Role of miR-34c microRNA in the late steps of spermatogenesis

  1. Jean-Pierre Rouault1
  1. 1Ecole Normale Supérieure de Lyon, Institut de Génomique Fonctionnelle de Lyon (IGFL), Université Lyon1, CNRS UMR 5242, INRA UMR1288, F-69364 Lyon, Cedex 07, France
  2. 2Faculté de Médecine, Institut de Génomique Fonctionnelle de Lyon (IGFL), Centre National de la Recherche Scientifique (CNRS) UMR5242, INRA UMR1288, Plateforme CLARA, 69495 Pierre-Bénite, France
  3. 3Institut des Sciences Pharmaceutiques et Biologiques (ISPB) de Lyon, Université Claude Bernard Lyon, 69622 Villeurbanne Cedex, France
  4. 4Groupe d’Étude de la Reproduction Chez l'Homme et les Mammifères (GERHM), Institut National de la Santé et de la Recherche Médicale (Inserm U625), Université Rennes 1, IFR140, Rennes, F-35042, France
  5. 5UMR-S625, Université Rennes1, Rennes, F-35042, France
  6. 6Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon1, 69622 Villeurbanne Cedex, France
  7. 7Unité Médicale d'Oncologie Moléculaire et Transfert (UMOMT), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon (HCL), 69495 Pierre-Bénite, France
  8. 8Centre National de la Recherche Scientifique (CNRS), UMR 6247, Institut National de la Santé et de la Recherche Médicale (Inserm), Université de Clermont, U931 Clermont-Ferrand, France
  • 9 Present address: Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London, United Kingdom.

Abstract

Spermatogenesis is a cyclic process in which diploid spermatogonia differentiate into haploid spermatozoa. This process is highly regulated, notably at the post-transcriptional level. MicroRNAs (miRNAs), single-stranded noncoding RNA molecules of about 20–25 nucleotides, are implicated in the regulation of many important biological pathways such as proliferation, apoptosis, and differentiation. We wondered whether miRNAs could play a role during spermatogenesis. The miRNA expression repertoire was tested in germ cells, and we present data showing that miR-34c was highly expressed only in these cells. Furthermore, our findings indicate that in male gonads, miR-34c expression is largely p53 independent in contrast to previous results showing a direct link in somatic cells between the miR-34 family and this tumor suppressor protein. In order to identify target genes involved in germinal lineage differentiation, we overexpressed miR-34c in HeLa cells, analyzed the transcriptome of these modified cells, and noticed a shift of the expression profile toward the germinal lineage. Recently, it has been shown that exogenous expression of Ddx4/Vasa in embryonic chicken stem cells (cESC) induces cESC reprogramming toward a germ cell fate. When we simultaneously expressed miR-34c in such cells, we could detect an up-regulation of germ cell-specific genes whereas the expression of other lineage specific markers remained unchanged. These data suggest that miR-34c could play a role by enhancing the germinal phenotype of cells already committed to this lineage.

Keywords:

Keywords

Footnotes

  • Reprint requests to: Jean-Pierre Rouault, Ecole Normale Supérieure de Lyon, Institut de Génomique Fonctionnelle de Lyon (IGFL), Université Lyon1, CNRS UMR 5242, INRA UMR1288, F-69364 Lyon, Cedex 07, France; e-mail: jean-pierre.rouault{at}ens-lyon.fr; fax: 33(0)472728992.

  • Article published online ahead of print. Article and publication date are at http://www.rnajournal.org/cgi/doi/10.1261/rna.1963810.

    • Received October 16, 2009.
    • Accepted December 23, 2009.
| Table of Contents