miR-30a inhibits glioma progression and stem cell‑like properties by repression of Wnt5a

  • Authors:
    • Yonghong Zhang
    • Zhi Wu
    • Lichao Li
    • Min Xie
  • View Affiliations

  • Published online on: June 16, 2017     https://doi.org/10.3892/or.2017.5728
  • Pages: 1156-1162
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

miR-30a has been found to be dysregulated in diverse cancers and involved in the regulation of tumor progression. However, there is scarce research on the role of miR-30a in glioma. In the present study, we assessed the expression level of miR-30a in glioma tissues and cell lines. The microRNA microarray analysis revealed low expression of miR-30a in glioma tissues and cells vs. the control. Furthermore, we found that stable miR-30a inhibited cell proliferation, G1 phase arrest and stem cell-like formation in glioma. Moreover, to investigate the molecular mechanism of miR-30a on glioma cell phenotypes, we identified Wnt5a as a new direct target gene for miR-30a by bioinformatic assay, luciferase assay and western blot analysis. Further functional studies suggested that miR-30a suppressed metastasis, sphere formation and glioma growth by targeting Wnt5a signal pathway. Collectively, our findings suggested for the first time that miR-30a may function as a tumor suppressor in glioma by targeting Wnt5a.

Introduction

With its incidence rate of almost 50%, glioma is the most common form among CNS gliomas. Although new therapeutic approaches have been developed, there are still many common problems that need to be resolved including therapy resistance to improve long-term survival. The origin of gliomas is largely unknown but there is increasing speculation that they might arise from glioma stem cells (GSCs), which might consist of transformed neural stem cells (14). Recent advances in our understanding of the biological features of glioma offer opportunities to the design of a new therapeutic strategy based on targeting essential signaling pathways.

MicroRNAs (miRNAs) are a family of endogenous small non-coding RNAs that regulate gene expression via the sequence-specific base pairing on the 3′-untranslated regions (3UTRs) of target mRNAs, leading to mRNA cleavage or translation inhibition (5). A great number of the human miRNAs function either as oncogenes or as tumor suppressors. Thus, their function acting as tumor-suppressor or carcinogenic miRNAs may vary depending on their targets (68), resulting in influencing glioma formation and growth. The miRNAs have been reported to be aberrantly overexpressed or downregulated during glioma progression, including miR-20a and miR-106a (9), miR-29a (10), miR-145 (11), miR-656 (12), miR-300 (13), miR-16 (14), miR-34a (15), miR-503 (16), miR-203 (17) miR-100 (18), miR-26a (19), miR-23b (20) and miR-218 (21). These miRNAs play oncogenic or tumor-suppressive roles in the regulation of cell growth, migration and invasion by repressing their target genes.

miRNA 30a (miR-30a) is a member of the miR-30 family, which consists of six distinct mature miRNA sequences. There is considerable evidence suggesting that the dysregulation of miR-30a is correlated with several types of malignant tumors, including breast, lung, thyroid, gastric cancer and leukemia (22). Metadherin (23), SNAIL1 (24), Beclin-1 (25) and PIK3CD (26) are potential targeted genes of miR-30a, which promotes glioma progression. However, there are relatively few studies available that report a role for miR-30a in tumorigenesis and progression of glioma. In the present study, we investigated the potential involvement of miR-30a by examining its expression and its effects on tumorigenesis, cell growth, cell cycle distribution, colony formation, migration, invasion and stem cell-like properties in glioma. Thus, mechanistic investigation revealed that miR-30a was shown to control Wnt5a expression in progression of glioma as a tumor suppressor.

Materials and methods

Clinical samples

Primary glioma tissue samples and normal samples were obtained from the patients in the First Hospital of Lanzhou University (Lanzhou, China). None of the patients had received either radiotherapy or chemotherapy. Both tumor and normal tissues were histologically confirmed by H&E (hematoxylin and eosin) staining. The tumor tissues derived from 16 cases with glioma, and the normal tissues derived from patients with brain injury. Informed consent was obtained from each patient and the research protocols were approved by the Ethics Committee of Lanzhou University Hospital.

