell proliferation and invasion via suppression of LGR4 expression (37). miR-218 has also been demonstrated to inhibit PCa cell growth and promote apoptosis by repressing TPD52 expression (38), as well as inhibit PCa cell migration and invasion via targeting LASP1 (39). We hypothesize that miR-218 may also inhibit PCa cell migration, EMT and CSC properties. However, the underlying role of miR-218 in regulating PCa stemness maintenance and EMT is poorly characterized at present.

In the present study, the expression of miR-218 was notably downregulated in PCa cells and the LNCaP/C4-2 cell constructs overexpressing miR-218 indicated that miR-218 inhibited PCa cell migration. Results of western blotting revealed that the overexpression of miR-218 downregulated the expression of vimentin, CD44, Oct4 and Nanog. In colony-forming assays, miR-218-overexpressing cells formed fewer and smaller colonies than the control cells. Consistently, the control cells generated more tumor-spheres than miR-218-overexpressing cells, suggesting that overexpression of miR-218 inhibited PCa stemness properties. Copious evidence has indicated that the Hedgehog-Gli signaling pathway serves a critical role in cancer cell EMT and CSC generation (1315,40). In the present study, the expression of Gli1 was inhibited by miR-218 overexpression. This indicates that miR-218 suppression of Gli1 may serve in the mechanism by which miR-218 inhibits EMT and stemness maintenance in PCa.

The regulatory mechanisms of human CSC maintenance and EMT are very complex. If miR-218 targets Gli1 by binding its 3′-UTR, or other molecular mediators between miR-218 and Gli1, the mechanism requires further investigation. In conclusion, the present study indicates that miR-218 served a critical role in inhibiting the migration, EMT and CSC properties of PCa cells. This provides a new insight for clarifying the potential mechanisms of PCa oncogenesis, and indicates that miR-218 may be a potential therapeutic target for PCa.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China, (grant nos., 81372736 and 81272811) awarded to YD and LZ, respectively.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

YD and DH designed and supervised all experiments and contributed to the manuscript preparation. BG performed the experiments, analyzed the data and contributed to the manuscript preparation. LM and JT contributed to the cell culture and lentivirus transfection. LZ and KW contributed to the data analysis. SX and XW contributed to western blot assay and manuscript preparation.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Egan A, Dong Y, Zhang H, Qi Y, Balk SP and Sartor O: Castration-resistant prostate cancer: Adaptive responses in the androgen axis. Cancer Treat Rev. 40:426–433. 2014. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, Bartel DP, Linsley PS and Johnson JM: Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 433:769–773. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Guo H, Ingolia NT, Weissman JS and Bartel DP: Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 466:835–840. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Iorio MV and Croce CM: MicroRNAs in cancer: Small molecules with a huge impact. J Clin Oncol. 27:5848–5856. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, et al: MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 65:7065–7070. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Zhang X, Tang J, Zhi X, Xie K, Wang W, Li Z, Zhu Y, Yang L, Xu H and Xu Z: miR-874 functions as a tumor suppressor by inhibiting angiogenesis through STAT3/VEGF-A pathway in gastric cancer. Oncotarget. 6:1605–1617. 2015.PubMed/NCBI

9 

Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, Patrawala L, Yan H, Jeter C, Honorio S, et al: The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med. 17:211–215. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Fu W, Tao T, Qi M, Wang L, Hu J, Li X, Xing N, Du R and Han B: MicroRNA-132/212 upregulation inhibits TGF-β-mediated epithelial-mesenchymal transition of prostate cancer cells by targeting SOX4. Prostate. 76:1560–1570. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Nishikawa R, Goto Y, Kojima S, Enokida H, Chiyomaru T, Kinoshita T, Sakamoto S, Fuse M, Nakagawa M, Naya Y, et al: Tumor-suppressive microRNA-29s inhibit cancer cell migration and invasion via targeting LAMC1 in prostate cancer. Int J Oncol. 45:401–410. 2014. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Xu X, Su B, Xie C, Wei S, Zhou Y, Liu H, Dai W, Cheng P, Wang F, Xu X and Guo C: Sonic hedgehog-Gli1 signaling pathway regulates the epithelial mesenchymal transition (EMT) by mediating a new target gene, S100A4, in pancreatic cancer cells. PLoS One. 9:e964412014. View Article : Google Scholar : PubMed/NCBI

