MicroRNA-101 induces apoptosis in cisplatin-resistant gastric cancer cells by targeting VEGF-C

Retraction in: /10.3892/mmr.2021.12333

  • Authors:
    • Guangyan Li
    • Fang Yang
    • Shiyu Gu
    • Zhenjuan Li
    • Minghui Xue
  • View Affiliations

  • Published online on: November 12, 2015     https://doi.org/10.3892/mmr.2015.4560
  • Pages: 572-578
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Deregulation of microRNAs (miRNAs) is known to be associated with drug resistance in human cancers. However, the precise role of miR‑101 in the cisplatin (DPP) resistance of human gastric cancer cells has not been elucidated, yet. The present study revealed that miR‑101 was markedly downregulated in gastric cancer cell lines compared to that in the normal gastric mucosa epithelial cell line GES1. Furthermore, a significant decrease in miR‑101 levels, accompanied with an increased expression of vascular endothelial growth factor (VEGF)‑C in DDP‑resistant SGC7901 gastric cancer cells (SGC7901/DDP) compared with those in native SGC7901 cells was observed. In addition, forced overexpression of miR‑101 significantly inhibited cell proliferation, while enhancing cisplatin‑induced apoptosis of SGC7901/DDP cells. A luciferase reporter assay confirmed that VEGF‑C was a direct target of miR‑101 in SGC7901/DDP cells. Forced overexpression of miR‑101 in SGC7901/DDP cells reduced the expression of VEGF‑C, while knockdown of miR‑101 expression significantly enhanced VEGF‑C expression in SGC7901/DDP cells. Finally, overexpression of VEGF‑C inhibited DDP‑induced apoptosis in SGC7901 cells. In conclusion, the results of the present study suggested that miR‑101 inhibited the proliferation and promoted DDP‑induced apoptosis of DDP‑resistant gastric cancer cells, at least in part via targeting VEGF-C.

Introduction

Gastric cancer is one of the most common types of solid tumor and features poor prognosis as well as high mortality. Traditional treatments, including surgical resection, chemotherapy and radiotherapy, show limited curative effects, mainly due to metastasis, recurrence and drug resistance (1,2). Therefore, it is urgent to clarify the underlying molecular mechanisms of gastric cancer for the development of novel molecular therapeutic tools.

MicroRNAs (miRs), a class of small non-coding RNAs, have an inhibitory role in the regulation of gene expression by either inducing mRNA degradation or suppressing protein translation (3). Accumulating evidence has revealed that deregulations of miRNAs participate in the development and progression of human malignances, including gastric cancer (46). Recently, miR-101 has been found to act as a tumor suppressor in multiple types of malignant tumor, including hepatocellular carcinoma, breast cancer, cervical cancer and lung cancer (711). For instance, Guo et al (12) found that miR-101 suppressed the epithelial-to-mesenchymal transition in ovarian carcinoma by targeting zinc finger E-box binding homeobox 1 and 2. Lin et al (13) reported that miR-101 inhibited the proliferation of papillary thyroid carcinoma cells by targeting Ras-related C3 botulinum toxin substrate 1. Recently, miR-101 was reported to be frequently downregulated in gastric cancer tissues, and overexpression of miR-101 inhibited gastric cancer cell migration and invasion, suggesting that miR-101 has a suppressive role in gastric cancer (14,15). However, the exact role of miR-101 in drug resistance in gastric cancer as well as the underlying mechanisms have not been elucidated, yet.

The present study aimed to reveal the role of miR-101 in the regulation of cisplatin (DPP) resistance in human gastric cancer cells and investigated the underlying molecular mechanisms.

Materials and methods

Reagents

Dulbecco's modified Eagle's medium (DMEM), TRIzol, fetal bovine serum (FBS), Opti-minimum essential medium (MEM), miRNA Reverse Transcription kit, SYBR Ex Taq kit and Lipofectamine 2000 were purchased from Invitrogen (Thermo Fisher Scientific, Waltham, MA, USA). The miRNA Q-PCR Detection kit was purchased from GeneCopoeia (Rockville, MD, USA). Mouse anti-vascular endothelial growth factor (VEGF)-C antibody (1:100; cat. no. ab63221), mouse anti-GAPDH antibody (1:50; cat. no. ab8245) and rabbit anti-mouse secondary antibody (cat. no. ab46540) were purchased from Abcam (Cambridge, UK). An enhanced chemiluminescence (ECL) kit was purchased from Pierce Biotechnology, Inc. (Rockford, IL, USA). A Quick-Change Site-Directed Mutagenesis kit was purchased from Stratagene (La Jolla, CA, USA). PsiCHECK™ 2 vector was purchased from Promega Corp. (Madison, WI, USA). Apoptosis Detection kit I was purchased from BD Biosciences (Franklin Lakes, NJ, USA).