Cell culture

Human glioma cells T98G, SHG44, U251, U87 and U373 were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA) were cultured in Dulbeccos modified Eagles medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 µg/ml streptomycin. All cells were cultured at 37°C in a humidified atmosphere containing 5% CO2. Human normal cells HA (ScienCell Research Laboratories, Carlsbad, CA, USA) isolated from human brain (cerebral cortex) were cultured in microglia medium with 10% FBS.

Oligonucleotide synthesis and lentiviral transduction

The oligonucleotide of mature miR-30a antagomir was chemosynthesized, amplified and cloned into GV232-Puro Vectors by Shanghai Genechem, Co., Ltd. (Shanghai, China). The correct sequences and insertions were confirmed by DNA sequencing. Cells were lentivirus-transfected with either the GV232-Puro-miR-30a recombined vector (LV-miR-30a) or emptyGV232-Puro vector (negative control, miR-control) using Lipofectamine 2000 transfection reagent (Invitrogen, Carlsbad, CA, USA). The transduced cells with a cell density of over 40% confluency were exposed to puromycin dihydrochloride (1 mg/ml; Sigma-Aldrich, St. Louis, MO, USA) for resistance selection. Stable cell lines were selected with 0.5 mg/ml puromycin in the first round of selection. Lentivirus-mediated silencing of miR-30a was verified by quantitative reverse transcription-PCR (qRT-PCR) and western blot analysis.

miRNA target validation

A fragment of Wnt5a 3UTR (untranslated regions) was amplified by PCR and cloned downstream of the firefly luciferase gene in pGL4 vector (Promega, Madison, WI, USA). The vector was named wild-type 3UTR. Site-directed mutagenesis of the miR-30a binding site in Wnt5a 3UTR was performed using GeneTailor Site-Directed Mutagenesis system (Invitrogen) and named mutant 3UTR.

RNA isolation and real-time RT-PCR

Total RNA was extracted using TRIzol reagent (Invitrogen) following the instructions. Briefly, the cells were lysed in TRIzol and then mixed with chloroform. The lysate was centrifuged to separate RNA, DNA and protein, the total RNA recovered was precipitated with isopropanol, washed in 75% ethanol to remove impurities before dissolved in water. Subsequently, 2 µg of RNA was treated with DNase to remove contaminating DNA prior to reverse transcription to cDNA using SYBR® PCR kit (Takara Bio, Shiga, Japan). With the primers purchased from Invitrogen, real-time RT-PCR was performed using a sequence detector (ABI Prism; Applied Biosystems, Foster City, CA, USA) to measure mRNA expression. The relative expression levels were quantified by comparing Ct values of the samples with those of the reference, the data were normalized to the internal control GAPDH.

Cell viability assay

To detect the growth of glioma cells and the growth curve, cell viability was assessed by MTT assay. Logarithmic phase cells were seeded in 96-well culture plates at a density of 5×103 cells/well [the edge wells of the plate are filled with aseptic phosphate-buffered saline buffer (PBS)]. The cells were incubated at 37°C, 5% CO2 until cells covered the bottom of the well. A total of 20 µl of the MTT solution was added to each well (5 mg/ml, 0.5% MTT) and the cells were cultured for 4 h at 37°C. After the incubation, the supernatant was discarded and 150 µl dimethyl sulfoxide was added to each well. Afterwards, the culture plate was shaken at low speed for 10 min until the crystals dissolved completely. The ELISA reader was used to measure the absorbance at 570 nm.

Colony formation assay

Cells in logarithmic growth phase were digested in 0.5% trypsin/0.04% EDTA and single cell suspension was prepared. Then, these cells were added to 6-well plates (200 cells/well) followed by incubation at 37°C in a humidified incubator containing 5% CO2 for 24 h. Non-adherent cells were removed. After culture for 10–14 days, colonies were present. These cells were seeded into 96-well plates followed by incubation at 37°C in an environment with saturated humidity and 5% CO2. The colony formation efficiency and the morphology of colonies were photographed using a microscope. The colony size and cells in each colony were measured.

Cell cycle assay

Total cells were collected by trypsinization. Bovine pancreatic RNAse (Wuhan Boster Biological Technology, Ltd., Wuhan, China) was added at a final concentration of 2 µg/ml, incubated at 37°C for 30 min, then 20 µg/ml propidium iodide (PI) was added and incubated for 20 min at 25°C. Cells (5×104) were analyzed by FACSCalibur flow cytometer (Becton-Dickinson, Franklin Lakes, NJ, USA) in each group.

