MicroRNA‑655 inhibits cell proliferation and invasion in epithelial ovarian cancer by directly targeting vascular endothelial growth factor

Retraction in: /10.3892/mmr.2022.12860

  • Authors:
    • Ziling Zhao
    • Shuna Yang
    • Yan Cheng
    • Xiuhua Zhao
  • View Affiliations

  • Published online on: May 29, 2018     https://doi.org/10.3892/mmr.2018.9090
  • Pages: 1878-1884
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In recent years, microRNAs (miRNAs/miRs) have been shown to be deregulated in epithelial ovarian cancer (EOC). Their deregulation has been suggested to be involved in EOC formation and progression through the regulation of the expression of numerous cancer‑related genes. Hence, it is of great importance to further determine the detailed roles and underlying mechanisms of miRNAs involved in EOC and to identify novel targets for diagnosis, prognosis and treatment of patients with EOC. In this study, the expression of miR‑655‑3p (miR‑655) was significantly downregulated in EOC tissues and four EOC cell lines. After miR‑655 was restored, functional assays revealed that cellular proliferation and invasion were considerably reduced in EOC. Additionally, vascular endothelial growth factor (VEGF) A was identified as a direct target gene of miR‑655 in EOC cells. Furthermore, VEGF knockdown could mimic the tumour‑suppressive roles of miR‑655 overexpression in EOC cells. Moreover, the introduction of VEGF abrogated the effects of miR‑655‑induced proliferation and invasion inhibition in EOC cells. Altogether, these findings indicated that miR‑655 may inhibit EOC cell proliferation and invasion by repressing VEGF. Thus, the miR‑655/VEGF pathway could serve as a novel therapeutic target for patients with EOC.

Introduction

Ovarian cancer is the fifth leading cause of cancer-related deaths resulting from gynaecological malignancies worldwide (1). Over 204,000 newly diagnosed cases and 125,000 mortalities related to ovarian cancer are estimated each year globally (2). Epithelial ovarian cancer (EOC) is a major subtype of ovarian cancer that accounts for 90% of ovarian cancer cases (3). Although recent advancements have been achieved in clinical and experimental oncology, the long-term survival of patients with EOC remains unsatisfactory with a five-year survival rate of only 35% (4). Malignant growth, tumour recurrence, metastasis and poor response to chemo-/radiotherapy are mainly responsible for the poor treatment outcomes of EOC patients (5). In addition, the molecular mechanisms underlying EOC oncogenesis and malignant development are poorly defined, thereby limiting the efficiency of clinical treatment (6). Therefore, identifying the molecular mechanisms of EOC pathogenesis and progression will greatly benefit the development of therapeutic methods to improve the prognosis of patients with EOC.

MicroRNAs (miRNAs/miRs) are a group of endogenous, non-coding and short RNA molecules that are normally expressed in animals, plants and certain viruses (7). miRNAs are considered as novel gene regulators that regulate gene expression by directly binding to the 3′-untranslated regions (3′-UTRs) of their target mRNAs to induce mRNA degradation and/or translation inhibition, thereby reducing the expression of the associated protein products (8). Half of the human miRNAs are located at cancer-related genomic regions, suggesting that miRNAs may play crucial roles in carcinogenesis and cancer progression (9). Considerable empirical evidence suggested that miRNAs are aberrantly expressed in almost all types of human malignancies and are implicated in the regulation of many cell behaviours, including cell growth, cycle, apoptosis, metastasis and differentiation (1012). miRNAs may play tumour-suppressive or oncogenic roles in human cancers depending on the cellular context and their target genes (13). Therefore, restoring or inhibiting miRNAs may be attractive therapeutic strategies for anticancer therapy.

miR-655-3p (miR-655) is aberrantly underexpressed in several types of human cancers, such as hepatocellular carcinoma (14,15), oesophageal squamous cell carcinoma (16,17) and breast cancer (18). However, the expression pattern and biological roles of miR-655 in EOC and the underlying mechanisms for its functional significance remain unknown. In this study, we detected miR-655 expression in EOC tissues and cell lines and determined the roles of miR-655 in EOC cells. We also investigated the molecular mechanisms by which miR-655 inhibits EOC cell progression. Our study identified the critical roles of miR-655 in EOC and discovered a potential molecular therapeutic target for patients with EOC.

