Demethylation drug 5-Aza-2'-deoxycytidine-induced upregulation of miR-200c inhibits the migration, invasion and epithelial-mesenchymal transition of clear cell renal cell carcinoma in vitro

Retraction in: /10.3892/ol.2021.12923

  • Authors:
    • Juan Jiang
    • Bo Yi
    • Siqing Ding
    • Jian Sun
    • Wei Cao
    • Mengzi Liu
  • View Affiliations

  • Published online on: March 22, 2016     https://doi.org/10.3892/ol.2016.4364
  • Pages: 3167-3172
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The microRNA (miR)-200 family has been found to be involved in the process of mesenchymal-epithelial transition during renal development. Deregulation of miR-200c has been suggested to be involved in clear cell renal cell carcinoma (ccRCC). However, the precise role of miR‑200c in the regulation of ccRCC metastasis has not been previously reported. In the present study, it was observed that miR‑200c was frequently downregulated in ccRCC tissue compared with matched adjacent normal tissue. The expression of miR‑200c was additionally reduced in ccRCC cell lines when compared with levels in normal renal cells. The DNA demethylation drug 5‑Aza‑2'‑deoxycytidine (Aza) was used to treat several ccRCC cell lines, and it was observed that the expression of miR‑200c was significantly increased following Aza treatment. Furthermore, treatment with Aza markedly inhibited ccRCC cell invasion and migration, while treatment with miR‑200c inhibitor significantly enhanced invasion and migration of ccRCC cells. In addition, Aza treatment significantly promoted expression of E‑cadherin and inhibited the expression of N‑cadherin, while the inhibition of miR‑200c downregulated E‑cadherin and upregulated the expression of N‑cadherin, suggesting that miR‑200c has a suppressive role in epithelial-mesenchymal transition (EMT) of ccRCC cells. In conclusion, it was suggested that demethylation drug Aza-induced upregulation of miR‑200c may inhibit migration, invasion and EMT in ccRCC cells.

Introduction

Clear cell renal cell carcinoma (ccRCC) is the most commonly observed carcinoma of the renal parenchyma, accounting for ~70% of renal cancer cases (1). Among urinary system tumors, the incidence of ccRCC is the third highest. Furthermore, ccRCC accounts for ~3% of adult cancer cases, and its mortality rate is on the increase (2,3). Although treatment is typically administered in the form of surgery combined with chemotherapy and radiotherapy, the median survival rate of ccRCC remains poor (4,5). Therefore, the development of effective therapeutic targets for ccRCC is urgently required.

MicroRNAs (miRs) are a type of small non-coding RNA (19–25 nucleotides) that possess prominent roles in the regulation of genes (6). MiRs typically inhibit gene expression by directly binding the 3′-untranslated region of their target messenger (m)RNAs, causing repression of translation or mRNA degradation (7). Deregulation of miRs has been revealed to have a significant role in tumorigenesis (8). In addition, the expression of miRs is regulated by the DNA methylation status in the CpG island of the promoter region, similar to protein-encoding genes, which are involved in the development and progression of human cancer (9). Therefore, epigenetic drugs, including the demethylation drug 5-Aza-2′-deoxycytidine (Aza), may be utilized for the treatment of human cancer, via mediation of the expression levels of tumor-associated miRs.

Deregulation of miR-200c has been identified to be associated with various types of human cancer, including ovarian, breast, prostate, gastric and bladder cancer (1014). miR-200c has been identified as being frequently downregulated in ccRCC (15). Furthermore, loss of miR-200c expression was observed to cause gain of function of oncogenes in ccRCC (16). Accordingly, the present study examined whether miR-200c acts as a tumor suppressor in ccRCC. However, the role of miR-200c in the regulation of ccRCC metastasis, as well as the epigenetic regulatory mechanism, has not previously been reported to the best of our knowledge.

The present study primarily aimed to evaluate the effect of epigenetic drug treatment on miR-200c expression in ccRCCs. The role of miR-200c in the regulation of migration, invasion and epithelial-mesenchymal transition (EMT) in ccRCC cells was additionally studied.

