Abstract

MicroRNAs are potent regulators of gene expression that have been widely implicated in reproduction and embryo development. Recent studies have demonstrated that miR-21, a microRNA extensively studied in the context of disease, is important in multiple facets of reproductive biology including folliculogenesis, ovulation, oocyte maturation and early mammalian development. Surprisingly, little is known about the mechanisms that regulate miR-21 and no studies have characterized these regulatory pathways in cumulus-oocyte complexes (COCs). We therefore investigated miR-21 in an in vitro model of bovine oocyte maturation. Levels of the primary transcript of miR-21 (pri-miR-21) and mature miR-21 increased markedly in COCs over the maturation period. Cloning of the bovine pri-miR-21 gene and promoter by 5′3′RACE (rapid amplification of cDNA ends) revealed a highly conserved region immediately upstream of the transcription start site and two alternatively-spliced variants of pri-miR-21. The promoter region contained several putative transcription factor binding sites, including two for signal transducer and activator of transcription 3 (STAT3). Mutation of these sites significantly decreased both the intrinsic activity of pri-miR-21 promoter-luciferase constructs and the response to leukemia inhibitory factor (LIF) (a STAT3 activator) in cultured MCF7 cells. In COCs, treatment with a STAT3 pathway inhibitor markedly decreased pri-miR-21 expression and prevented cumulus expansion. Pri-miR-21 expression was also inhibited by the protein synthesis inhibitor cycloheximide, suggesting that a protein ligand or signaling cofactor synthesized during maturation is necessary for transcription. Together these studies represent the first investigation of signaling pathways that directly influence miR-21 expression in bovine oocytes and cumulus cells.

Highlights

  • Oocyte maturation consists of nuclear and cytoplasmic events that are required for the oocyte to acquire competency for fertilization, and subsequent embryo development should fertilization occur

  • In order to confirm this presence of mature miR-21 in the somatic and oocyte compartments of the cumulus-oocyte complexes (COCs) after in vitro maturation, RNA in situ hybridization was performed with Locked Nucleic Acid (LNA) oligonucleotide probes specific for miR-21 or the small nuclear RNA U6, which was used as control (Fig. 1C). miR-21 and snRNA U6 are both detectable in the mature oocyte and its surrounding cells, in confirmation of the qRT-PCR results. miR-21 is strongly expressed in the cumulus compartment where it localizes to the cytoplasm as expected for a mature miRNA, while snRNA U6 is detected predominantly in cumulus cell nuclei

  • Based on the observation that signal transducer and activator of transcription 3 (STAT3) activation was robust after 6 hours of in vitro maturation (IVM), we evaluated pri-miR-21 transcript abundance in cumulus cells of COCs removed from culture at 7 hours of maturation under standard conditions, supplemented with 10 μM Stattic or dimethyl sulfoxide (DMSO) vehicle

Read more

Summary

Introduction

Oocyte maturation consists of nuclear and cytoplasmic events that are required for the oocyte to acquire competency for fertilization, and subsequent embryo development should fertilization occur. Comparative approaches, small RNA-sequencing and bioinformatics analysis have revealed that a substantial number of miRNA genes are expressed in bovine oocytes and their surrounding cumulus cells, a subset of which undergo dynamic changes in abundance over the course of in vitro maturation (IVM) and early embryonic development[26,27,28,29]. These profiles of global miRNA changes occurring in COCs have been useful for generating hypotheses regarding the potential regulatory function of miRNA during IVM, mechanistic studies that establish particular roles for specific miRNAs are limited. We document that miR-21 is induced under multiple IVM culture conditions after oocyte aspiration from follicles, and suggest that miR-21 is regulated by ligands synthesized and secreted by the COC itself

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