[HTML][HTML] Suppressing miR-199a-3p by promoter methylation contributes to tumor aggressiveness and cisplatin resistance of ovarian cancer through promoting DDR1 …

Y Deng, F Zhao, L Hui, X Li, D Zhang, W Lin… - Journal of ovarian …, 2017 - Springer
Y Deng, F Zhao, L Hui, X Li, D Zhang, W Lin, Z Chen, Y Ning
Journal of ovarian research, 2017Springer
Abstract Background Discoidin Domain Receptor 1 (DDR1) belongs to the family of collagen
receptor tyrosine kinases that confers the progression of various cancers. Aberrant
expression of DDR1 was detected in several human cancers including ovarian cancer,
which had been shown to increase the migration and invasion of tumor cells. However, the
precise mechanisms underlying the abnormal expression of DDR1 in ovarian cancer has
not been well investigated in previous studies. Results In this work, a negative correlation …
Background
Discoidin Domain Receptor 1 (DDR1) belongs to the family of collagen receptor tyrosine kinases that confers the progression of various cancers. Aberrant expression of DDR1 was detected in several human cancers including ovarian cancer, which had been shown to increase the migration and invasion of tumor cells. However, the precise mechanisms underlying the abnormal expression of DDR1 in ovarian cancer has not been well investigated in previous studies.
Results
In this work, a negative correlation between DDR1 and a tumor suppressor miRNA, miR-199a-3p, was observed in ovarian cancer tissues. Furthermore, in vitro experimental results confirmed that miR-199a-3p decreased the expression of DDR1 via targeting the 3’UTR of DDR1 mRNA. To explore the mechanisms for miR-199a-3p silence in ovarian cancer, the methylation status of the miR-199a promoter was analyzed in ovarian epithelial or cancer cells by methylation-specific PCR and bisulphite sequencing. As expected, the miR-199a promoter was hypermethylated in ovarian cancer cells but not in normal ovarianepithelial cells. Interestingly, knockdown of DNA methyltransferase 3A (DNMT3A) notably increased miR-199a-3p level and then attenuated the expression of DDR1 in ovarian cancer cells, which suggested that DNMT3A was responsible for the miR-199a promoter hypermethylation. Phenotype experiments showed that overexpression of miR-199a-3p significantly impaired the migratory, invasive, and tumorigenic capabilities of ovarian cancer cells as well as enhanced cisplatin resistance through inhibiting DDR1 expression.
Conclusion
These findings demonstrate a critical role of miR-199a-3p/DDR1 pathway in ovarian cancer development.
Springer