Materials and methods

Reagents and materials

Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), TRIzol® reagent, Bicinchoninic Acid (BCA) Protein Assay kit and Lipofectamine™ 2000 were obtained from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Demethylation drug Aza was obtained from Sigma-Aldrich (St. Louis, MO, USA). The miRNeasy mini kit was obtained from Qiagen, Inc. (Valencia, CA, USA). Mouse anti-E-cadherin, N-cadherin and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) monoclonal antibodies, and rabbit anti-mouse secondary antibody were obtained from Abcam (Cambridge, MA, USA). The Cell Invasion Assay kit was purchased from Merck Millipore (Darmstadt, Germany).

Cell culture

The SN12C, A704, 786-O, ACHN and TK10 human ccRCC cell lines, and HEK293 normal renal cells were obtained from the Cell Bank of Central South University (Changsha, China). The cells were cultured in DMEM with 10% FBS in a humidified atmosphere containing 5% CO2 at 37°C.

Reverse transcription-polymerase chain reaction assay

According to the manufacturer's protocol, total RNA was extracted from tissues and cells using TRIzol® reagent (Thermo Fisher Scientific, Inc.) The SYBR® GreenER™ miRNA qRT-PCR Kit (Thermo Fisher Scientific, Inc.) and Prism 7500 SDS (Applied Biosystems; Thermo Fisher Scientific, Inc.) were used to convert RNA into cDNA, according to the manufacturer's protocol. A negative control (no RNA) and a reverse transcription-negative control were used. Subsequently, the miRNA levels were evaluated using the miRNeasy mini kit (Qiagen, Inc.). The U6 small nuclear RNA was used for normalization. Specific primer sets for miR-200c (cat. no. HmiRQP0227) and U6 (cat. no. HmiRQP900) were obtained from GeneCopoeia, Inc., Rockville, MD, USA. The PCR cycling conditions were as follows: 95°C for 5 min, 40 cycles of denaturation at 95°C for 15 sec and an annealing/elongation step at 60°C for 30 sec. The relative expression of miRNA was quantified using the 2−ΔΔCq method (17).

Western blot analysis

Western blot analysis was applied to examine the protein expression levels in each group. The cells were lysed in cold radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology, Shanghai, China). The BCA Protein Assay kit was utilized to determine the protein concentration in accordance with the manufacturer's protocol. Subsequently, the protein was separated using 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and transferred to a polyvinylidene difluoride (PVDF) membrane. The PVDF membrane was blocked by 5% non-fat dried milk in phosphate-buffered saline (PBS) for 4 h. Subsequently, the PVDF membrane was incubated with specific primary antibodies [mouse anti-human E-cadherin (cat. no. ab1416; 1:200), N-cadherin (cat. no. ab19348; 1:100) and GAPDH (cat. no. ab8245; 1:1:200) monoclonal antibodies] for 3 h. After washing with PBS three times, each for 5 min, the PVDF membrane was incubated with the appropriate secondary antibody [horseradish peroxidase-conjugated rabbit anti-mouse IgG polyclonal secondary antibody (cat. no. ab6728; 1:10,000)]. After washing with PBS three times, each for 5 min, an enhanced chemiluminescence Western Blotting kit (Thermo Fisher Scientific, Inc.) was utilized to detect the immune complexes on the PVDF membrane.

Epigenetic drug treatment of cells

Human ccRCC A704 and TK10 cells were treated with Aza (0.1, 1 and 10 nM) for 72 h.

Wound scratch assay

A wound scratch assay was performed to determine the cell migratory capacity of each group. Cells were cultured to 100% confluence, and a wound of ~1 mm width was made using a plastic scriber. Subsequently, the cells were washed with PBS and cultured at 37°C with 5% CO2 for 36 h. Following culturing, the cells in each group were fixed and observed under a microscope (CX41; Olympus Corporation, Tokyo, Japan).