Cell culture

The SNU5, HGC27, BGC823, SGC7901 and AGS human gastric cancer cell lines, the DDP-resistant gastric cancer cell line SGC7901/DDP, as well as the normal gastric mucosa epithelial cell line GES1 were purchased from the Cell Bank of the Type Culture Collection of Chinese Academy of Sciences (Shanghai, China). Cells were cultured in DMEM with 10% FBS, 100 IU/ml penicillin and 100 mg/ml streptomycin (Sigma-Aldrich, St. Louis, MO, USA) at 37°C in a humidified atmosphere containing 5% CO2.

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

Total RNA was extracted with TRIzol reagent according to the manufacturer's instructions. The integrity of the large RNAs was confirmed by 1% denatured agarose gel electrophoresis. RT-qPCR was then performed to detect the expression of miR-101. In accordance with the manufacturer's instructions, RNA was reverse-transcribed into cDNA by using an miRNA Reverse Transcription kit. The cDNA was used for the amplification of mature miR-101 through PCR alongside U6 small nuclear RNA as an endogenous control. Real-time PCR was performed in an ABI 7500 thermocycler (Applied Biosystems; Thermo Fisher Scientific) using the miRNA Q-PCR Detection kit in accordance with the manufacturer's instructions. The PCR cycling conditions used were as follows: 94°C for 3 min, followed by 40 cycles of 94°C for 30 sec, 56°C for 30 sec and 72°C for 30 sec. To determine the mRNA levels of VEGF-C, the respective cDNA obtained by reverse transcription and the SYBR Ex Taq kit were used for PCR amplification. GAPDH was used as the endogenous control gene. The PCR cycling conditions used were as follows: 94°C for 3 min, followed by 40 cycles of 94°C for 30 sec, 60°C for 30 sec and 72°C for 30 sec. The following specific primers were used: VEGF-C forward, 5′-GAGGAGCAGTTACGGTCTGTG-3′ and reverse, 5′-TCCTTTCCTTAGCTGACACTTGT-3′. GAPDH forward, 5′-CTGGGCTACACTGAGCACC-3′ and reverse, 5′-AAGTGGTCGTTGAGGGCAATG-3′. The primers for U6 and miR-101 were as follows: MicroRNA-101, forward 5′-TACAGTACTGTGATAACTGAA-3′ and reverse 5′-GCGTGCTACAGTACTGTGATAACTG-3′; and U6, forward 5′-CTCGCTTCGGCAGCACA-3′ and reverse 5′-AACGCTTCACGAATTTGCGT-3′. The primers were supplied by Sango Biotech Co., Ltd. (Shanghai, China). The relative mRNA expression of VEGF-C was normalized to GAPDH. The relative expression was analyzed using the 2−ΔΔCt method (16).

Transfection

Transfection was performed using the Lipofectamine 2000 reagent according to the manufacturer's instructions. At the time of transfection, cells were at least 70% confluent. Oligonucleotides and plasmids were incubated in Opti-MEM medium. For miR-101 functional analysis, SGC7901/DDP cells were transfected with miR-101 mimics or miR-101 inhibitor (all from Thermo Fisher Scientific), respectively. For VEGF-C functional analysis, SGC7901/DDP cells were transfected with a blank vector (PcDNA™ 3.1 (+) mammalian expression vector; cat. no. 790-20; (Thermo Fisher Scientific)) or the VEGF-C plasmid (PcDNA™ 3.1 (+)-VEGF-C expression plasmid; Genetalks, Changsha, China), respectively.

Bioinformatics analysis

Bioinformatics analyses were performed to predict the putative target genes of miR-101 using TargetScan software (www.targetscan.org/).