Materials and methods

Patients and tissue samples

In total, 23 pairs of EOC tissues and adjacent non-neoplastic ovarian tissues were obtained from patients (age range, 48–74 years; mean age, 59 years; 17 serous, 6 non-serous) who underwent surgical resection at the Yidu Central Hospital of Weifang between August 2015 and February 2017. All patients had not received chemotherapy, radiotherapy or any other treatments prior to surgery. All tissues were quickly snap-frozen in liquid nitrogen and stored at −80°C for future use. This study was approved by the Ethics Committee of the Yidu Central Hospital of Weifang, and written informed consent was obtained from all EOC patients enrolled in this study.

Cell culture and transfection

Four human EOC cell lines (SKOV3, OVCAR3, ES-2 and CAOV-3) were ordered from the American Type Culture Collection (Manassas, VA, USA). EOC cell lines were cultured in Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS), 100 mg/ml penicillin and 100 mg/ml streptomycin (all from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA). A human normal ovarian epithelial cell line (NOEC) was purchased from ScienCell Research Laboratories (cat. no. 7310; Carlsbad, CA, USA), and was grown in Ham's F-12 supplemented with 20% FBS, 120 mg/ml streptomycin and 120 mg/ml penicillin (all from Gibco; Thermo Fisher Scientific, Inc.). All these cells were maintained at 37°C in a humidified atmosphere containing 5% CO2. Cells in logarithmic phase were harvested for subsequent usage.

miR-655 mimics and negative control mimic (miR-NC) were purchased from GenePharma (Shanghai, China). To knock down VEGF expression, small interfering RNA (siRNA) targeting the expression of VEGF (si-VEGF) and negative control siRNA (si-NC) were chemically synthesized by Ribobio (Guangzhou, China). VEGF restoration plasmid (pcDNA3.1-VEGF) and empty pcDNA3.1 plasmid were produced by GeneCopoeia (Guangzhou, China). Cells were seeded into 6-well plates one day prior to transfection. These molecular products were introduced into cells using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) in accordance with the manufacturer's protocol.

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

The total RNA was extracted from tissue samples or cultured cells by using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.), following the manufacturer's instructions. To quantify miR-655 expression, the complementary DNA was synthesised using a TaqMan MicroRNA Reverse Transcription Kit and then subjected to qPCR by using a TaqMan MicroRNA assay kit (both from Applied Biosystems; Thermo Fisher Scientific, Inc.). U6 snRNA was used as the internal control for miR-655 expression. To detect VEGF mRNA expression, reverse transcription was performed using a PrimeScript® RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China). Subsequently, VEGF expression was determined using a SYBR Premix Ex Taq kit (Takara Biotechnology Co., Ltd.). GAPDH served as the internal reference for VEGF expression. Relative gene expression was analysed by the 2−ΔΔCq method (19).

MTT assay

The transfected cells were collected at 24 h post-transfection, prepared as a single-cell suspension and seeded at a density of 3,000 cells per well into 96-well plates. Cell proliferation was determined using MTT assay at 0, 24, 48 and 72 h post-inoculation. At every time point, 20 µl of MTT solution (5 mg/ml; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was added into each well, which was then incubated at 37°C for another 4 h. After the supernatant was discarded, 150 µl of dimethyl sulfoxide (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was added into each well to dissolve the crystals. After gentle agitation for 15 min, the absorbance was detected at a wavelength of 490 nm with an enzyme-linked immunosorbent detector (De Tie Inc., Nanjing, China).