Cell invasion assay

The cells treated with 0.1, 1 and 10 nM Aza for 72 h were starved in serum-free medium for 24 h, and subsequently re-suspended in serum-free medium. The cells were added to the upper Transwell chamber, while the lower chamber was filled with base medium containing 10% FBS. Following incubation for 24 h, cells that had migrated to the lower chamber were stained with crystal violet (Beyotime Institute of Biotechnology) for 20 min, and subsequently washed and air-dried. Invasive cells were observed under a microscope.

Statistical analysis

Data were expressed as the mean ± standard deviation of three independent experiments, and analyzed using SPSS version 17.0 (SPSS, Inc., Chicago, IL, USA). The differences between groups were determined using one-way analysis of variance. P<0.05 was considered to indicated a statistically significant difference.

Results

miR-200c is frequently downregulated in ccRCC tissues and cell lines

To identify the role of miR-200c in ccRCC, the expression levels of miR-200c in six ccRCC tissues and their matched adjacent normal tissues were initially determined. As shown in Fig. 1A, miR-200c was frequently downregulated in ccRCC tissue compared with matched adjacent normal tissue, suggesting that deregulation of miR-200c may play a role in the development and progression of ccRCC. Subsequently, the detailed role of miR-200c in ccRCC was studied in vitro. The expression levels of miR-200c were examined in the SN12C, A704, 786-O, ACHN and TK10 ccRCC cell lines and HEK293 normal renal cells. As shown in Fig. 1B, the expression levels of miR-200c were significantly reduced in ccRCC cell lines compared with normal renal HEK293 cells. As ccRCC A704 and TK10 cells demonstrated the most significant decrease in miR-200c expression, the two cell lines were utilized in the subsequent experiments.

Treatment with demethylation drug promotes the expression of miR-200c in ccRCC cells

The methylation status in the CpG island of the gene promoter is associated with gene transcription. In general, hypermethylated status in the CpG island of the gene promoter inhibits gene transcription (18). Therefore, demethylation treatment frequently promotes gene transcription. However, whether the expression of miR-200c is mediated by methylation has not been previously studied to the best of our knowledge. In the present study, A704 and TK10 cells ccRCC were treated with demethylation drug Aza. Following treatment with Aza (0.1, 1 and 10 nM) for 72 h, the expression levels of miR-200c were examined in each group. As shown in Fig. 2, the expression levels of miR-200c were significantly increased following treatment with Aza. Furthermore, the higher the concentration of Aza used, the higher the observed expression levels of miR-200c in the two ccRCC cell lines (Fig. 2). According to the above data, 10 nM Aza was used in the subsequent experiments.

miR-200c has an inhibitory role in the regulation of ccRCC cell migration

Cell migration has a significant role in tumor metastasis (19). Therefore, the present study investigated the effect of Aza treatment on ccRCC cell migration. As shown in Fig. 3A, treatment with Aza significantly inhibited the migration of ccRCC cells. Subsequently, it was investigated whether Aza-induced inhibition of cell migration was due to upregulation of miR-200c. Aza-treated ccRCC cells were transfected with miR-200c inhibitor, which is able to reverse upregulation of miR-200c induced by treatment with Aza (Fig. 3B). It was subsequently identified that migration was significantly increased in the Aza + miR-200c inhibitor group compared with the Aza alone group (Fig. 3C), suggesting that the inhibitory effect of Aza treatment on ccRCC cell migration may be at least partly attributed to Aza-induced upregulation of miR-200c. Therefore, miR-200c possesses a suppressive role in the regulation of ccRCC cell migration.