14 

Takebe N, Harris PJ, Warren RQ and Ivy SP: Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nat Rev Clin Oncol. 8:97–106. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Clement V, Sanchez P, de Tribolet N, Radovanovic I and Ruiz i Altaba A: HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr Biol. 17:165–172. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Ahmed A, Ali S and Sarkar FH: Advances in androgen receptor targeted therapy for prostate cancer. J Cell Physiol. 229:271–276. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Brabletz T, Jung A, Spaderna S, Hlubek F and Kirchner T: Opinion: Migrating cancer stem cells-an integrated concept of malignant tumour progression. Nat Rev Cancer. 5:744–749. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Deshmukh A, Deshpande K, Arfuso F, Newsholme P and Dharmarajan A: Cancer stem cell metabolism: A potential target for cancer therapy. Mol Cancer. 15:692016. View Article : Google Scholar : PubMed/NCBI

19 

Colak S and Medema JP: Cancer stem cells-important players in tumor therapy resistance. FEBS J. 281:4779–4791. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Thiery JP, Acloque H, Huang RY and Nieto MA: Epithelial-mesenchymal transitions in development and disease. Cell. 139:871–890. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Dave B, Mittal V, Tan NM and Chang JC: Epithelial-mesenchymal transition, cancer stem cells and treatment resistance. Breast Cancer Res. 14:2022012. View Article : Google Scholar : PubMed/NCBI

22 

Miska EA: How microRNAs control cell division, differentiation and death. Curr Opin Genet Dev. 15:563–568. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Garzon R, Marcucci G and Croce CM: Targeting microRNAs in cancer: Rationale, strategies and challenges. Nat Rev Drug Discov. 9:775–789. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Li X, Yu Z, Li Y, Liu S, Gao C, Hou X, Yao R and Cui L: The tumor suppressor miR-124 inhibits cell proliferation by targeting STAT3 and functions as a prognostic marker for postoperative NSCLC patients. Int J Oncol. 46:798–808. 2015. View Article : Google Scholar : PubMed/NCBI

25 

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. View Article : Google Scholar : PubMed/NCBI

26 

Zhou N, Fei D, Zong S, Zhang M and Yue Y: MicroRNA-138 inhibits proliferation, migration and invasion through targeting hTERT in cervical cancer. Oncol Lett. 12:3633–3639. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Davidson MR, Larsen JE, Yang IA, Hayward NK, Clarke BE, Duhig EE, Passmore LH, Bowman RV and Fong KM: MicroRNA-218 is deleted and downregulated in lung squamous cell carcinoma. PLoS One. 5:e125602010. View Article : Google Scholar : PubMed/NCBI

28 

Yu H, Gao G, Jiang L, Guo L, Lin M, Jiao X, Jia W and Huang J: Decreased expression of miR-218 is associated with poor prognosis in patients with colorectal cancer. Int J Clin Exp Pathol. 6:2904–2911. 2013.PubMed/NCBI

29 

Guan H, Wei G, Wu J, Fang D, Liao Z, Xiao H, Li M and Li Y: Down-regulation of miR-218-2 and its host gene SLIT3 cooperate to promote invasion and progression of thyroid cancer. J Clin Endocrinol Metab. 98:E1334–E1344. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Wang XX, Ge SJ, Wang XL, Jiang LX, Sheng MF and Ma JJ: miR-218 tissue expression level is associated with aggressive progression of gastric cancer. Genet Mol Res. 15:2016.