Transwell invasion assay

Transwell chambers coated with Matrigel (8 µm pores; BD Biosciences, San Jose, CA, USA) were employed to assess cell invasive ability. A total of 1×105 transfected cells, which were suspended in 200 µl of DMEM without FBS, were plated into the upper chambers. The lower chambers were filled with 500 µl of DMEM supplemented with 20% FBS to serve as a chemoattractant. After 24 h incubation at 37°C, the cells remaining on the upper surface of the Transwell chambers were gently removed using a cotton swab. The invaded cells were fixed in 75% ethanol, stained with 0.5% crystal violet and washed with PBS. The number of invaded cells was counted under an inverted microscope (Olympus, Tokyo, Japan) in five randomly selected fields.

Bioinformatic prediction and luciferase reporter assay

TargetScan (https://www.targetscan.org) and microRNA.org (www.microrna.org/microrna/) were used to predict the putative targets of miR-655. The wild-type (Wt) 3′-UTR of VEGF containing predicted miR-655 binding sequences and mutant (Mut) 3′-UTR of VEGF was chemically produced by GenePharma, and subcloned into the pGL3 reporter vector (Promega, Madison, WI, USA). Cells were plated into 24-well plates, and cotransfected with Wt or Mut VEGF 3′-UTR reported plasmid and miR-655 mimics or miR-NC using Lipofectamine 2000, according to the manufacturer's instructions. The pRL-TK plasmid with constitutive expression of Renilla luciferase (Promega) was also transfected into cells. After 48 h of incubation, luciferase activities were analzyed using a dual-luciferase reporter assay system (Promega). Firefly luciferase activities were normalized to Renilla luciferase activities.

Western blot analysis

To extract the proteins, cells or homogenised tissues were lysed in radioimmunoprecipitation assay lysis buffer (Nanjing KeyGen Biotech Co., Ltd., Nanjing, China). The protein concentrations were evaluated using a BCA Protein Assay kit (Nanjing KeyGen Biotech Co., Ltd.). Equal amounts of protein were separated by 10% SDS-PAGE and transferred onto polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were blocked with TBS containing 0.1% Tween-20 (TBST) with 5% skim milk and incubated overnight at 4°C with primary antibodies: Mouse anti-human monoclonal VEGF (ab1316; 1:500 dilution; Abcam, Cambridge, UK) and mouse anti-human monoclonal GAPDH antibody (ab110305; 1:500 dilution; Abcam). Thereafter, the membranes were washed three times with TBST and incubated with goat anti-mouse horseradish peroxidase-conjugated IgG secondary antibodies (ab205719; 1:5,000 dilution; Abcam) at room temperature for 2 h. After thrice washing with TBST, the protein signals were visualised using an enhanced chemiluminescence detection system (Pierce; Thermo Fisher Scientific, Inc.). GAPDH was used as the loading control.

Statistical analysis

Data were expressed as the mean ± standard deviation, and analyzed with SPSS software version 18.0 (SPSS, Inc., Chicago, IL, USA). Group differences were evaluated using two-tailed Student's t-test or one-way analysis of variance followed by Student-Newman-Keuls post hoc test. The association between expression levels of miR-655 and VEGF mRNA was determined using Spearman's correlation analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-655 expression is downregulated in EOC tissues and cell lines

To investigate the functional significance of miR-655 in EOC, we measured miR-655 expression in 23 pairs of EOC tissues and adjacent non-neoplastic ovarian tissues by using RT-qPCR. The results revealed that miR-655 expression was decreased in EOC tissues relative to that in non-neoplastic ovarian tissues (P<0.05; Fig. 1A). In addition, the expression level of miR-655 in four human EOC cell lines (SKOV3, OVCAR3, ES-2 and CAOV-3) and a human normal ovarian epithelial NOEC cell line was detected. Compared with NOEC, miR-655 was significantly downregulated in the four EOC cell lines (P<0.05; Fig. 1B). SKOV3 and OVCAR3 cells expressed relatively lower miR-655 expression compared with the other two EOC cell lines. Therefore, they were selected for the subsequent experiments.