Luciferase reporter assay

In accordance with the manufacturer's instructions, the Quick-Change Site-Directed Mutagenesis kit was used to generate a mutant-type 3′-untranslated region (3′-UTR) of VEGF-C. The wild-type or mutant-type 3′-UTR of VEGF-C was inserted into the psiCHECK™ 2 vector, respectively. After SGC7901/DDP cells were cultured to ~70% confluence, they were transfected with psiCHECK™ 2-VEGF-C-3′-UTR or psiCHECK™ 2-mutant VEGF-C-3′-UTR vector, with or without 100 nM miR-101 mimics, respectively. After transfection for 48 h, the luciferase activities were determined on a LD400 luminometer (Beckman Coulter, Brea, CA, USA). Renilla luciferase activity was normalized to firefly luciferase activity.

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

At 24 h following transfection, cells (5,000 per well) were seeded in a 96-well plate. After incubation for 48 h, the cells, which were not incubated with DPP, were incubated with MTT (0.5 mg/ml) at 37°C for 4 h. The medium was removed and the precipitated formazan was dissolved in 100 ml dimethyl sulfoxide. The absorbance at 570 nm was then detected using a microplate reader (PHERAstar FS; Life Technologies, Grand Island, NY, USA).

Apoptosis assay

Cells were treated with DPP (1 mM; TCI Chemicals, Shanghai, China) for 6 h, following which cell apoptosis was examined. The cells were seeded in six-well plates. For each group, flow cytometry was used (Accuri C5 Flow Cytometer; BD Biosciences, Franklin Lakes, NJ, USA) to determine the apoptotic rate of SGC7901/DDP cells by determining the relative amount of Annexin V-fluorescein isothiocyanate-positive/propidium iodide-negative cells using an Apoptosis Detection Kit I according to the manufacturer's instructions.

Western blot analysis

Cells were lysed with radioimunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Haimen, China) to extract the protein. After the protein concentration was determined, equal amounts (25 µg/lane) were loaded onto a 12% sodium dodecyl sulfate polyacrylamide gel (Beyotime Institute of Biotechnology), subjected to electrophoresis and transferred onto a polyvinylidene difluoride (PVDF) membrane (Thermo Fisher Scientific). The PVDF membrane was then incubated with mouse monoclonal anti-VEGF-C antibody and mouse monoclonal anti-GAPDH antibody, respectively, at room temperature for 3 h. After washing with phosphate-buffered saline containing Tween 20 three times, the PVDF membrane was incubated with the horseradish peroxidase-conjugated rabbit anti-mouse secondary antibody at room temperature for 1 h. An ECL kit was used for chemiluminescent detection. Images were captured using a ChemiDoc MP system (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The relative protein expression was analyzed by Image-Pro plus software (version 6.0; Media Cybernetics, Rockville, MD, USA). The protein concentrations were determined using a Pierce BCA Protein Assay kit (Thermo Fisher Scientific), in accordance with the manufacturer's protocol.

Statistical analysis

Values are expressed as the mean ± standard deviation of three independent experiments. Statistical analysis of differences was performed using the two-tailed Student's t test. SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA) was used to perform statistical analyses. P<0.05 was considered to indicate a statistically significant difference between values.

Results

miR-101 is frequently downregulated in gastric cancer cell lines

The present study first determined the expression levels of miR-101 in five common gastric cancer cell lines, SNU5, HGC27, BGC823, SGC7901 and AGS, as well as in the normal gastric mucosa epithelial cell line GES1 as a control using real-time RT-qPCR. As shown in Fig. 1, miR-101 was frequently downregulated in gastric cancer cell lines compared to that in the GES1 normal gastric mucosa epithelial cell line. As the downregulation of miR-101 expression was most significant in SGC7901 cells, the present study used DDP-resistant SGC7901/DDP cells in all subsequent experiments to investigate the role of miR-101 in the drug resistance of gastric cancer.

