MicroRNA‑301a targets WNT1 to suppress cell proliferation and migration and enhance radiosensitivity in esophageal cancer cells

  • Authors:
    • Huafang Su
    • Youyi Wu
    • Ya Fang
    • Lanxiao Shen
    • Zhenghua Fei
    • Congying Xie
    • Ming Chen
  • View Affiliations

  • Published online on: October 16, 2018     https://doi.org/10.3892/or.2018.6799
  • Pages: 599-607
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Esophageal cancer (EC) is one of the leading causes of death among malignancies. Radiotherapy for esophageal squamous cell carcinoma (ESCC) patients is limited by resistance to ionizing radiation (IR). An increasing body of evidence has demonstrated that aberrant expression of microRNA‑301a (miR‑301a) contributes to cancer progression and sensitivity to radiation. The aim of the present study was to investigate the exact functions and potential mechanisms of miR‑301a in ESCC radioresistance. Initially, the miR‑301a‑transfected radioresistant ESCC cells KYSE‑150R exhibited a decreased proliferation rate, and enhanced radiosensitivity and migration, whereas downregulation of miR‑301a in radiosensitive KYSE‑150 cells produced the opposite results. miR‑301a regulates WNT1 expression at both the mRNA and protein levels. Furthermore, dual‑luciferase reporter assays revealed that WNT1 was a target gene of miR‑301a. In addition, the expression of miR‑301a markedly affected the expression of Wnt/β‑catenin‑related proteins such as β‑catenin and cyclin D1. Finally, overexpression of miR‑301a inhibited epithelial‑mesenchymal transition (EMT) conversion by directly targeting Snail and vimentin in radioresistant‑ESCC cell lines; however, no inhibitory effects were exerted on Twist. Collectively, these results indicated that miR‑301a increased the radiosensitivity and inhibited the migration of radioresistant‑ESCC cells by targeting WNT1, thereby inactivating the Wnt/β‑catenin signaling pathway and EMT reversal. Thus, miR‑301a may be a potential therapeutic target for the treatment of EC radioresistance.

Introduction

Esophageal cancer (EC) is the eighth most commonly diagnosed cancer and the sixth leading cause of cancer-associated mortality worldwide (1). Radiation therapy (RT) is a key part of multimodality EC therapy (2,3). Radiotherapy resistance often leads to subsequent recurrence and metastasis (4,5).

MicroRNAs (miRNAs/miRs) are a family of endogenous non-coding RNAs comprised of 18–25 nucleotides in length that function to regulate the stability of target genes by directly binding to the 3′ untranslated region (3′ UTR) (6,7). An increasing body of evidence has demonstrated that miRNAs can act as tumor promoters or suppressors to regulate various basic cellular functions (810). Furthermore, some studies have revealed that miRNAs directly affect radioresistance by regulating specific pathways (11), including the repair of DNA double strand breaks (12), cell cycle checkpoint activation (13), apoptosis (14) and autophagy (15). For example, Lin28-let7 regulates the radiosensitivity of human cancers through activated KRAS signaling (16).

miR-301a, located on chromosome 17q22-17q23, is an important miRNA that has been studied in a variety of tumor types (17). miR-301a expression is upregulated in several types of cancer including pancreatic (18), prostate cancer (19) and malignant melanoma (20); it also acts as a candidate oncogene by promoting tumor proliferation and invasion. Recently, a previous study demonstrated that miR-301a promoted radioresistance in prostate cancer cells by downregulating N-myc downstream-regulated gene 2 (NDRG2) (21). However, the mechanism underlying the role of miR-301a in EC radioresistance remains unclear.

In our previous study, we used a human miRNA microarray to detect the differential expression of miRNAs, comparing the human radioresistant esophageal squamous cell carcinoma (ESCC) line KYSE-150R and the parental cell line KYSE-150 (22). Based on the predicted genes and pathways of the miRNA target, the expression of miR-301a was significantly downregulated in KYSE-150R cells and it was thought to play an important role in regulating the radiosensitivity of ESCC cells. In the present study, we confirmed the effect of miR-301a on cell proliferation, epithelial-mesenchymal transition (EMT) and radiosensitivity in ESCC cells. It was revealed that miR-301a suppressed cell proliferation and migration, and enhanced radiosensitivity in ESCC cells by directly targeting WNT1.