miR-655 inhibits EOC cellular proliferation and invasion in vitro

Given that miR-655 was underexpressed in EOC, we hypothesised that miR-655 may play crucial roles in EOC progression. To test our hypothesis, miR-655 mimics was transfected into SKOV3 and OVCAR3 cells to increase endogenous miR-655 level. The results of RT-qPCR analysis revealed that transfection with miR-655 mimics significantly increased the miR-655 expression level in SKOV3 and OVCAR3 cells compared with that in cells transfected with miR-NC (P<0.05; Fig. 2A). MTT assay was performed to investigate the effects of miR-655 overexpression on EOC cellular proliferation. As shown in Fig. 2B, resumption expression of miR-655 significantly reduced the proliferation of SKOV3 and OVCAR3 cells (P<0.05; Fig. 2B). To investigate whether miR-655 affects EOC cell invasion ability, Transwell invasion assay was conducted on SKOV3 and OVCAR3 cells that were transfected with miR-655 mimics or miR-NC. The results indicated that miR-655 upregulation substantially reduced the invasion abilities of SKOV3 and OVCAR3 cells (P<0.05; Fig. 2C). Taken together, miR-655 may play a tumour-suppressive role in EOC development.

VEGF is a direct target gene of miR-655 in EOC cells

To clarify the mechanism underlying the tumour-suppressive roles of miR-655 in EOC, bioinformatics analysis was performed to predict the putative targets of miR-655. VEGF, a well-known oncogene in EOC (2024), was a candidate target gene of miR-655 (Fig. 3A). Luciferase reporter assay was performed to examine whether miR-655 could directly interact with the 3′-UTR of VEGF. The results revealed that ectopic expression of miR-655 significantly reduced the luciferase activities of the reporter plasmid carrying the wild-type (Wt) 3′-UTR of VEGF in SKOV3 and OVCAR3 cells (P<0.05). However, upregulation of miR-655 exhibited no significant effect on the luciferase activities of the reporter plasmid containing the mutant (Mut) 3′-UTR of VEGF (Fig. 3B).

To explore the relationship between miR-655 and VEGF in EOC, RT-qPCR analysis was conducted to detect VEGF mRNA expression in 23 pairs of EOC tissues and adjacent non-neoplastic ovarian tissues. The expression level of VEGF mRNA was significantly higher in EOC tissues than in non-neoplastic ovarian tissues (P<0.05; Fig. 3C). Furthermore, Spearman's correlation analysis indicated an inverse association between miR-655 and VEGF mRNA in EOC tissues (r=−0.5564, P=0.0058; Fig. 3D). Moreover, we evaluated the effects of miR-655 overexpression on VEGF expression in SKOV3 and OVCAR3 cells by using RT-qPCR and Western blot analysis, respectively. Enforced expression of miR-655 in SKOV3 and OVCAR3 cells significantly reduced VEGF expression at both mRNA (P<0.05; Fig. 3E) and protein (P<0.05; Fig. 3F) levels. These results suggested that VEGF is a direct target of miR-655 in EOC cells.

VEGF knockdown simulates the tumour-suppressing roles of miR-655 overexpression in EOC

Considering that VEGF is a direct target of miR-655, we hypothesised that the tumour-suppressive roles of miR-655 in EOC cells could be imitated by VEGF knockdown. To confirm this hypothesis, VEGF siRNA was transfected into SKOV3 and OVCAR3 cells to knock down VEGF expression. VEGF protein expression was silenced effectively in SKOV3 and OVCAR3 cells after transfection with VEGF siRNA (P<0.05; Fig. 4A). Similar to miR-655 restoration, VEGF knockdown retarded the proliferation (Fig. 4B, P<0.05) and invasion (P<0.05; Fig. 4C) of SKOV3 and OVCAR3 cells. These results further suggested that VEGF is a direct target of miR-655 in EOC.