Upregulation of miR-200c induced by demethylation drug inhibits ccRCC cell invasion

As cell invasion additionally participates in tumor metastasis, the present study investigated the effects of Aza and miR-200c on ccRCC cell invasion. Similar to the migration data, treatment with Aza significantly inhibited the invasiveness of ccRCC A704 and TK10 cells, compared with the control group. However, the inhibitory effect of Aza treatment on cell invasion was significantly attenuated by inhibition of miR-200c (Fig. 4). Based on these findings, it was suggested that miR-200c possesses an inhibitory role in the regulation of ccRCC cell invasion.

miR-200c has a suppressive effect on EMT in ccRCC cells

Furthermore, the effects of Aza treatment and miR-200c on EMT were investigated in ccRCC cells. The protein expression levels of E-cadherin and N-cadherin, which are associated with EMT, were determined. As shown in Fig. 5, Aza treatment significantly inhibited the expression of N-cadherin and promoted the expression of E-cadherin, while the downregulation of miR-200c markedly promoted the expression of N-cadherin and suppressed the expression of E-cadherin, suggesting that miR-200c had a suppressive role in EMT of ccRCC cells. These findings were consistent with the results of the aforementioned cell migration and invasion assays.

Discussion

In the present study, it was demonstrated that the expression levels of miR-200c were significantly reduced in ccRCC tissues and cell lines, when compared with the levels in adjacent normal tissues and the normal renal cell line. In addition, it was identified that treatment with the demethylation drug Aza markedly upregulated the expression of miR-200c in ccRCC cells. Furthermore, the upregulation of miR-200c induced by Aza treatment markedly inhibited the migration, invasion and EMT in ccRCC cells, while the knockdown of miR-200c significantly promoted the migration, invasion and EMT in ccRCC cells, suggesting that miR-200c potentially acted as a tumor suppressor in the regulation of ccRCC metastasis.

Nakada et al (15) investigated the expression profiles of miRs in renal cell carcinoma, including ccRCC and chromophobe renal cell carcinoma, and identified that miR-200c was significantly downregulated in ccRCC. In addition, Senanayake et al (20) identified that miR-200c was downregulated and its target Activin A Receptor, Type IIB was markedly expressed in renal childhood neoplasms (20). In the present study, it was additionally demonstrated that miR-200c was frequently downregulated in ccRCC tissues and cell lines. The abovementioned findings suggested that the deregulation of miR-200c may have a role in the development and progression of ccRCC. However, the detailed role of miR-200c in the regulation of ccRCC metastasis has not been previously studied to the best of our knowledge.

DNA methylation in the CpG island of the gene promoter is the most frequent epigenetic modification in eukaryotic genomes, and hypermethylation typically inhibits gene transcription (21). However, the epigenetic regulatory mechanism underlying miR-200c expression has not been previously studied in human cancer to the best of our knowledge. Aza is a DNA methyltransferase inhibitor, which may cause DNA demethylation (22). In the present study, it was observed that treatment with Aza significantly promoted the expression of miR-200c, in a dose-dependent manner. Accordingly, the gene transcription of miR-200c in ccRCC cells was mediated by the DNA methylation status in the CpG island of the promoter region. Furthermore, as the expression of miR-200c was frequently reduced in ccRCC tissues and cell lines, the results of the present study suggested that hypermethylation of the miR-200c promoter may be a significant cause of downregulation of miR-200c in ccRCC.

Subsequently, the present study identified a significant decrease in the migration and invasion in ccRCC cells treated with Aza. However, knockdown of miR-200c enhanced ccRCC cell migration and invasion. As Aza treatment markedly upregulated the expression levels of miR-200c, the results of the present study suggest that miR-200c may have a suppressive effect on ccRCC cell migration and invasion, and the inhibitory effect of Aza treatment on ccRCC cell migration and invasion may be partly due to the direct upregulation of miR-200c expression levels. A suppressive role of miR-200c in cell migration and invasion has additionally been identified in other types of human cancer (23,24). Liu et al (13) identified that miR-200c inhibited invasion, migration and proliferation of bladder cancer cells. Li et al (25) showed that miR-200c inhibited metastasis and invasion of human non-small cell lung cancer cells (25). Therefore, the present study expanded the current understanding of miR-200c functioning in human cancer.

