Abstract

Abstract MicroRNAs (miRNAs), a class of non-coding small RNA, regulate gene expression through base-pairing binding to the complementary sequence in the 3’ untranslated region (3’ UTR) of mRNA. miRNAs contribute to the timing of development, apoptosis, cell cycle progression, cellular proliferation, stem cell self-renewal, cancer initiation and metastasis. The expression of miRNA is regulated during cell-cycle transition and cellular contact in part via active degradation. Aberrant expression of miRNAs or mutations of miRNA genes have been described in many types of tumors, including mammary tumors. The RNase III endoribonuclease Dicer cleaves long double-stranded RNA (dsRNA) or stem-loop-stem structured pre-miRNA to form mature miRNAs. RNAi-mediated knock-down of Dicer in human cells led to defects in both miRNA production and shRNA-mediated RNAi. Initially cloned as a breakpoint rearrangement in parathyroid adenoma, the cyclin D1 gene encodes the regulatory subunit of the holoenzyme that phosphorylates and inactivates both the pRb tumor suppressor and the key inducer of mitochondrial biogenesis NRF-1. In addition, a DNA bound form of cyclin D1 regulates gene expression. Cyclin D1 expression is induced during mammary gland and retinal differentiation, and deletion of the murine cyclin D1 gene resulted in failed terminal alveolar breast bud development and retinal degeneration. Diverse biological functions regulated by cyclin D1 include the induction of cellular proliferation, angiogenesis, cellular migration, DNA damage repair, mitochondrial biogenesis, stem cell maintenance, and miRNA expression. Cyclin D1 was shown to regulate the miR-17/20 locus and found to bind the miR-17/20 regulatory region. In order to determine further the mechanism by which cyclin D1 regulates non-coding RNA, we conducted studies of miRNA processing. We established cyclin D1-/- mouse embryonic fibroblasts cells (MEFs) and cyclin D1 knockdown (KD) MCF-7 human breast cancer cells. miRNA analysis indicated an induction of mature miRNA expression in cyclin D1 overexpressing cells. Analysis of the miRNA processing regulators demonstrated the selective induction of Dicer expression by cyclin D1. In cyclin D1-/- cells the reduction of Dicer abundance was accompanied by impairment of pre-miRNA to mature miRNA processing, which was restored with cyclin D1 rescue. Transient transgenic expression of cyclin D1 in the mouse mammary gland, or sustained transgenic expression of cyclin D1 induced mouse mammary gland tumors, recapitulated the induction of Dicer expression. Cyclin D1 and Dicer expression were correlated in luminal A and basal-like human breast cancer. Cyclin D1 regulation of cellular proliferation and migration was dependent upon Dicer. By demonstrating cyclin D1 induced Dicer abundance and function in tissue culture and in vivo, we provide evidence for novel crosstalk between the cell-cycle and non-coding miRNA biogenesis. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P4-07-05.

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