VEGF reversed the miR-655 overexpression-induced suppression of cellular proliferation and invasion of EOC

To ascertain whether VEGF mediates the inhibitory roles of miR-655 overexpression in EOC cells, SKOV3 and OVCAR3 cells were simultaneously co-transfected with miR-655 mimic and VEGF restoration plasmid pcDNA3.1-VEGF or empty pcDNA3.1 plasmid. After transfection, Western blot analysis demonstrated that the downregulation of VEGF protein caused by miR-655 overexpression was recovered in SKOV3 and OVCAR3 cells after cotransfection with pcDNA3.1-VEGF (P<0.05; Fig. 5A). In addition, subsequent functional assays indicated that recovered VEGF expression could significantly eliminate the suppressive effects on proliferation (P<0.05; Fig. 5B) and invasion (P<0.05; Fig. 5C) of SKOV3 and OVCAR3 cells induced by miR-655 overexpression. These findings suggested that miR-655 inhibits EOC cell proliferation and invasion partly by downregulating VEGF expression.

Discussion

In recent years, miRNAs have been shown to be deregulated in EOC, and their deregulation is involved in EOC formation and progression by regulating the expression of numerous cancer-related genes (2527). Moreover, miRNAs have been recognised as promising prognosis biomarkers and effective therapeutic targets of EOC (28). Hence, it is of great importance to further determine the detailed roles and underlying mechanisms of miRNAs involved in EOC and to identify novel targets for diagnosis, prognosis and treatment of patients with EOC. Here, we found that miR-655 expression was significantly downregulated in EOC tissues and cell lines. Functional analysis revealed that the enforced expression of miR-655 significantly restricted EOC cell proliferation and invasion in vitro. Additionally, we identified VEGF as a direct target gene of miR-655 in EOC cells. Furthermore, inhibition of VEGF simulated the inhibitory effects of miR-655 overexpression in EOC cell proliferation and invasion. Moreover, recovered VEGF expression effectively counteracted the inhibitory effects on EOC cells due to miR-655 overexpression. Our findings suggested that miR-655 suppresses cell proliferation and invasion in EOC by directly targeting VEGF.

miR-655 is differentially expressed in multiple human cancer types. For instance, miR-655 is downregulated in hepatocellular carcinoma, and their downregulation is strongly correlated with tumour size, microvascular invasion, portal vein tumour thrombosis status, TNM stage and distant metastasis (14,15). Hepatocellular carcinoma patients with low miR-655 expression exhibits shorter survival periods than patients with high miR-655 levels. In addition, miR-655 is identified as an independent risk factor for patients with hepatocellular carcinoma (14). In oesophageal squamous cell carcinoma, miR-655 expression is low in tumour tissues, and this low miR-655 expression is significantly associated with lymph node metastases (16). Oesophageal squamous cell carcinoma patients with low miR-655 expression have poorer progression-free survival compared with patients with high miR-655 levels (17). In triple-negative breast cancer, miR-655 expression is reduced in tumour tissues and cell lines, and this decreased miR-655 expression is strongly correlated with the molecular-based classification and lymph node metastasis of triple-negative breast cancer (18). These findings suggested that miR-655 may be a valuable biomarker for the diagnosis and prognosis of these specific tumour types.

