Abstract

BackgroundThough androgen deprivation therapy is the standard treatment for prostate cancer (PCa), most patients would inevitably progress to castration-resistant prostate cancer (CRPC) which is the main cause of PCa death. Therefore, the identification of novel molecular mechanism regulating cancer progression and achievement of new insight into target therapy would be necessary for improving the benefits of PCa patients. This study aims to study the function and regulatory mechanism of HOTAIR/EZH2/miR-193a feedback loop in PCa progression.MethodsMSKCC and TCGA datasets were used to identify miR-193a expression profile in PCa. Cell Counting Kit-8 (CCK-8) assays, colony formation, invasion, migration, flow cytometry, a xenograft model and Gene Set Enrichment Analysis were used to detect and analyze the biological function of miR-193a. Then, we assessed the role of HOTAIR and EZH2 in regulation of miR-193a expression by using plasmid, lentivirus and small interfering RNA (siRNA). Luciferase reporter assays and chromatin immunoprecipitation assays were performed to detect the transcriptional activation of miR-193a by EZH2 and HOTAIR. Further, qRT-PCR and luciferase reporter assays were conducted to examine the regulatory role of miR-193a controlling the HOTAIR expression in PCa. Finally, the correlation between HOTAIR, EZH2 and miR-193a expression were analyzed using In situ hybridization and immunohistochemistry.ResultsWe found that miR-193a was significantly downregulated in metastatic PCa through mining MSKCC and TCGA datasets. In vitro studies revealed that miR-193a inhibited PCa cell growth, suppressed migration and invasion, and promoted apoptosis; in vivo results demonstrated that overexpression of miR-193a mediated by lentivirus dramatically reduced PCa xenograft tumor growth. Importantly, we found EZH2 coupled with HOTAIR to repress miR-193a expression through trimethylation of H3K27 at miR-193a promoter in PC3 and DU145 cells. Interestingly, further evidence illustrated that miR-193a directly targets HOTAIR showing as significantly reduced HOTAIR level in miR-193a overexpressed cells and tissues. The expression level of miR-193a was inversely associated with that of HOTAIR and EZH2 in PCa.ConclusionThis study firstly demonstrated that miR-193a acted as tumor suppressor in CRPC and the autoregulatory feedback loop of HOTAIR/EZH2/miR-193a served an important mechanism in PCa development. Targeting this aberrantly activated feedback loop may provide a potential therapeutic strategy.

Highlights

  • Though androgen deprivation therapy is the standard treatment for prostate cancer (PCa), most patients would inevitably progress to castration-resistant prostate cancer (CRPC) which is the main cause of PCa death

  • Experimental scheme and miR-193a expression correlates with prostate cancer clinical features In our previous microarray analysis [16], we have detected a total of 452 miRNAs that were differentially expressed between CRPC and Androgen dependent prostate cancer (ADPC)

  • We discovered that miR-193a levels were significantly downregulated in metastatic prostate cancer via reanalysis of Memorial Sloan Kettering Cancer Center (MSKCC) and The Cancer Genome Atlas (TCGA) datasets, and its expression was negatively correlated with the advanced stages and high gleason scores of PCa patients

Read more

Summary

Introduction

Though androgen deprivation therapy is the standard treatment for prostate cancer (PCa), most patients would inevitably progress to castration-resistant prostate cancer (CRPC) which is the main cause of PCa death. The biggest challenge for PCa treatment is that most patients would inevitably progress to castration-resistant prostate cancer within 2 years’ androgen deprivation therapy, which is considered as main cause of prostate cancer patient death. Given its dysregulation in PCa and its oncogenic role in PCa cells proliferation and metastasis, substantial efforts have been dedicated to identify its underlying molecular regulatory mechanisms and its potential therapy application in PCa. EZH2 is one of the core enzymatic subunit of histone methyltransferase polycomb repressor complex 2 (PRC2) which methylates lysine of histone H3 (H3K27) to promote transcriptional silencing of many tumor suppressive genes [5, 6]. The exact functional role of EZH2-mediated epigenetic silencing of tumor suppressive miRNAs in prostate cancer progression has not been systematically studied

Objectives
Methods
Results
Discussion
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