Western blot analysis

Total cells were lysed using the RIPA (Bio-Rad Laboratories, Inc., Philadelphia, PA, USA) buffer (50 mM Tris-HCl, pH 8.0, 150 mM NaCl, 1 mM dithiothreitol, 0.1% SDS). Proteins were separated by SDS-PAGE and transferred to polyvinylidene difluoride (PVDF) membranes (Millipore, Bedford, MA, USA) at 55 V for 4 h at 4°C. After blocking, the membranes were incubated with primary antibodies overnight at 4°C, washed three times with TBS Tween-20, and followed by secondary antibodies conjugated with horseradish peroxidase at 1:5,000 dilution in TBS for 1 h at 25°C. Western blotting were visualized on X-ray film by an automated chemiluminescence system.

Luciferase assay

For reporter assays, wild-type or mutation 3UTR vector and the control vector pRL-CMV (cytomegalovirus; coding for Renilla luciferase; Promega) were cotransfected. Luciferase activity was measured 36 h after transfection. Firefly and Renilla luciferase reporter activity was measured using Luc-Pair Duo-luciferase Assay kit 2.0 as per manufacturers instructions.

Migration assay

In order to evaluate the effect of metastatic properties, Transwell migration assays was conducted. Glioma cells (1×105) were placed in the top chamber onto the non-coated membrane and allowed to migrate toward DMEM medium containing fetal calf serum (FCS) in the lower chamber. After incubation with methanol, cells were fixed for 24 h and stained with 0.1% crystal violet (Sigma-Aldrich). The number of cells was counted using a light microscope.

Invasion assay

In order to evaluate the effect of metastatic properties, invasion assay was conducted. Cells (1×105) were placed in the top chamber onto the Matrigel coated membrane. Each well was coated freshly with Matrigel (60 µg; BD Biosciences, San Jose, CA, USA). Cells were placed in serum-free medium or growth factors, and medium containing serum was used as a chemoattractant in the lower chamber. After incubating for 48 h, cells that did invade via the pores were removed by a cotton swab. Cancer stem cells (CSCs) on the lower surface of the membrane were fixed in methanol and then stained with crystal violet. The number of cells was counted using a light microscope (27).

Tumor sphere assay

Sphere formation assay (27) was performed as described. In brief, cells were plated in 6-well ultralow attachment plates (Corning Inc., Corning, NY, USA) at a density of 1×103 cells/ml in DMEM supplemented with 1% N2 supplement (Invitrogen), 2% B27 supplement (Invitrogen), 20 ng/ml human platelet growth factor (Sigma-Aldrich), 100 ng/ml epidermal growth factor (Invitrogen) and 1% antibiotic-antimycotic (Invitrogen) at 37°C in a humidified atmosphere containing 5% CO2. Sphere were collected after 7 days and dissociated with accutase (Innovative Cell Technologies, Inc., San Diego, CA, USA). The cells obtained from dissociation were sieved through a 40-µm filter, and counted by coulter counter using trypan blue dye.

Mouse xenograft models

The mice (6-weeks-old) were purchased from Beijing Weitongli, Co., Ltd. (Beijing, China). A total of 1.0×106 cells, either stably expressing glioma cells LV-miR-30a or LV-miR control and LV-miR-30a with Wnt5a overexpression or LV-miR control with Wnt5a overexpression, were injected subcutaneously into the abdomen of each mouse, respectively. After the tumors were ~100 mm3, mice were examined for the effects of tumor burden and tumor growth, every two days and tumor measurements were performed weekly. Tumor volume was calculated using the formula: Tumor volume = [length × width2]/2 as previously reported. Approximately 3 weeks after inoculation, the mice were euthanized by subcutaneous injection with sodium pentobarbital (40 mg/kg) and the tumors were weighed, all mice were handled according to the protocol approved by the Committee on the Ethics of Animal Experiments of the hospital. All tumors were dissected, and sizes and weights were measured and recorded (28).

Statistical analysis

All data were expressed as the mean ± SD of at least three independent experiments. Differences between the groups were analyzed by one- or two-way ANOVA, followed by Bonferonis multiple comparison tests using PRISM statistical analysis software (GrafPad Software, Inc., San Diego, CA, USA). Differences at P<0.05 were considered as significance level.