Dysregulation of miR-655 is closely associated with carcinogenesis and cancer progression of various human malignancies. For example, miR-655 overexpression restricts cell proliferation, migration, invasion and epithelial-to-mesenchymal transition of hepatocellular carcinoma by directly targeting ADAM10, ZEB1 and TGFBR2 and indirectly regulating the β-catenin pathway (15,29). Wang (16) and Chang (17) et al revealed that ectopic expression of miR-655 inhibits cell growth and metastasis in vitro by blocking PTTG1. Lv et al reported that upregulation of miR-655 significantly reduces cell migration, invasion and epithelial-to-mesenchymal transition of triple-negative breast cancer by regulating Prrx1 (18). Liang et al found that miR-655 re-expression restricts cell proliferation and motility and induces the apoptosis of pituitary tumour by regulating the p53/PTTG1 feedback loop (30). Zhang et al showed that restoration expression of miR-655 represses cell proliferation and invasion and induces the apoptosis of retinoblastoma by directly targeting PAX6 and suppressing the ERK and p38 MAPK signalling pathways (31). These findings suggested that miR-655 might be an attractive therapeutic target for patients with these cancer types.

Identification of the targets of miR-655 in EOC is vital for the development of effective therapeutic strategies for patients with this disease. In our study, VEGF, a 35–45 kD heparin-binding glycoprotein, was demonstrated to be a direct target gene of miR-655 in EOC. It is reported to be overexpressed in several types of human cancer, such as gastric cancer (32), retinoblastoma (33), hepatocellular carcinoma (34) and colorectal cancer (35). VEGF expression is also upregulated in EOC, and this upregulation is correlated with tumour grade and stage (20,36). EOC patients with high VEGF expression show shorter overall survival than those with low expression. VEGF expression is an independent prognostic factor in EOC patients (20,21). Aberrantly highly expressed VEGF is involved in EOC onset and development, and it regulates diverse biological processes, including cell proliferation, migration, invasion, apoptosis and angiogenesis (2124). Hence, VEGF knockdown using miR-655-based targeted therapy may be an effective therapeutic method for patients with EOC.

In conclusion, the present study showed that miR-655 was downregulated in EOC tissues and cell lines. In vitro functional experiments demonstrated that miR-655 inhibited cellular proliferation and invasion of EOC. Mechanistically, VEGF was validated as a direct target gene of miR-655 in EOC cells. The present results indicated that miR-655 may be a potential therapeutic target in EOC. However, we do not employ laser microdissection method to collect the adjacent non-neoplastic ovarian tissues. This is a limitation of our study.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

XZ and ZZ designed the work that led to the submission. ZZ and SY performed functional experiments. YC analysed the data obtained from this research. ZZ and SY drafted the manuscript.

Ethics approval and consent to participate

The present study was approved by the Research Ethics Committee of Yidu Central Hospital of Weifang (Yidu, China), and was performed in accordance with the Declaration of Helsinki and the guidelines of the Ethics Committee of Yidu Central Hospital of Weifang Hospital. Written informed consent was obtained from all patients for the use of their clinical tissues.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chornokur G, Amankwah EK, Schildkraut JM and Phelan CM: Global ovarian cancer health disparities. Gynecol Oncol. 129:258–264. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Zhang B, Cai FF and Zhong XY: An overview of biomarkers for the ovarian cancer diagnosis. Eur J Obstet Gynecol Reprod Biol. 158:119–123. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Suh DH, Kim JW, Kim K, Kim HJ and Lee KH: Major clinical research advances in gynecologic cancer in 2012. J Gynecol Oncol. 24:66–82. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2012. CA Cancer J Clin. 62:10–29. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Lengyel E: Ovarian cancer development and metastasis. Am J Pathol. 177:1053–1064. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Despierre E, Lambrechts D, Neven P, Amant F, Lambrechts S and Vergote I: The molecular genetic basis of ovarian cancer and its roadmap towards a better treatment. Gynecol Oncol. 117:358–365. 2010. View Article : Google Scholar : PubMed/NCBI

7 

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

8 

He L and Hannon GJ: MicroRNAs: Small RNAs with a big role in gene regulation. Nat Rev Genet. 5:522–531. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Zaman MS, Maher DM, Khan S, Jaggi M and Chauhan SC: Current status and implications of microRNAs in ovarian cancer diagnosis and therapy. J Ovarian Res. 5:442012. View Article : Google Scholar : PubMed/NCBI