Materials and methods

Cell culture

The human ESCC cell line KYSE-150 was purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). The radioresistant cell line KYSE-150R was previously established at our department by gradient dose irradiation treatment (22). KYSE-150 and KYSE-150R cells were maintained in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with 100 U/ml of penicillin, 100 mg/ml of streptomycin and 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.), in an incubator at 37°C containing 5% CO2. The cell lines were sub-cultured every 2 to 3 days following digestion at room temperature with 1 ml 0.25% Trypsin-EDTA (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). The viability was reported as the percentage of the number of viable cells to the total number of cells. The average viability over 95% was determined by Trypan Blue staining, at 37°C in an incubator containing 5% CO2.

Cell transfection

Cultured cells were transfected with miR-301a mimic or inhibitor (Ambion; Thermo Fisher Scientific, Inc.) using Lipofectamine 2000™ reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Cells were transfected with 0, 5, 10 and 20 nM miR-301a mimic or inhibitor or negative control (NC) for 48 h, unless indicated otherwise.

Transwell assay

After transfection with miR-301a mimic or inhibitor, 5×104 cells were plated into the top chamber of the insert (8 µm pore size; BD Biosciences, Franklin Lakes, NJ, USA) in medium without serum. The lower chamber was loaded with medium containing 10% FBS. The cells were incubated for 12 h and the cells that had migrated to the bottom of the membrane were fixed with methanol and stained with crystal violet. The number of stained cells were counted at least five random microscopic fields by a light microscope at a magnification of ×200 (Olympus Corp., Tokyo, Japan).

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assay

Cells were seeded into a 96-well plate in triplicate at a concentration of 4×103 cells/well. Cell growth was measured by MTT bromide assay every 24 h from Day 1 to Day 6. Cells were incubated with 5 mg/ml MTT for 4 h, and subsequently solubilized in dimethyl sulfoxide (DMSO) (100 µl/well). The absorbance was measured at 570 nm using an ELISA reader.

Clonogenic assay

Cells in the exponential phase of growth were plated in triplicate onto 6-well plates with ~1×103 cells/well. These logarithmic growth cells were irradiated with doses of 0, 2, 4, 6, 8, or 10 Gy using X-ray from a linear accelerator (Varian 2300C/D; Varian Medical Systems, Salt Lake, UT, USA) with an average dose rate of 100 cGy/min. Immediately following irradiation, the cells were incubated for 10 days at 37°C in 5% CO2 to allow for colony formation and then fixed with pure ethanol. Visible colonies consisting of at least 50 cells were stained with 0.5% crystal violet (Sigma-Aldrich; Merck KGaA) and counted. The surviving fraction was then estimated.

Dual-luciferase reporter assay

The wild-type (wt) or mutated (mut) 3′ UTR of WNT1, containing three binding sites of miR-301a, were cloned into the luciferase vector pMIR reporter. Cells were transfected with the WNT1 luciferase reporter vector, Renilla luciferase vector (Promega Corp., Madison, WI, USA) and the miRNA mimic or inhibitor using Lipofectamine 2000™ reagent (Invitrogen; Thermo Fisher Scientific, Inc.). After 48 h, the cells were harvested, lysed and luciferase activity was assessed using the Dual-Luciferase Reporter Assay system (Promega Corp.) according to the manufacturer's protocol. Renilla luciferase was used for normalization.

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

Total RNA was extracted using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Total RNA (1 µg) was reverse transcribed to cDNA using the SuperScript II reverse transcription kit (Thermo Fisher Scientific, Inc.). RT-qPCR was performed using SYBR-Green Master Mix (Applied Biosystems; Thermo Fisher Scientific, Inc.) and the ABI PRISM 7900 Sequence Detection System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The RT-qPCR thermocycling conditions were as follows: 95°C for 10 min, followed by 40 cycles at 95°C for 15 sec and 60°C for 1 min. β-actin was used as an internal control and the fold-changes of target genes were calculated using the 2−ΔΔCq method (23). Primer sequences of Wnt pathway- and EMT-associated genes are presented in Table I.