Inverse expression of miR-101 and VEGF-C in DDP-resistant gastric cancer cells

RT-qPCR was performed to determine the expression of miR-101 in SGC7901/DDP cells as well as in SGC7901 cells as a control. As shown in Fig. 2A, the expression levels of miR-101 in SGC7901/DDP cells were significantly lower than those in SGC7901 cells. Furthermore, RT-qPCR and western blot analysis was used to assess the mRNA and protein expression of VEGF-C in these two cell lines. As shown in Fig. 2B and C, the mRNA and protein levels of VEGF-C were markedly increased in SGC7901/DDP cells compared to those in SGC7901 cells. These findings suggested that deregulation of miR-101 and VEGF-C may be involved in the drug resistance of SGC7901/DDP gastric cancer cells.

miR-101 inhibits proliferation and promotes DDP-induced apoptosis of SGC7901/DDP cells

The role of miR-101 in the proliferation and DDP-induced apoptosis of SGC7901/DDP cells was explored. After transfection of SGC7901/DDP cells with miR-101 mimics or inhibitor, the levels of miR-101 were determined. As shown in Fig. 3A, the levels of miR-101 were significantly upregulated after transfection with miR-101 mimics, while transfection with miR-101 inhibitor significantly reduced the expression of miR-101 in SGC7901/DDP cells when compared to that in the control group. Subsequently, the cell proliferation in each group was determined. As demonstrated in Fig. 3B, upregulation of miR-101 significantly inhibited the proliferation, while downregulation of miR-101 significantly promoted the proliferation of SGC7901/DDP cells when compared with that in the control group. Furthermore, the apoptotic rate following DDP treatment was determined in each group. As shown in Fig. 3C, overexpression of miR-101 promoted DDP-induced apoptosis, while inhibition of miR-101 expression suppressed DDP-induced apoptosis when compared to that in the control group. These findings indicated that miR-101 inhibited the proliferation of SGC7901/DDP cells while enhancing their sensitivity to DDP with an increased apoptotic rate.

VEGF-C is a direct target of miR-101 in SGC7901/DDP cells

According to bioinformatics analysis, VEGF-C was predicted to be a potential target gene of miR-101. To verify this, the wild-type and mutant-type 3′-UTRs of VEGF-C were generated (Fig. 4A) and inserted into luciferase reporter vectors, which were transfected into DDP-resistant human gastric cancer SGC7901/DDP cells. As shown in Fig. 4B, the luciferase activity was significantly decreased in SGC7901/DDP cells co-transfected with miR-101 mimics and the plasmid expressing the wild-type VEGF-C 3′-UTR. However, the luciferase activity was unaltered in cells co-transfected with miR-101 mimics and the plasmid expressing the mutant-type VEGF-C 3′-UTR. These results indicated that VEGF-C is a target gene of miR-101 in SGC7901/DDP human gastric cancer cells. Furthermore, the present study investigated the effects of the upregulation and downregulation of miR-101 on the expression of VEGF-C in SGC7901/DDP cells. As demonstrated in Fig. 4C, following overexpression of miR-101, the protein expression of VEGF-C was significantly decreased. By contrast, inhibition of miR-101 enhanced the protein expression of VEGF-C in SGC7901/DDP cells. All of these findings suggested that miR-101 negatively mediates the expression of VEGF-C in SGC7901/DDP cells.

Overexpression of VEGF-C inhibits DDP-induced apoptosis in SGC7901/DDP cells

While the abovementioned findings suggested that miR-101 promotes DDP-induced apoptosis in SGC7901/DDP cells through directly targeting VEGF-C, it remained to be clarified whether VEGF-C is indeed involved in the DDP resistance in gastric cancer cells. To confirm this speculation, SGC7901/DDP cells were transfected with VEGF-C to upregulate the VEGF-C overexpression plasmid, following which the protein expression of VEGF-C was significantly increased (Fig. 5A). Subsequently, the cells' sensitivity to DDP-induced apoptosis was tested. As shown in Fig. 5B, overexpression of VEGF-C significantly inhibited DDP-induced apoptosis in SGC7901/DDP cells when compared with that in the untransfected control SGC7901/DDP cells. These findings suggested that VEGF-C is indeed involved in the DDP resistance of gastric cancer cells.