N-cadherin is a cytoskeletal linker protein, which has a critical role in the regulation of cell motility (26). E-cadherin is a cell-cell adhesion molecule, and its upregulation promotes cell adhesion, while inhibiting cell motility (27,28). In the present study, it was observed that treatment with Aza led to a decreased expression of N-cadherin with an increased expression of E-cadherin in ccRCC cells, indicating that EMT was suppressed. By contrast, the knockdown of miR-200c resulted in an increased N-cadherin expression with a reduced E-cadherin expression in ccRCC cells, indicating the EMT was upregulated. As Aza treatment also enhanced miR-200c expression in ccRCC cells, the results of the present study suggest that the inhibitory effect of miR-200c on ccRCC cell migration and invasion may be partly due to inhibition of the EMT.

In conclusion, the present study has demonstrated that miR-200c was significantly downregulated in cells due to the hypermethylation status of its promoter. Furthermore, the upregulation of miR-200c induced by epigenetic drug treatment inhibited the migration, invasion and EMT of ccRCC cells. Therefore, epigenetic drugs that are able to mediate the expression of miR-200c may be utilized for the treatment of ccRCC metastasis.

References

1 

Novick AC: Kidney cancer: Past, present, and future. Urol Oncol. 25:188–195. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Chow WH, Dong LM and Devesa SS: Epidemiology and risk factors for kidney cancer. Nat Rev Urol. 7:245–257. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Azeem K, Kollarova H, Horakova D, Magnuskova S and Janout V: Genetic syndromes associated with renal cell carcinoma: A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 155:231–238. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Diamond E, Riches J, Faltas B, Tagawa ST and Nanus DM: Immunologics and chemotherapeutics for renal cell carcinoma. Semin Intervent Radiol. 31:91–97. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Rydzanicz M, Wrzesiński T, Bluyssen HA and Wesoły J: Genomics and epigenomics of clear cell renal cell carcinoma: Recent developments and potential applications. Cancer Lett. 341:111–126. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. 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 

Lu J, Getz G, Miska EA, et al: MicroRNA expression profiles classify human cancers. Nature. 435:834–838. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Watanabe K and Takai D: Disruption of the expression and function of microRNAs in lung cancer as a result of epigenetic changes. Front Genet. 4:2752013. View Article : Google Scholar : PubMed/NCBI

10 

Gao YC and Wu J: MicroRNA-200c and microRNA-141 as potential diagnostic and prognostic biomarkers for ovarian cancer. Tumour Biol. 36:4843–4850. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Sun X, Luo S, He Y, et al: Screening of the miRNAs related to breast cancer and identification of its target genes. Eur J Gynaecol Oncol. 35:696–700. 2014.PubMed/NCBI

12 

Zhou X, Wang Y, Shan B, et al: The downregulation of miR-200c/141 promotes ZEB1/2 expression and gastric cancer progression. Med Oncol. 32:4282015. View Article : Google Scholar : PubMed/NCBI

13 

Liu L, Qiu M, Tan G, et al: miR-200c inhibits invasion, migration and proliferation of bladder cancer cells through down-regulation of BMI-1 and E2F3. J Transl Med. 12:3052014. View Article : Google Scholar : PubMed/NCBI

14 

Shi R, Xiao H, Yang T, et al: Effects of miR-200c on the migration and invasion abilities of human prostate cancer Du145 cells and the corresponding mechanism. Front Med. 8:456–463. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Nakada C, Matsuura K, Tsukamoto Y, et al: Genome-wide microRNA expression profiling in renal cell carcinoma: Significant down-regulation of miR-141 and miR-200c. J Pathol. 216:418–427. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Liu H, Brannon AR, Reddy AR, et al: Identifying mRNA targets of microRNA dysregulated in cancer: With application to clear cell renal cell carcinoma. BMC Syst Biol. 4:512010. View Article : Google Scholar : PubMed/NCBI