Table I.

Primer sequences of Wnt pathway- and EMT-associated genes.

Table I.

Primer sequences of Wnt pathway- and EMT-associated genes.

Target geneForward primer (5′-3′)Reverse primer (5′-3′)
WNT1 TAAGCAGGTTCGTGGAGGAG GGTTTCTGCTACGCTGCTG
E-cadherin GACCGGTGCAATCTTCAAA TTGACGCCGAGAGCTACAC
Twist TCCATTTTCTCCTTCTCTGGAA CCTTCTCGGTCTGGAGGAT
Vimentin GCAAAGATTCCACTTTGCGT GAAATTGCAGGAGGAGATGC
Snail TCTGAGTGGGTCTGGAGGTG CTCTAGGCCCTGGCTGCTAC
TCF4 CATAGGGAGTCCCATCTCCA GGACCAACTTCTTTGGCAAG
Cyclin D1 GGCGGATTGGAAATGAACTT TCCTCTCCAAAATGCCAGAG
β-actin AAAGACCTGTACGCCAACAC GTCATACTCCTGCTTGCTGAT
Western blot analysis

Proteins were extracted from cultured cells using RIPA buffer (Thermo Fisher Scientific, Inc). Protein concentration was determined using the BCA protein assay kit (Thermo Fisher Scientific, Inc.). Lysates containing 100 µg of protein were mixed with loading buffer containing 5% β-mercaptoethanol. Extracted proteins were subjected to 10% SDS-PAGE and transferred to PVDF membranes (0.45 µm). The membranes were then incubated with 5% non-fat dry milk in phosphate-buffered saline (PBS) for 1 h at room temperature to block non-specific binding sites. Then, the membranes were incubated with primary antibodies overnight at 4°C, followed by a secondary antibody (1:20,000; anti-rabbit or anti-mouse IgG, HRP-linked antibody, cat. nos. 7071 and 7072, respectively; Cell Signaling Technology, Inc.) incubation for 2 h at room temperature. The protein bands were visualized with the SuperSignal West Pico Chemiluminescent kit (Pierce; Thermo Fisher Scientific, Inc.). The following primary antibodies were used: Cyclin D1 (1:1,000; cat. no. 2978), E-cadherin (1:1,000; cat. no. 3195), β-catenin (1:1,000; cat. no. 8480) and Snail (1:1,000; cat. no. 3879; all from CST Biological Reagents Co., Ltd., Shanghai, China), WNT1 (1:1,000; cat. no. ab15251; Abcam, Cambridge, MA, USA) and vimentin (1:1,000; cat. no. 5741; CST Biological Reagents Co., Ltd.) and Tubulin (1:4,000; cat. no. T5168; Sigma-Aldrich; Merck KGaA).

Immunofluorescence staining

Cells were fixed with 4% formaldehyde, permeabilized with 0.5% Triton, and then incubated with an E-cadherin (1:500) or vimentin (1:500) antibody overnight. After washing with PBS, the cells were incubated with Alexa Fluor 488-labeled secondary antibody (1,000-fold dilution; cat. no. 34201; Invitrogen; Thermo Fisher Scientific, Inc.) for 45 min. Cells were counterstained with DAPI to label the nuclei. Images were captured with a Nikon fluorescent microscope.

Statistical analysis

Data were expressed as the mean ± standard deviation from at least three independent experiments. Student's t-test was used to compare the differences between two groups. All statistical analyses were performed using SPSS 15.0 software (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

miR-301a inhibits cell proliferation and migration, and increases radioresistant cell sensitivity to irradiation in ESCC

The present study initially transfected KYSE-150R cells with the miR-301a mimic in order to upregulate miR-301a in vitro (P<0.01; Fig. 1A). The overexpression of miR-301a in KYSE-150R cells decreased cell viability when compared with cells transfected with NC mimic cells (P<0.05; Fig. 1B). The results of the colony formation assays revealed that overexpressing miR-301a significantly enhanced the effects of radiotherapy in KYSE-150R cells (P<0.05; Fig. 1C). Upregulation of miR-301a in KYSE-150R cells led to a 30–40% reduction in the levels of migration when compared with the NC-mimic cells (P<0.01; Fig. 1D).