Discussion

miR-101 has been reported be downregulated in human gastric cancer and has an inhibitory role in mediating the proliferation, migration and invasion of gastric cancer cells (14). However, the exact role of miR-101 in the drug resistance of gastric cancer as well as the underlying mechanism have not been elucidated, to the best of our knowledge. The present study found that miR-101 was frequently downregulated in gastric cancer cell lines when compared with that in normal gastric mucosa epithelial cells, consistent with the results of previous studies (14,15). It has been reported that the expression levels of miR-101 in gastric cancer tissues were significantly reduced when compared to those in their matched normal tissues, and lower levels of miR-101 were associated with increased tumor invasion and lymph-node metastasis, suggesting that downregulation of miR-101 may have a role in the progression of gastric cancer (14,15).

To reveal the role of miR-101 in the regulation of drug resistance in gastric cancer, the DDP-resistant gastric cancer cell line SGC7901/DDP was used, in which miR-101 was downregulated compared to that in SGC7901 cells. Furthermore, forced overexpression of miR-101 significantly inhibited the proliferation and promoted DDP-induced apoptosis in SGC7901/DDP cells, indicating that miR-101 sensitized SGC7901/DDP cells to DDP. In fact, similar findings have also been reported in other types of human cancer. For instance, Xu et al (7) reported that miR-101 enhanced DDP-induced apoptosis in hepatocellular carcinoma cells, probably via inhibition of autophagy. In analogy with this, Chang et al (17) also found that miR-101 enhanced osteosarcoma-cell chemosensitivity to DDP through suppressing autophagy.

Apart from mediating autophagy, miR-101 has also been demonstrated to be associated with sensitivity to drugs in several types of human cancer, probably through inhibiting the expression of genes associated with drug resistance. For instance, Liu et al (18) found that overexpression of miR-101 inhibited the expression of zeste homolog 2 and re-sensitized drug-resistant epithelial ovarian cancer cells to DDP (18). Bu et al (19) found that enforced expression of miR-101 promoted cisplatin sensitivity in human bladder cancer cells via inhibition of the cyclooxygenase-2 pathway. As one miR can mediate numerous target genes, the present study further explored the potential targets of miR-101 involved in the DDP resistance of gastric cancer. Through performing a bioinformatics analysis and a luciferase reporter assay, VEGF-C was identified as a target gene of miR-101, and miR-101 was revealed to negatively regulate the protein expression of VEGF-C through directly binding to the 3′-UTR of VEGF-C mRNA in SGC7901/DDP cells. In addition, it was found that the mRNA expression of VEGF was also negatively affected by miR-101, and there may be additional indirect regulatory pathways involved in miR-101-mediated VEGF expression.

VEGF-C, a member of the VEGF family, can promote angiogenesis and endothelial cell growth, as well as affect the permeability of blood vessels, through directly binding and activating VEGF receptor 2 and 3 (2022). A recent study suggested that de-regulation of VEGF-C is associated with the development and progression of gastric cancer (23). Wang et al (23) reported that high expression VEGF-C was significantly correlated with extensive lymph-node metastasis, an advanced stage and the Lauren type of gastric adenocarcinoma. In addition, the expression of VEGF-C was shown to be associated with poor survival in gastric cancer tissues (24). These previous studies suggested that VEGF-C has an oncogenic role in gastric cancer. In addition, VEGF-C was found to be involved in drug resistance of several types of human malignancies (2527). For instance, Li et al (26) found that VEGF-C was significantly upregulated in the Lewis murine lung carcinoma cell line which is resistant to the treatment with a potent VEGF inhibitor, while inhibition of VEGF-C in resistant cells suppressed endothelial-cell migration and partially restored sensitivity to anti-VEGF therapy. Hua et al (27) reported that VEGF-C modulated chemoresistance in acute myeloid leukemic cells through an endothelin-1-dependent induction of cyclooxygenase-2. The present study found that overexpression of VEGF-C significantly inhibited DDP-induced apoptosis in gastric cancer cells, suggesting that VEGF-C is indeed involved in drug resistance in gastric cancer. Cho et al (25) also showed that VEGF-C depletion sensitized RhoGDI2-overexpressing cells to DDP-induced apoptosis in vitro and in vivo, while secreted VEGF-C conferred DDP resistance to RhoGDI2-overexpressing gastric cancer cells. Furthermore, the present study showed that the expression of VEGF-C was markedly upregulated in DDP-resistant gastric cancer cells, accompanied with the downregulation of miR-101. Based on these findings, it is indicated that the miR-101/VEGF-C axis is involved in the regulation of DDP resistance in gastric cancer.