17 

Livak and Schmittgen: 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

18 

Furuta M, Kozaki KI, Tanaka S, et al: miR-124 and miR-203 are epigenetically silenced tumor-suppressive microRNAs in hepatocellular carcinoma. Carcinogenesis. 31:766–776. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Funasaka T, Raz A and Nangia-Makker P: Galectin-3 in angiogenesis and metastasis. Glycobiology. 24:886–891. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Senanayake U, Das S, Vesely P, et al: miR-192, miR-194, miR-215, miR-200c and miR-141 are downregulated and their common target ACVR2B is strongly expressed in renal childhood neoplasms. Carcinogenesis. 33:1014–1021. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Niu Y, Des Marais TL, Tong Z, Yao Y and Costa M: Oxidative stress alters global histone modification and DNA methylation. Free Radic Biol Med. 82:22–28. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Zhang D, Li Q, Rao L, Yi B and Xu Q: Effect of 5-Aza-2′- deoxycytidine on odontogenic differentiation of human dental pulp cells. J Endod. 41:640–645. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Lu YX, Yuan L, Xue XL, et al: Regulation of colorectal carcinoma stemness, growth, and metastasis by an miR-200c-Sox2-negative feedback loop mechanism. Clin Cancer Res. 20:2631–2642. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Tamagawa S, Beder LB, Hotomi M, et al: Role of miR-200c/miR-141 in the regulation of epithelial-mesenchymal transition and migration in head and neck squamous cell carcinoma. Int J Mol Med. 33:879–886. 2014.PubMed/NCBI

25 

Li J, Tan Q, Yan M, et al: miRNA-200c inhibits invasion and metastasis of human non-small cell lung cancer by directly targeting ubiquitin specific peptidase 25. Mol Cancer. 13:1662014. View Article : Google Scholar : PubMed/NCBI

26 

Hristov M, Erl W and Weber PC: Endothelial progenitor cells: Mobilization, differentiation, and homing. Arterioscler Thromb Vasc Biol. 23:1185–1189. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Kumar KJ, Vani MG, Chueh PJ, Mau JL and Wang SY: Antrodin C inhibits epithelial-to-mesenchymal transition and metastasis of breast cancer cells via suppression of Smad2/3 and β-catenin signaling pathways. PLoS One. 10:e01171112015. View Article : Google Scholar : PubMed/NCBI

28 

Wang M, Liu X, Jiang G, Chen H, Guo J and Weng X: Relationship between LSD1 expression and E-cadherin expression in prostate cancer. Int Urol Nephrol. 47:485–490. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 11 Issue 5

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
Jiang J, Yi B, Ding S, Sun J, Cao W and Liu M: Demethylation drug 5-Aza-2'-deoxycytidine-induced upregulation of miR-200c inhibits the migration, invasion and epithelial-mesenchymal transition of clear cell renal cell carcinoma in vitro Retraction in /10.3892/ol.2021.12923. Oncol Lett 11: 3167-3172, 2016
APA
Jiang, J., Yi, B., Ding, S., Sun, J., Cao, W., & Liu, M. (2016). Demethylation drug 5-Aza-2'-deoxycytidine-induced upregulation of miR-200c inhibits the migration, invasion and epithelial-mesenchymal transition of clear cell renal cell carcinoma in vitro Retraction in /10.3892/ol.2021.12923. Oncology Letters, 11, 3167-3172. https://doi.org/10.3892/ol.2016.4364
MLA
Jiang, J., Yi, B., Ding, S., Sun, J., Cao, W., Liu, M."Demethylation drug 5-Aza-2'-deoxycytidine-induced upregulation of miR-200c inhibits the migration, invasion and epithelial-mesenchymal transition of clear cell renal cell carcinoma in vitro Retraction in /10.3892/ol.2021.12923". Oncology Letters 11.5 (2016): 3167-3172.
Chicago
Jiang, J., Yi, B., Ding, S., Sun, J., Cao, W., Liu, M."Demethylation drug 5-Aza-2'-deoxycytidine-induced upregulation of miR-200c inhibits the migration, invasion and epithelial-mesenchymal transition of clear cell renal cell carcinoma in vitro Retraction in /10.3892/ol.2021.12923". Oncology Letters 11, no. 5 (2016): 3167-3172. https://doi.org/10.3892/ol.2016.4364