Next, the present study suppressed the expression of miR-301a in KYSE-150 cells via transfection with a miR-301a inhibitor (P<0.05; Fig. 2A). The miR-301a inhibitor groups exhibited enhanced cell proliferation when compared with the NC inhibitor group (P<0.05; Fig. 2B). The KYSE-150 cells with reduced miR-301a levels had a significantly greater survival fraction when compared with the NC cells (P<0.05; Fig. 2C), indicating that these cells had decreased radiosensitivity. Anti-miR301a cells displayed significantly higher migration potential, compared to the NC group (P<0.01; Fig. 2D).

miR-301a directly targets the coding region of WNT1

Next the present study evaluated the potential targets of miR-301a. Bioinformatics analysis was performed to identify potential candidate targets of miR-301a using publicly available algorithms. As revealed in Fig. 3A, the 3′ UTR WNT1 contained the target sequence of miR-301a. Subsequently, RT-qPCR and western blotting were employed to determine the ability of miR-301a to regulate the expression of WNT1. Overexpression of miR-301a markedly downregulated the mRNA and protein levels of WNT1 in KYSE-150R cells. Conversely, inhibition of miR-301a markedly increased the expression of WNT1 at the mRNA and protein levels in the KYSE-150 group (Fig. 3B and C).

To further explore whether miR-301a could interact with the 3′ UTR of WNT1, the present study performed luciferase reporter assays. As indicated in Fig. 3D, the miR-301a mimic significantly inhibited luciferase activity in cells expressing pMIR-WNT1-wt 3′ UTR. However, co-transfection with the pMIR-WNT1-mut plasmid and the miR-301a mimic did not significantly alter the luciferase activity when compared with the NC group (P>0.05). Collectively, these results demonstrated that WNT1 is directly targeted by miR-301a.

Comparisons between the expression of Wnt/β-catenin signaling pathway-related genes and proteins among the different groups

RT-qPCR and western blot analyses were conducted to investigate whether miR-301a expression could markedly affect the expression of Wnt/β-catenin-related proteins. As revealed in Fig. 4A, miR-301a mimic-transfected cells (at the greatest concentration of 20 nM) had significantly lower mRNA expression of transcription factor 4 (TCF4) and cyclin D1 when compared to the KYSE-150R control groups. In miR-301a inhibitor-transfected cells, TCF4 and cyclin D1 were significantly increased. Furthermore, the protein expression of β-catenin and cyclin D1 was reduced in the miR-301a mimic-transfected group. However, the miR-301a inhibitor-transfected group exhibited the opposite results (P<0.05; Fig. 4B).

miR-301a affects EMT-related protein expression

Our previous study demonstrated that EMT mediates radioresistance in human ESCC cells (24). Therefore, in the present study classic EMT biomarkers and transcription factors were detected in the different cell groups. The results of the RT-qPCR assay and western blotting revealed that the gene and protein expression levels of E-cadherin in KYSE-150 cells transfected with miR-301a inhibitors were decreased than those observed in the NC groups, while E-cadherin expression in the miR-301a-mimic groups was significantly increased when compared with the NC groups (Fig. 5A and B). Furthermore, expression of vimentin and Snail in the miR-301a-inhibitor group was upregulated, whereas the expression of vimentin and Snail was downregulated in the miR-301a-mimic group. However, the regulation of miR-301a did not significantly affect Twist expression (P≥0.05; Fig. 5A and B).

Immunofluorescence analysis was conducted to confirm the localization of vimentin and E-cadherin in the miR-301a overexpression and knockout groups. Intense staining in the cell membrane indicated that E-cadherin was shuttled to the cell membrane in miR-301a-overexpressing cells, while less staining in knockout miR-301a cells indicated that E-cadherin was downregulated (Fig. 5C). Conversely the expression of vimentin in the cell membrane of miR-301a mimic-transfected cells was reduced. These results indicated that miR-301a may be an upstream regulator of E-cadherin and vimentin expression, and an inducer of EMT phenotypes.