In conclusion, the present study showed that miR-101 inhibited the proliferation and promoted DDP-induced apoptosis in DPP-resistant gastric cancer cells, at least in part through inhibition of VEGF-C expression by targeting the 3′UTR of VEGF-C mRNA. Therefore, the present study provided novel insight into the drug resistance in gastric cancer, and indicated that miR-101 may be used for the treatment of drug-resistant gastric cancer.

References

1 

Pasechnikov V, Chukov S, Fedorov E, Kikuste I and Leja M: Gastric cancer: Prevention, screening and early diagnosis. World J Gastroenterol. 20:13842–13862. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Shi J, Qu YP and Hou P: Pathogenetic mechanisms in gastric cancer. World J Gastroenterol. 20:13804–13819. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Baer C, Claus R and Plass C: Genome-wide epigenetic regulation of miRNAs in cancer. Cancer Res. 73:473–477. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Bouyssou JM, Manier S, Huynh D, Issa S, Roccaro AM and Ghobrial IM: Regulation of microRNAs in cancer metastasis. Biochim Biophys Acta. 1845:255–265. 2014.PubMed/NCBI

6 

Matuszcak C, Haier J, Hummel R and Lindner K: MicroRNAs: Promising chemoresistance biomarkers in gastric cancer with diagnostic and therapeutic potential. World J Gastroenterol. 20:13658–13666. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Xu Y, An Y, Wang Y, Zhang C, Zhang H, Huang C, Jiang H, Wang X and Li X: miR-101 inhibits autophagy and enhances cisplatin-induced apoptosis in hepatocellular carcinoma cells. Oncol Rep. 29:2019–2024. 2013.PubMed/NCBI

8 

Wang L, Li L, Guo R, Li X, Lu Y, Guan X, Gitau SC, Wang L, Xu C, Yang B and Shan H: miR-101 promotes breast cancer cell apoptosis by targeting janus kinase 2. Cell Physiol Biochem. 34:413–422. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Liang X, Liu Y, Zeng L, Yu C, Hu Z, Zhou Q and Yang Z: miR-101 Inhibits the G1-to-S phase transition of cervical cancer cells by targeting Fos. Int J Gynecol Cancer. 24:1165–1172. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Zhang JG, Guo JF, Liu DL, Liu Q and Wang JJ: MicroRNA-101 exerts tumor-suppressive functions in non-small cell lung cancer through directly targeting enhancer of zeste homolog 2. J Thorac Oncol. 6:671–678. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Konno Y, Dong P, Xiong Y, Suzuki F, Lu J, Cai M, Watari H, Mitamura T, Hosaka M, Hanley SJ, et al: MicroRNA-101 targets EZH2, MCL-1 and FOS to suppress proliferation, invasion and stem cell-like phenotype of aggressive endometrial cancer cells. Oncotarget. 5:6049–6062. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Guo F, Cogdell D, Hu L, Yang D, Sood AK, Xue F and Zhang W: MiR-101 suppresses the epithelial-to-mesenchymal transition by targeting ZEB1 and ZEB2 in ovarian carcinoma. Oncol Rep. 31:2021–2028. 2014.PubMed/NCBI

13 

Lin X, Guan H, Li H, Liu L, Liu J, Wei G, Huang Z, Liao Z and Li Y: miR-101 inhibits cell proliferation by targeting Rac1 in papillary thyroid carcinoma. Biomed Rep. 2:122–126. 2014.PubMed/NCBI

14 

He XP, Shao Y, Li XL, Xu W, Chen GS, Sun HH, Xu HC, Xu X, Tang D, Zheng XF, et al: Downregulation of miR-101 in gastric cancer correlates with cyclooxygenase-2 overexpression and tumor growth. FEBS J. 279:4201–4212. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Wang HJ, Ruan HJ, He XJ, Ma YY, Jiang XT, Xia YJ, Ye ZY and Tao HQ: MicroRNA-101 is down-regulated in gastric cancer and involved in cell migration and invasion. Eur J Cancer. 46:2295–2303. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Liu SQ, Yu HC, Gong YZ and Lai NS: Quantitative measurement of HLA-B27 mRNA in patients with ankylosing spondylitis–correlation with clinical activity. J Rheumatol. 33:1128–1132. 2006.PubMed/NCBI