Results

Low expression of miR-30a in human glioma

To explore the possible action of miR-30a in glioma, the expression of miR-30a was examined and quantified by real-time RT-PCR at both tissue and cell levels. We examined the expression of miR-30a in 16 cases with glioma and their compared normal tissues. As shown in Fig. 1A, the expression levels of miR-30a in glioma samples were lower than those in normal samples. Similarly, miR-30a in human glioma cells was shown significantly reduced comparing with control cells (Fig. 1B). It is suggested that miR-30a may function as a tumor suppressor in the progression of human glioma.

miR-30a suppresses proliferation and mediates the accumulation of G1-phase glioma cells

To investigate the effect of miR-30a on cell proliferation, U87 and U251 cells were transfected with LV-miR-control or LV-miR-30a, respectively. MTT and colony formation assay showed that miR-30a inhibited cell proliferation in U251 cells (Fig. 2A and C) and U87 cells (Fig. 2B and D). In addition, there was no difference on cell proliferation between cells with the LV-miR-control and the control cells (data not shown). To further ascertain miR-30a mediating growth inhibition, cells with LV-miR-30a and cell cycle distribution were examined. U251 cells infected with LV-miR-30a had an increased percentage of cells in G1 phase but fewer cells in S phase comparing with the control (Fig. 2E and F). As shown in Fig. 2, there was a correlation between the growth-suppressive effect of miR-30a and the G0/G1 phase arrest. Therefore, the accumulation of G1-phase glioma cells mediated by miR-30a is as a direct cause of the cell proliferation inhibition.

miR-30a inhibits stem cell-like properties in glioma

To investigate the effects of miR-30a on glioma metastasis and stem cell-like properties, the Transwell migration assays and invasion assays of U251 cells with LV-miR-30a were performed. It is shown that upregulation of miR-30a significantly decreased migration (Fig. 3A) and invasion (Fig. 3B) exposed to TGF-β. The sphere formation of U251 cells with LV-miR-30a was much lower than the control (Fig. 3C). Metastasis associated markers were also detected by western blot analysis and the results showed that vimentin and SNAIL1 decreased in the U251 cells with LV-miR-30a (Fig. 3D). The above indicated that miR-30a prevented the glioma cell from stem-like cells.

Wnt5a is a target gene for miR-30a

To further explore the possible molecular mechanisms of miR-30a-mediated glioma progress inhibition, we applied the bioinformatic analysis to search the potential targets of miR-30a. It is shown that Wnt5a is considered to be directly suppressed by miR-30a (Fig. 4A). The luciferase activity of pGL4-Wnt5α-WT in U251 cells was much lower than in control cells (Fig. 4B). Moreover, pGL4-Wnt5a-Mut luciferase activity was rescued. Furthermore, we examined whether miR-30a could regulate the expression of endogenous Wnt5a in U251 cells. Comparing with the control, the mRNA levels of endogenous Wn5a (Fig. 4C) were downregulated when cells were transected with miR-30a. The expression of Wnt5a increased in the cells with anti-miR-30a (Fig. 4D and E). These data indicated that Wnt5a acted as a new target gene for miR-30a.

miR-30a inhibits metastasis and sphere formation by targeting Wnt5a signal pathway in glioma cells

In view of the fact that Wnt5a was the latent target of miR-30a, over-expression of Wnt5a was performed to test whether miR-30a regulates metastasis and sphere formation by targeting Wnt5a signal pathway in glioma cells. It is suggested that miR-30a in U251 cells inhibited cell migration and invasion with Wnt5a overexpression (Fig. 5A and B). Sphere formation assay also demonstrated that miR-30a inhibited the self-renewal ability of glioma cells (Fig. 5C). Since Wnt signaling pathway is an important pathway involved in primary glioma, we then detected several downstream proteins in Wnt signaling pathway. The protooncogene cMyc, downstream of the Wnt singling pathway, was decreased by miR-30a overexpression. A similar result was obtained for phosphorylated JNK, an important downstream protein of Wnt5a (Fig. 5D).

miR-30a inhibits glioma growth by targeting Wnt5a signal pathway in vivo

Based on the above effect of miR-30a observed in in vitro experiments, we further investigate miR-30a mediating growth inhibition in vivo. The xenograft model of glioma in nude mice was applied using U251-miR-30a and U251-miR control. Next, we measured the tumor size every 5 days and plotted the growth curve against the average tumor size. The results showed that miR-30a suppressed glioma growth in vivo (Fig. 6A).