31 

Wang HT, Liu AG, Luo DS, Zhou ZN, Lin HG, Chen RZ, He JS and Chen K: miR-218 expression in osteosarcoma tissues and its effect on cell growth in osteosarcoma cells. Asian Pac J Trop Med. 7:1000–1004. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Wang G, Fu Y, Liu G, Ye Y and Zhang X: miR-218 inhibits proliferation, migration, and EMT of gastric cancer cells by targeting WASF3. Oncol Res. 25:355–364. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Jiang Z, Song Q, Zeng R, Li J, Li J, Lin X, Chen X, Zhang J and Zheng Y: MicroRNA-218 inhibits EMT, migration and invasion by targeting SFMBT1 and DCUN1D1 in cervical cancer. Oncotarget. 7:45622–45636. 2016.PubMed/NCBI

34 

Tu Y, Gao X, Li G, Fu H, Cui D, Liu H, Jin W and Zhang Y: MicroRNA-218 inhibits glioma invasion, migration, proliferation, and cancer stem-like cell self-renewal by targeting the polycomb group gene Bmi1. Cancer Res. 73:6046–6055. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Kogo R, How C, Chaudary N, Bruce J, Shi W, Hill RP, Zahedi P, Yip KW and Liu FF: The microRNA-218~Survivin axis regulates migration, invasion, and lymph node metastasis in cervical cancer. Oncotarget. 6:1090–1100. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Cheng Y, Yang X, Deng X, Zhang X, Li P, Tao J and Lu Q: MicroRNA-218 inhibits bladder cancer cell proliferation, migration, and invasion by targeting BMI-1. Tumour Biol. 36:8015–8023. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Li F, Gu C, Tian F, Jia Z, Meng Z, Ding Y and Yang J: MiR-218 impedes IL-6-induced prostate cancer cell proliferation and invasion via suppression of LGR4 expression. Oncol Rep. 35:2859–2865. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Han G, Fan M and Zhang X: MicroRNA-218 inhibits prostate cancer cell growth and promotes apoptosis by repressing TPD52 expression. Biochem Biophys Res Commun. 456:804–809. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Nishikawa R, Goto Y, Sakamoto S, Chiyomaru T, Enokida H, Kojima S, Kinoshita T, Yamamoto N, Nakagawa M, Naya Y, et al: Tumor-suppressive microRNA-218 inhibits cancer cell migration and invasion via targeting of LASP1 in prostate cancer. Cancer Sci. 105:802–811. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Inaguma S, Kasai K, Hashimoto M and Ikeda H: GLI1 modulates EMT in pancreatic cancer-letter. Cancer Res. 72:3702–3703. 3704–3705. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 16 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guan B, Mu L, Zhang L, Wang K, Tian J, Xu S, Wang X, He D and Du Y: MicroRNA‑218 inhibits the migration, epithelial‑mesenchymal transition and cancer stem cell properties of prostate cancer cells. Oncol Lett 16: 1821-1826, 2018
APA
Guan, B., Mu, L., Zhang, L., Wang, K., Tian, J., Xu, S. ... Du, Y. (2018). MicroRNA‑218 inhibits the migration, epithelial‑mesenchymal transition and cancer stem cell properties of prostate cancer cells. Oncology Letters, 16, 1821-1826. https://doi.org/10.3892/ol.2018.8877
MLA
Guan, B., Mu, L., Zhang, L., Wang, K., Tian, J., Xu, S., Wang, X., He, D., Du, Y."MicroRNA‑218 inhibits the migration, epithelial‑mesenchymal transition and cancer stem cell properties of prostate cancer cells". Oncology Letters 16.2 (2018): 1821-1826.
Chicago
Guan, B., Mu, L., Zhang, L., Wang, K., Tian, J., Xu, S., Wang, X., He, D., Du, Y."MicroRNA‑218 inhibits the migration, epithelial‑mesenchymal transition and cancer stem cell properties of prostate cancer cells". Oncology Letters 16, no. 2 (2018): 1821-1826. https://doi.org/10.3892/ol.2018.8877