17 

Chang Z, Huo L, Li K, Wu Y and Hu Z: Blocked autophagy by miR-101 enhances osteosarcoma cell chemosensitivity in vitro. Scientific World Journal. 2014:7947562014. View Article : Google Scholar : PubMed/NCBI

18 

Liu L, Guo J, Yu L, Cai J, Gui T, Tang H, Song L, Wang J, Han F, Yang C, et al: miR-101 regulates expression of EZH2 and contributes to progression of and cisplatin resistance in epithelial ovarian cancer. Tumour Biol. 35:12619–12626. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Bu Q, Fang Y, Cao Y, Chen Q and Liu Y: Enforced expression of miR-101 enhances cisplatin sensitivity in human bladder cancer cells by modulating the cyclooxygenase-2 pathway. Mol Med Rep. 10:2203–2209. 2014.PubMed/NCBI

20 

Shibuya M: VEGF-VEGFR signals in health and disease. Biomol Ther (Seoul). 22:1–9. 2014. View Article : Google Scholar

21 

Goel HL and Mercurio AM: VEGF targets the tumour cell. Nat Rev Cancer. 13:871–882. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Liang X, Xu F, Li X, Ma C, Zhang Y and Xu W: VEGF signal system: The application of antiangiogenesis. Curr Med Chem. 21:894–910. 2014. View Article : Google Scholar

23 

Wang L, Li HG, Wen JM, Peng TS, Zeng H and Wang LY: Expression of CD44v3, erythropoietin and VEGF-C in gastric adenocarcinomas: Correlations with clinicopathological features. Tumori. 100:321–327. 2014.PubMed/NCBI

24 

Cao W, Fan R, Yang W and Wu Y: VEGF-C expression is associated with the poor survival in gastric cancer tissue. Tumour Biol. 35:3377–3383. 2014. View Article : Google Scholar

25 

Cho HJ, Kim IK, Park SM, Baek KE, Nam IK, Park SH, Ryu KJ, Choi J, Ryu J, Hong SC, et al: VEGF-C mediates RhoGDI2-induced gastric cancer cell metastasis and cisplatin resistance. Int J Cancer. 135:1553–1563. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Li D, Xie K, Ding G, Li J, Chen K, Li H, Qian J, Jiang C and Fang J: Tumor resistance to anti-VEGF therapy through up-regulation of VEGF-C expression. Cancer Lett. 346:45–52. 2014. View Article : Google Scholar

27 

Hua KT, Lee WJ, Yang SF, Chen CK, Hsiao M, Ku CC, Wei LH, Kuo ML and Chien MH: Vascular endothelial growth factor-C modulates proliferation and chemoresistance in acute myeloid leukemic cells through an endothelin-1-dependent induction of cyclooxygenase-2. Biochim Biophys Acta. 1843:387–397. 2014. View Article : Google Scholar

Related Articles

Journal Cover

January-2016
Volume 13 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li G, Yang F, Gu S, Li Z and Xue M: MicroRNA-101 induces apoptosis in cisplatin-resistant gastric cancer cells by targeting VEGF-C Retraction in /10.3892/mmr.2021.12333. Mol Med Rep 13: 572-578, 2016
APA
Li, G., Yang, F., Gu, S., Li, Z., & Xue, M. (2016). MicroRNA-101 induces apoptosis in cisplatin-resistant gastric cancer cells by targeting VEGF-C Retraction in /10.3892/mmr.2021.12333. Molecular Medicine Reports, 13, 572-578. https://doi.org/10.3892/mmr.2015.4560
MLA
Li, G., Yang, F., Gu, S., Li, Z., Xue, M."MicroRNA-101 induces apoptosis in cisplatin-resistant gastric cancer cells by targeting VEGF-C Retraction in /10.3892/mmr.2021.12333". Molecular Medicine Reports 13.1 (2016): 572-578.
Chicago
Li, G., Yang, F., Gu, S., Li, Z., Xue, M."MicroRNA-101 induces apoptosis in cisplatin-resistant gastric cancer cells by targeting VEGF-C Retraction in /10.3892/mmr.2021.12333". Molecular Medicine Reports 13, no. 1 (2016): 572-578. https://doi.org/10.3892/mmr.2015.4560