Abstract

The tumor suppressor gene, Ras-association domain family (RASSF)2A, is inactivated by promoter hypermethylation in many cancers. The current study was performed to evaluate the methylation status of RASSF2A in epithelial ovarian cancer (EOC) tissues and plasma, and correlations with gene expression and clinicopathologic characteristics. We detected methylation of the RASSF2A gene in tissues and corresponding plasma samples from 47 EOC patients and 14 patients with benign ovarian tumors and 10 with normal ovarian tissues. The methylation status was determined by methylation-specific PCR while gene expression of mRNA was examined by RT-PCR. The EOC cell line, SKOV3, was treated with 5-aza-2'-deoxycytidine (5-aza- dC). RASSF2A mRNA expression was significantly low in EOC tissues. The frequency of aberrant methylation of RASSF2A was 51.1% in EOC tissues and 36.2% in corresponding plasma samples, whereas such hypermethylation was not detected in the benign ovarial tumors and normal ovarian samples. The expression of RASSF2A mRNA was significantly down-regulated or lost in the methylated group compared to the unmethylated group (p<0.05). After treatment with 5-aza-dC, RASSF2A mRNA expression was significantly restored in the Skov3 cell line. Epigenetic inactivation of RASSF2A through aberrant promoter methylation may play an important role in the pathogenesis of EOC. Methylation of the RASSF2A gene in plasma may be a valuable molecular marker for the early detection of EOC.

Highlights

  • Epithelial ovarian cancer (EOC) is the third leading type of female gynecologic carcinomas, and has a high mortality rate (Siegel et al, 2012)

  • RASSF2A mRNA expression was absent in 42.6% (20/47) of epithelial ovarian cancer (EOC)

  • RASSF2A mRNA expression was significantly lower in EOCs compared with benign ovarian tumors or normal ovarian samples (p

Read more

Summary

Introduction

Epithelial ovarian cancer (EOC) is the third leading type of female gynecologic carcinomas, and has a high mortality rate (Siegel et al, 2012). It is increasingly recognized that both genetic and epigenetic events play a role in the development of ovarian cancer, with epigenetic changes occurring early in the carcinogenic process (Wei et al, 2006; Jones et al, 2007). There is evidence indicating that epigenetic silencing of gene expression by promoter hypermethylation is a major mechanism for the inactivation of tumor suppressor genes (TSGs) in cancer (Mutskov et al, 2002; Herman et al, 2003; Qiu H et al, 2011). Promoter hypermethylation is a common epigenetic mechanism for TSG inactivation in human cancer and a promising target for molecular detection (Jones et al, 1999)

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call