Discussion

Acquired radioresistance is considered to be an important cause of treatment failure in EC patients. Our previous study revealed that miR-301a was a candidate in the aberrant profile of radiosensitive miRNAs and was associated with ESCC radiosensitivity (22). However, there is limited research on its function. In the present study, miR-301a was revealed to regulate the radiosensitivity of ESCC by targeting WNT1. The results indicated that miR-301a may be a new type of radiosensitivity-related miRNA for future EC radioresistant therapeutic strategies.

miR-301a has been identified as a candidate oncogene in different types of cancers. In colorectal cancer, ectopic miR-301a expression was observed to promote migration and invasion by downregulating transforming growth factor β receptor (17). Nam et al (25) demonstrated that miR-301a played an oncogenic role in prostate cancer by directly targeting the p63 tumor suppressor. Kawano et al (26) demonstrated that restoration of miR-301a downregulation decreased ESC proliferation and tumorigenesis, and that the oncogenic activity may involve the negative regulation of the target gene, phosphatase and tensin homolog. In the present study, upregulation of miR-301a in KYSE-150R cells was revealed to inhibit cell proliferation and migration, and increase cell sensitivity to irradiation. Furthermore, inhibition of miR-301a in KYSE-150 cells reduced cell sensitivity to irradiation. However, miR-301a function in the present study was markedly different to that reported in previous studies. This may be due to the fact that the different functions of miRNAs depend on the particular type of cancer cell line used because of its particular genetic background.

miRNAs exert their biological functions by binding with the 3′ UTR of their target genes. Wang et al (21) observed that higher expression levels of miR-301a and miR-301b resulted in elevated autophagy and increased radioresistance in LNCaP cells by targeting NDRG2. The present study confirmed that WNT1 was a target of miR-301a based on the results of the luciferase reporter assays. In agreement with this, the present study also revealed that miR-301a overexpression significantly inhibited WNT1 expression at both the protein and mRNA levels, indicating that miR-301a suppressed ESCC cell activities by targeting WNT1.

Numerous previous studies have focused on WNT1, a major member of Wnt/β-catenin signaling, and have often reported that it is altered in EC. The Wnt/β-catenin signaling pathway plays a crucial role in EC biological processes, including cell proliferation, apoptosis, and motility. Recently, previous research has reported that aberrant activation of canonical Wnt/β-catenin signaling after long-term exposure to fractionated irradiation was associated with the development of radioresistance in many types of human cancers. In our pervious study, significantly upregulated expression levels of WNT1, catenin β1 and cyclin D1 were observed in the radioresistant ESCC KYSE-150R cell line, indicating that the Wnt/β-catenin signaling pathway was activated (24). As crucial biological regulators, miRNAs inhibit targets of signaling pathways and can promote or inhibit cancer progression in a context- or target-dependent manner. (27). miR-185-3p regulated nasopharyngeal carcinoma radioresistance by targeting WNT2B (28). The present study revealed that β-catenin and cyclin D1 may be key molecules in the Wnt/β-catenin signaling pathway, as their expression was altered when miR-301a was upregulated. These studies provide the basis for the future exploration of the potential mechanisms underlying miR-301a functions and how WNT1 is involved in EC radiation resistance.

Notably, the present study demonstrated that miR-301a not only enhanced radiosensitivity but also affected EMT by changing vimentin and E-cadherin expression. EMT is a vitally important biological process during which epithelial cells lose their polarity and change to a mesenchymal phenotype (29). Numerous studies have demonstrated that EMT plays an important role in cancer malignant behaviors, including radiation resistance (30), drug resistance (31) and cancer stem cells (32). The epithelial marker E-cadherin and the mesenchymal marker vimentin are often used as markers of EMT during metastatic progression. Our previous study reported that the ESCC cell line KYSE-150R presented with typical morphological changes of the EMT phenotype with significantly decreased E-cadherin and increased vimentin expression. The results of the present study revealed that miR-301a and WNT1 affected the expression of EMT-related biomolecules, indicating that miR-301a and WNT1 may induce EC radioresistance through EMT.