10 

Alvarez-Garcia I and Miska EA: MicroRNA functions in animal development and human disease. Development. 132:4653–4662. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Katz B, Tropé CG, Reich R and Davidson B: MicroRNAs in ovarian cancer. Hum Pathol. 46:1245–1256. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Zhang S, Lu Z, Unruh AK, Ivan C, Baggerly KA, Calin GA, Li Z, Bast RC Jr and Le XF: Clinically relevant microRNAs in ovarian cancer. Mol Cancer Res. 13:393–401. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Lin S and Gregory RI: MicroRNA biogenesis pathways in cancer. Nat Rev Cancer. 15:321–333. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Zhao XQ, Liang B, Jiang K and Zhang HY: Down-regulation of miR-655-3p predicts worse clinical outcome in patients suffering from hepatocellular carcinoma. Eur Rev Med Pharmacol Sci. 21:748–752. 2017.PubMed/NCBI

15 

Wu G, Zheng K, Xia S, Wang Y, Meng X, Qin X and Cheng Y: MicroRNA-655-3p functions as a tumor suppressor by regulating ADAM10 and β-catenin pathway in hepatocellular carcinoma. J Exp Clin Cancer Res. 35:892016. View Article : Google Scholar : PubMed/NCBI

16 

Wang Y, Zang W, Du Y, Ma Y, Li M, Li P, Chen X, Wang T, Dong Z and Zhao G: Mir-655 up-regulation suppresses cell invasion by targeting pituitary tumor-transforming gene-1 in esophageal squamous cell carcinoma. J Transl Med. 11:3012013. View Article : Google Scholar : PubMed/NCBI

17 

Chang P, Wang X, Zhou Y and Hou Y: Analysis of the correlation between the expression of miR-655 and esophageal cancer prognosis. Oncol Lett. 13:4691–4694. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Lv ZD, Kong B, Liu XP, Jin LY, Dong Q, Li FN and Wang HB: miR-655 suppresses epithelial-to-mesenchymal transition by targeting Prrx1 in triple-negative breast cancer. J Cell Mol Med. 20:864–873. 2016. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Shen W, Li HL, Liu L and Cheng JX: Expression levels of PTEN, HIF-1α, and VEGF as prognostic factors in ovarian cancer. Eur Rev Med Pharmacol Sci. 21:2596–2603. 2017.PubMed/NCBI

21 

Whynott RM, Manahan P and Geisler JP: Vascular endothelial growth factor (VEGF) and cyclooxygenase 2 (COX 2) immunostaining in ovarian cancer. Eur J Gynaecol Oncol. 37:164–166. 2016.PubMed/NCBI

22 

Li J, Li L, Li Z, Gong G, Chen P, Liu H, Wang J, Liu Y and Wu X: The role of miR-205 in the VEGF-mediated promotion of human ovarian cancer cell invasion. Gynecol Oncol. 137:125–133. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Leng R, Zha L and Tang L: miR-718 represses VEGF and inhibits ovarian cancer cell progression. FEBS Lett. 588:2078–2086. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Chen Y, Zhang L, Liu WX and Wang K: VEGF and SEMA4D have synergistic effects on the promotion of angiogenesis in epithelial ovarian cancer. Cell Mol Biol Lett. 23:22018. View Article : Google Scholar : PubMed/NCBI

25 

Wang YQ, Guo RD, Guo RM, Sheng W and Yin LR: MicroRNA-182 promotes cell growth, invasion, and chemoresistance by targeting programmed cell death 4 (PDCD4) in human ovarian carcinomas. J Cell Biochem. 114:1464–1473. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Wu H, Xiao Z, Wang K, Liu W and Hao Q: miR-145 is downregulated in human ovarian cancer and modulates cell growth and invasion by targeting p70S6K1 and MUC1. Biochem Biophys Res Commun. 441:693–700. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Zhou F, Chen J and Wang H: MicroRNA-298 inhibits malignant phenotypes of epithelial ovarian cancer by regulating the expression of EZH2. Oncol Lett. 12:3926–3932. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Corney DC and Nikitin AY: MicroRNA and ovarian cancer. Histol Histopathol. 23:1161–1169. 2008.PubMed/NCBI

