miR‑92b promotes autophagy and suppresses viability and invasion in breast cancer by targeting EZH2

2018 
MicroRNAs (miRs) are a small non-coding RNA family with a length of 18–22 nucleotides. They are able to regulate gene expression by either triggering target messenger RNA degradation or by inhibiting mRNA translation. Enhancer of zeste homolog 2 (EZH2) is the core enzymatic subunit of polycomb repressor complex 2 and is responsible for the trimethylation of histone 3 on lysine 27 (H3K27me3); it is also able to silence a bundle of tumor suppressor genes through promoter binding. However, little is known regarding the effect of miR-92b on cell autophagy, viability and invasion as well as how it interacts with EZH2. The present study investigated the major role of miR-92b in the autophagy, viability and invasion of breast cancer. It was revealed that in MCF-7 and MDA-MB-453 cells, the expression of miR-92b promoted autophagy induced by starvation and rapamycin treatment. The results of in vitro experiments results demonstrated that miR-92b inhibited breast cancer cell viability, invasion and migration. To further elucidate the regulatory mechanisms of miR-92b in autophagy, a dual luciferase reporter assay was performed to determine whether miR-92b targeted the EZH2 gene. The expression of miR-92b was negatively correlated to EZH2 mRNA expression in breast cancer. Depletion of EZH2 induced phenocopied effects on miR-92b overexpression, thereby demonstrating its importance in autophagy. These results indicated that miR-92b may serve an important role in breast cancer in controlling autophagy, viability and invasion. The present study indicated that miR-92b and EZH2 may serve as potential biomarkers for cancer detection and highlighted their possible therapeutic implications.
    • Correction
    • Source
    • Cite
    • Save
    • Machine Reading By IdeaReader
    43
    References
    23
    Citations
    NaN
    KQI
    []