Discussion

Although previous studies have shown that miR-30a may act as a tumor suppressor or oncogene, the detailed mechanism of miR-30a involvement in glioma is not well understood. In the present study, we found that the glioma tissues had a significant downregulation of the expression of miR-30a comparing with the normal tissues. From this finding, we aimed to clear the suppressor role of miR-30a in glioma. Thus, we also discovered that miR-30a induced G1 arrest of glioma cells and suppressed cell proliferation. Our finding suggested that miR-30a suppressed the progression of glioma by targeting Wnt5a for the first time. In addition, overexpression of miR-30a in U251 cells restrained oncogenesis in nude mice, which showed that miR-30a acts as a tumor suppressor in glioma.

Over the past few decades, many studies have focused on the cancer-related role of miR-30 family. This family is widely known to be implicated in various cellular processes including cell differentiation, organ development and substance metabolism (29,30). Several studies indicated that miR-30a could regulate breast cancer cell proliferation and migration and reduce the oncogenic abilities of breast and lung cancer depending on their targets (3133). In order to investigate potential target genes of miR-30a, we used bioinformatics analyses to search the latent target genes and than discovered that Wnt5a was a novel potential target gene of miR-30a. Wnt family members are classified into two groups: Wnt1, Wnt3a and Wnt7a, activate the β-catenin pathway, and have been shown to be present in mammals (34,35). Another group including Wnt2, Wnt4, Wnt5a, Wnt5b, Wnt6 and Wnt11, activate a β-catenin-independent pathway that primarily regulates cell motility and polarity (34,35). Moreover, β-catenin-independent pathway is known to activate various protein kinases including protein kinase C (PKC), Ca2+/calmodulin-dependent protein kinase II, Rho-associated kinase and c-jun N-terminal kinase (JNK). In particular, Wnt5a is a representative of the Wnt family that activate the β-catenin-independent pathway and distinct routes (36,37). Previous research showed that Wnt5a stimulates proliferation and migration in cancer cells. Moreover, Wnt5a expression is related with the aggressiveness of ocular melanoma, lung, breast and gastric cancer, indicating that Wnt5a has oncogenic properties (36,37). A recent study showed that the glioma cells were found to express significantly high levels of Wnt5a. It was also suggested that Wnt5a promotes invasion activities of glial cells at least via the activation and expression of JNK and matrix metalloproteinase-1 (MMP-1) (37). In this study, we proved that Wnt5a was a novel target gene of miR-30a that could inhibit metastasis and sphere formation.

In conclusion, we identified miR-30a as a tumor suppressor miRNA in glioma, and low miR-30a expression may become an unfavorable prognostic factor in patients with glioma. Furthermore, miR-30a functions as the tumor suppressor in human glioma by targeting Wnt5a. These findings indicated that miR-30a acts as a potential target for treating glioma and the key function of miR-30a in glioma oncogenesis. It also has great value in early diagnosis, and in prognostic diagnosis. This study offers theoretical basis to better understand the molecular mechanism of glioma and its potential therapeutic strategies.

Acknowledgements

The present study was supported by the Natural Science Foundation of Gansu Province (no. 1606RJZA348) and the Science and Technology Project of Lanzhou city (2015–2-67).

References

1 

Godlewski J, Newton HB, Chiocca EA and Lawler SE: MicroRNAs and glioblastoma; the stem cell connection. Cell Death Differ. 17:221–228. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Li Z, Wang H, Eyler CE, Hjelmeland AB and Rich JN: Turning cancer stem cells inside out: An exploration of glioma stem cell signaling pathways. J Biol Chem. 284:16705–16709. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Wen PY and Kesari S: Malignant gliomas in adults. N Engl J Med. 359:492–507. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Gladson CL, Prayson RA and Liu WM: The pathobiology of glioma tumors. Annu Rev Pathol. 5:33–50. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Kartha RV and Subramanian S: Competing endogenous RNAs (ceRNAs): New entrants to the intricacies of gene regulation. Front Genet. 5:82014.eCollection. View Article : Google Scholar : PubMed/NCBI