However, there are some limitations in our study. For example, we only used one radioresistant cell line to conduct the study. In vivo models will be set up to define the roles of miR-301a in EC radioresistance in further research. In conclusion, upregulation of miR-301a inactivated the Wnt/β-catenin signaling pathway by targeting WNT1, leading to increased radiosensitivity and reduced migration of cancer cells. In addition, targeting this novel miR-301a/Wnt/β-catenin axis may be a promising strategy for the future treatment of EC radioresistance.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of Zhejiang Province (grant nos. LQ15H160013, LY17H160051 and LY15H280013) and the Natural Science Foundation of China (grant no. 81602658).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

MC designed the experiments. HS, YW, LS, and ZF performed the biological experiments. MC, YW, LS and HS provided administrative support and funded the experiments. HS, YF, ZF and CX analyzed the data. HS, YF, CX and MC drafted the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Napier KJ, Scheerer M and Misra S: Esophageal cancer: A Review of epidemiology, pathogenesis, staging workup and treatment modalities. World J Gastrointest Oncol. 6:112–120. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Fokas E, Weiss C and Rödel C: The role of radiotherapy in the multimodal management of esophageal cancer. Dig Dis. 31:30–37. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Sjoquist KM, Burmeister BH, Smithers BM, Zalcberg JR, Simes RJ, Barbour A and Gebski V: Australasian Gastro-Intestinal Trials Group: Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: An updated meta-analysis. Lancet Oncol. 2:681–692. 2011. View Article : Google Scholar

4 

Barker HE, Paget JT, Khan AA and Harrington KJ: The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat Rev Cancer. 15:409–425. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Maier P, Hartmann L, Wenz F and Herskind C: Cellular pathways in response to ionizing radiation and their targetability for tumor radiosensitization. Int J Mol Sci. 17(pii): E1022016. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Garzon R, Marcucci G and Croce CM: Targeting microRNAs in cancer: Rationale, strategies and challenges. Nat Rev Drug Discov. 9:775–778. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Hummel R, Hussey DJ and Haier J: MicroRNAs: Predictors and modifiers of chemo- and radiotherapy in different tumour types. Eur J Cancer. 46:298–311. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Farazi TA, Spitzer JI, Morozov P and Tuschl T: miRNAs in human cancer. J Pathol. 223:102–115. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Zhao L, Lu X and Cao Y: MicroRNA and signal transduction pathways in tumor radiation response. Cell Signal. 25:1625–1634. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Hu B, Wang X, Hu S, Ying X, Wang P, Zhang X, Wang J, Wang H and Wang Y: miR-21-mediated radioresistance occurs via promoting repair of DNA double strand breaks. J Biol Chem. 292:3531–3540. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Bhattacharya A, Schmitz U, Wolkenhauer O, Schönherr M, Raatz Y and Kunz M: Regulation of cell cycle checkpoint kinase WEE1 by miR-195 in malignant melanoma. Oncogene. 32:3175–3183. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Wang S, Pan Y, Zhang R, Xu T, Wu W, Zhang R, Wang C, Huang H, Calin CA, Yang H and Claret FX: Hsa-miR-24-3p increases nasopharyngeal carcinoma radiosensitivity by targeting both the 3′UTR and 5′UTR of Jab1/CSN5. Oncogene. 35:6096–6108. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Wang P, Zhang J, Zhang L, Zhu Z, Fan J, Chen L, Zhuang L, Luo J, Chen H, Liu L, et al: MicroRNA 23b regulates autophagy associated with radioresistance of pancreatic cancer cells. Gastroenterology. 145:1133–1143. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Oh JS, Kim JJ, Byun JY and Kim IA: Lin28-let7 modulates radiosensitivity of human cancer cells with activation of K-Ras. Int J Radiat Oncol Biol Phys. 76:5–8. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Zhang W, Zhang T, Jin R, Zhao H, Hu J, Feng B, Zang L, Zheng M and Wang M: MicroRNA-301a promotes migration and invasion by targeting TGFBR2 in human colorectal cancer. J Exp Clin Cancer Res. 33:1132014. View Article : Google Scholar : PubMed/NCBI

18 

Xia X, Zhang K, Luo G, Cen G, Cao J, Huang K and Qiu Z: Downregulation of miR-301a-3p sensitizes pancreatic cancer cells to gemcitabine treatment via PTEN. Am J Transl Res. 9:1886–1895. 2017.PubMed/NCBI