29 

Harazono Y, Muramatsu T, Endo H, Uzawa N, Kawano T, Harada K, Inazawa J and Kozaki K: miR-655 is an EMT-suppressive microRNA targeting ZEB1 and TGFBR2. PLoS One. 8:e627572013. View Article : Google Scholar : PubMed/NCBI

30 

Liang HQ, Wang RJ, Diao CF, Li JW, Su JL and Zhang S: The PTTG1-targeting miRNAs miR-329, miR-300, miR-381, and miR-655 inhibit pituitary tumor cell tumorigenesis and are involved in a p53/PTTG1 regulation feedback loop. Oncotarget. 6:29413–29427. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Zhang M, Li Q, Pan Y, Wang H, Liu G and Yin H: MicroRNA-655 attenuates the malignant biological behaviours of retinoblastoma cells by directly targeting PAX6 and suppressing the ERK and p38 MAPK signalling pathways. Oncol Rep. 39:2040–2050. 2018.PubMed/NCBI

32 

Ji YN, Wang Q, Li Y and Wang Z: Prognostic value of vascular endothelial growth factor A expression in gastric cancer: A meta-analysis. Tumour Biol. 35:2787–2793. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Youssef NS and Said AM: Immunohistochemical expression of CD117 and vascular endothelial growth factor in retinoblastoma: Possible targets of new therapies. Int J Clin Exp Pathol. 7:5725–5737. 2014.PubMed/NCBI

34 

Yamaguchi R, Yano H, Iemura A, Ogasawara S, Haramaki M and Kojiro M: Expression of vascular endothelial growth factor in human hepatocellular carcinoma. Hepatology. 28:68–77. 1998. View Article : Google Scholar : PubMed/NCBI

35 

Wen L, Wang R, Lu X and You C: Expression and clinical significance of vascular endothelial growth factor and fms-related tyrosine kinase 1 in colorectal cancer. Oncol Lett. 9:2414–2418. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Premalata CS, Umadevi K, Shobha K, Anurekha M and Krishnamoorthy L: Expression of VEGF-A in epithelial ovarian cancer: Correlation with morphologic types, grade and clinical stage. Gulf J Oncolog. 1:49–54. 2016.PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 18 Issue 2

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
Zhao Z, Yang S, Cheng Y and Zhao X: MicroRNA‑655 inhibits cell proliferation and invasion in epithelial ovarian cancer by directly targeting vascular endothelial growth factor Retraction in /10.3892/mmr.2022.12860. Mol Med Rep 18: 1878-1884, 2018
APA
Zhao, Z., Yang, S., Cheng, Y., & Zhao, X. (2018). MicroRNA‑655 inhibits cell proliferation and invasion in epithelial ovarian cancer by directly targeting vascular endothelial growth factor Retraction in /10.3892/mmr.2022.12860. Molecular Medicine Reports, 18, 1878-1884. https://doi.org/10.3892/mmr.2018.9090
MLA
Zhao, Z., Yang, S., Cheng, Y., Zhao, X."MicroRNA‑655 inhibits cell proliferation and invasion in epithelial ovarian cancer by directly targeting vascular endothelial growth factor Retraction in /10.3892/mmr.2022.12860". Molecular Medicine Reports 18.2 (2018): 1878-1884.
Chicago
Zhao, Z., Yang, S., Cheng, Y., Zhao, X."MicroRNA‑655 inhibits cell proliferation and invasion in epithelial ovarian cancer by directly targeting vascular endothelial growth factor Retraction in /10.3892/mmr.2022.12860". Molecular Medicine Reports 18, no. 2 (2018): 1878-1884. https://doi.org/10.3892/mmr.2018.9090