6 

Li M, Li J, Liu L, Li W, Yang Y and Yuan J: MicroRNA in human glioma. Cancers (Basel). 5:1306–1331. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Zhang Y, Dutta A and Abounader R: The role of microRNAs in glioma initiation and progression. Front Biosci (Landmark Ed). 17:700–712. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Wong JW: MicroRNA-induced silencing of glioma progression. J Neurosci. 30:3868–3869. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Wang Z, Wang B, Shi Y, Xu C, Xiao HL, Ma LN, Xu SL, Yang L, Wang QL, Dang WQ, et al: Oncogenic miR-20a and miR-106a enhance the invasiveness of human glioma stem cells by directly targeting TIMP-2. Oncogene. 34:407–419. 2015. View Article : Google Scholar

10 

Zhao D, Jiang X, Yao C, Zhang L, Liu H, Xia H and Wang Y: Heat shock protein 47 regulated by miR-29a to enhance glioma tumor growth and invasion. J Neurooncol. 118:39–47. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Wan X, Cheng Q, Peng R, Ma Z, Chen Z, Cao Y and Jiang B: ROCK1, a novel target of miR-145, promotes glioma cell invasion. Mol Med Rep. 9:1877–1882. 2014.PubMed/NCBI

12 

Guo M, Jiang Z, Zhang X, Lu D, Ha AD, Sun J, Du W, Wu Z, Hu L, Khadarian K, et al: miR-656 inhibits glioma tumorigenesis through repression of BMPR1A. Carcinogenesis. 35:1698–1706. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Zhang D, Yang G, Chen X, Li C, Wang L, Liu Y, Han D, Liu H, Hou X, Zhang W, et al: mir-300 promotes self-renewal and inhibits the differentiation of glioma stem-like cells. J Mol Neurosci. 53:637–644. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Yang TQ, Lu XJ, Wu TF, Ding DD, Zhao ZH, Chen GL, Xie XS, Li B, Wei YX, Guo LC, et al: MicroRNA-16 inhibits glioma cell growth and invasion through suppression of BCL2 and the nuclear factor-κB1/MMP9 signaling pathway. Cancer Sci. 105:265–271. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Li SZ, Hu YY, Zhao J, Zhao YB, Sun JD, Yang YF, Ji CC, Liu ZB, Cao WD, Qu Y, et al: MicroRNA-34a induces apoptosis in the human glioma cell line, A172, through enhanced ROS production and NOX2 expression. Biochem Biophys Res Commun. 444:6–12. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Zhang Y, Chen X, Lian H, Liu J, Zhou B, Han S, Peng B, Yin J, Liu W and He X: MicroRNA-503 acts as a tumor suppressor in glioblastoma for multiple antitumor effects by targeting IGF-1R. Oncol Rep. 31:1445–1452. 2014.PubMed/NCBI

17 

Chen Z, Li D, Cheng Q, Ma Z, Jiang B, Peng R, Chen R, Cao Y and Wan X: MicroRNA-203 inhibits the proliferation and invasion of U251 glioblastoma cells by directly targeting PLD2. Mol Med Rep. 9:503–508. 2014.PubMed/NCBI

18 

Alrfaei BM, Vemuganti R and Kuo JS: microRNA-100 targets SMRT/NCOR2, reduces proliferation, and improves survival in glioblastoma animal models. PLoS One. 8:e808652013. View Article : Google Scholar : PubMed/NCBI

19 

Guo P, Nie Q, Lan J, Ge J, Qiu Y and Mao Q: C-Myc negatively controls the tumor suppressor PTEN by upregulating miR-26a in glioblastoma multiforme cells. Biochem Biophys Res Commun. 441:186–190. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Jiang J, Yang J, Wang Z, Wu G and Liu F: TFAM is directly regulated by miR-23b in glioma. Oncol Rep. 30:2105–2110. 2013.PubMed/NCBI