19 

Damodaran C, Das TP, Papu John AM, Suman S, Kolluru V, Morris TJ, Faber EN, Rai SN, Messer JC, Alatassi H, et al: miR-301a expression: A prognostic marker for prostate cancer. Urol Oncol. 34:e13–e20. 2016. View Article : Google Scholar

20 

Cui L, Li Y, Lv X, Li J, Wang X, Lei Z and Li X: Expression of microRNA-301a and its functional roles in malignant melanoma. Cell Physiol Biochem. 40:230–244. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Wang W, Liu M, Guan Y and Wu Q: Hypoxia-responsive miR-301a and miR-301b promote radioresistance of prostate cancer cells via downregulating NDRG2. Med Sci Monit. 22:2126–2132. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Su H, Jin X, Zhang X, Xue S, Deng X, Shen L, Fang Y and Xie C: Identification of microRNAs involved in the radioresistance of esophageal cancer cells. Cell Biol Int. 38:318–325. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Su H, Jin X, Zhang X, Zhao L, Lin B, Li L, Fei Z, Shen L, Fang Y, Pan H, et al: FH535 increases the radiosensitivity and reverses epithelial-to-mesenchymal transition of radioresistant esophageal cancer cell line KYSE-150R. J Transl Med. 13:1042015. View Article : Google Scholar : PubMed/NCBI

25 

Nam RK, Benatar T, Wallis CJ, Amemiya Y, Yang W, Garbens A, Naeim M, Sherman C, Sugar L and Seth A: MiR-301a regulates E-cadherin expression and is predictive of prostate cancer recurrence. Prostate. 76:869–884. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Kawano M, Tanaka K, Itonaga I, Iwasaki T and Tsumura H: MicroRNA-301a promotes cell proliferation via PTEN targeting in Ewing's sarcoma cells. Int J Oncol. 48:1531–1540. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Song JL, Nigam P, Tektas SS and Selva E: microRNA regulation of Wnt signaling pathways in development and disease. Cell Signal. 27:1380–1391. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Li G, Wang Y, Liu Y, Su Z, Liu C, Ren S, Deng T, Huang D, Tian Y and Qiu Y: miR-185-3p regulates nasopharyngeal carcinoma radioresistance by targeting WNT2B in vitro. Cancer Sci. 105:1560–1568. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, et al: The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 133:704–715. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Marie-Egyptienne DT, Lohse I and Hill RP: Cancer stem cells, the epithelial to mesenchymal transition (EMT) and radioresistance: Potential role of hypoxia. Cancer Lett. 341:63–72. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Shibue T and Weinberg RA: E MTCS Cs, and drug resistance: The mechanistic link and clinical implications. Nat Rev Clin Oncol. 14:611–629. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Martin-Belmonte F and Perez-Moreno M: Epithelial cell polarity, stem cells and cancer. Nat Rev Cancer. 12:23–38. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 41 Issue 1

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
Su H, Wu Y, Fang Y, Shen L, Fei Z, Xie C and Chen M: MicroRNA‑301a targets WNT1 to suppress cell proliferation and migration and enhance radiosensitivity in esophageal cancer cells. Oncol Rep 41: 599-607, 2019
APA
Su, H., Wu, Y., Fang, Y., Shen, L., Fei, Z., Xie, C., & Chen, M. (2019). MicroRNA‑301a targets WNT1 to suppress cell proliferation and migration and enhance radiosensitivity in esophageal cancer cells. Oncology Reports, 41, 599-607. https://doi.org/10.3892/or.2018.6799
MLA
Su, H., Wu, Y., Fang, Y., Shen, L., Fei, Z., Xie, C., Chen, M."MicroRNA‑301a targets WNT1 to suppress cell proliferation and migration and enhance radiosensitivity in esophageal cancer cells". Oncology Reports 41.1 (2019): 599-607.
Chicago
Su, H., Wu, Y., Fang, Y., Shen, L., Fei, Z., Xie, C., Chen, M."MicroRNA‑301a targets WNT1 to suppress cell proliferation and migration and enhance radiosensitivity in esophageal cancer cells". Oncology Reports 41, no. 1 (2019): 599-607. https://doi.org/10.3892/or.2018.6799