21 

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

22 

Zhang N, Wang X, Huo Q, Sun M, Cai C, Liu Z, Hu G and Yang Q: MicroRNA-30a suppresses breast tumor growth and metastasis by targeting metadherin. Oncogene. 33:3119–3128. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Kumarswamy R, Mudduluru G, Ceppi P, Muppala S, Kozlowski M, Niklinski J, Papotti M and Allgayer H: MicroRNA-30a inhibits epithelial-to-mesenchymal transition by targeting Snai1 and is downregulated in non-small cell lung cancer. Int J Cancer. 130:2044–2053. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Zhu H, Wu H, Liu X, Li B, Chen Y, Ren X, Liu CG and Yang JM: Regulation of autophagy by a beclin 1-targeted microRNA, miR-30a, in cancer cells. Autophagy. 5:816–823. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Zhong M, Bian Z and Wu Z: miR-30a suppresses cell migration and invasion through downregulation of PIK3CD in colorectal carcinoma. Cell Physiol Biochem. 31:209–218. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Ouzounova M, Vuong T, Ancey PB, Ferrand M, Durand G, Kelm Le-Calvez F, Croce C, Matar C, Herceg Z and Hernandez-Vargas H: MicroRNA miR-30 family regulates non-attachment growth of breast cancer cells. BMC Genomics. 14:1392013. View Article : Google Scholar : PubMed/NCBI

27 

Zhao S, Ye X, Xiao L, Lian X, Feng Y, Li F and Li L: MiR-26a inhibits prostate cancer progression by repression of Wnt5a. Tumour Biol. 35:9725–9733. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Wu Y, Dong L, Bao S, Wang M, Yun Y and Zhu R: FK228 augmented temozolomide sensitivity in human glioma cells by blocking PI3K/AKT/mTOR signal pathways. Biomed Pharmacother. 84:462–469. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Shivdasani RA: MicroRNAs: Regulators of gene expression and cell differentiation. Blood. 108:3646–3653. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Cohen S: MicroRNAs in animal development. Editorial. Semin Cell Dev Biol. 21:7272010. View Article : Google Scholar : PubMed/NCBI

31 

Zou Z, Ni M, Zhang J, Chen Y, Ma H, Qian S, Tang L, Tang J, Yao H, Zhao C, et al: MiR-30a can inhibit DNA replication by targeting RPA1 thus slowing cancer cell proliferation. Biochem J. 473:2131–2139. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Kumarswamy R, Mudduluru G, Ceppi P, Muppala S, Kozlowski M, Niklinski J, Papotti M and Allgayer H: MicroRNA-30a inhibits epithelial-to-mesenchymal transition by targeting Snai1 and is downregulated in non-small cell lung cancer. Int J Cancer. 130:2044–2053. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Cheng CW1, Wang HW, Chang CW, Chu HW, Chen CY, Yu JC, Chao JI, Liu HF, Ding SL and Shen CY: MicroRNA-30a inhibits cell migration and invasion by downregulating vimentin expression and is a potential prognostic marker in breast cancer. Breast Cancer Res Treat. 134:1081–1093. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Kikuchi A, Yamamoto H, Sato A and Matsumoto S: Wnt5a: Its signalling, functions and implication in diseases. Acta Physiol (Oxf). 204:17–33. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Nishita M, Enomoto M, Yamagata K and Minami Y: Cell/tissue-tropic functions of Wnt5a signaling in normal and cancer cells. Trends Cell Biol. 20:346–354. 2010. View Article : Google Scholar : PubMed/NCBI

36 

McDonald SL and Silver A: The opposing roles of Wnt-5a in cancer. Br J Cancer. 101:209–214. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Katoh M: WNT signaling in stem cell biology and regenerative medicine. Curr Drug Targets. 9:565–570. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 38 Issue 2

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
Zhang Y, Wu Z, Li L and Xie M: miR-30a inhibits glioma progression and stem cell‑like properties by repression of Wnt5a. Oncol Rep 38: 1156-1162, 2017
APA
Zhang, Y., Wu, Z., Li, L., & Xie, M. (2017). miR-30a inhibits glioma progression and stem cell‑like properties by repression of Wnt5a. Oncology Reports, 38, 1156-1162. https://doi.org/10.3892/or.2017.5728
MLA
Zhang, Y., Wu, Z., Li, L., Xie, M."miR-30a inhibits glioma progression and stem cell‑like properties by repression of Wnt5a". Oncology Reports 38.2 (2017): 1156-1162.
Chicago
Zhang, Y., Wu, Z., Li, L., Xie, M."miR-30a inhibits glioma progression and stem cell‑like properties by repression of Wnt5a". Oncology Reports 38, no. 2 (2017): 1156-1162. https://doi.org/10.3892/or.2017.5728