Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biomolecules ; 9(10)2019 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-31561560

RESUMO

In female mammals, germ cells enter meiosis in the fetal ovaries, while in males, meiosis is prevented until postnatal development. Retinoic acid (RA) is considered the main inducer of meiotic entry, as it stimulates Stra8 which is required for the mitotic/meiotic switch. In fetal testes, the RA-degrading enzyme CYP26B1 prevents meiosis initiation. However, the role of endogenous RA in female meiosis entry has never been demonstrated in vivo. In this study, we demonstrate that some effects of RA in mouse fetal gonads are not recapitulated by the invalidation or up-regulation of CYP26B1. In organ culture of fetal testes, RA stimulates testosterone production and inhibits Sertoli cell proliferation. In the ovaries, short-term inhibition of RA-signaling does not decrease Stra8 expression. We develop a gain-of-function model to express CYP26A1 or CYP26B1. Only CYP26B1 fully prevents STRA8 induction in female germ cells, confirming its role as part of the meiotic prevention machinery. CYP26A1, a very potent RA degrading enzyme, does not impair the formation of STRA8-positive cells, but decreases Stra8 transcription. Collectively, our data reveal that CYP26B1 has other activities apart from metabolizing RA in fetal gonads and suggest a role of endogenous RA in amplifying Stra8, rather than being the initial inducer of Stra8. These findings should reactivate the quest to identify meiotic preventing or inducing substances.


Assuntos
Gônadas/metabolismo , Ácido Retinoico 4 Hidroxilase/metabolismo , Tretinoína/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Gônadas/citologia , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Receptores do Ácido Retinoico/metabolismo , Testosterona/análise , Testosterona/biossíntese
2.
Cell Cycle ; 7(5): 656-64, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18256537

RESUMO

During mouse fetal development, meiosis is initiated in female germ cells only, with male germ cells undergoing mitotic arrest. Retinoic acid (RA) is degraded by Cyp26b1 in the embryonic testis but not in the ovary where it initiates the mitosis/meiosis transition. However the role of RA status in fetal germ cell proliferation has not been elucidated. As expected, using organ cultures, we observed that addition of RA in 11.5 days post-conception (dpc) testes induced Stra8 expression and meiosis. Surprisingly, in 13.5 dpc testes although RA induced Stra8 expression it did not promote meiosis. On 11.5 and 13.5 dpc, RA prevented male germ cell mitotic arrest through PI3K signaling. Therefore 13.5 dpc testes appeared as an interesting model to investigate RA effects on germ cell proliferation/differentiation independently of RA effect on the meiosis induction. At this stage, RA delayed SSEA-1 extinction, p63gamma expression and DNA hypermethylation which normally occur in male mitotic arrested germ cells. In vivo, in the fetal male gonad, germ cells cease their proliferation and loose SSEA-1 earlier than in female gonad and RA administration maintained male germ cell proliferation. Lastly, inhibition of endogenous Cyp26 activity in 13.5 dpc cultured testes also prevented male germ cell mitotic arrest. Our data demonstrate that the reduction of RA levels, which occurs specifically in the male fetal gonad and was known to block meiosis initiation, is also necessary to allow the establishment of the germ cell mitotic arrest and the correct further differentiation of the fetal germ cells along the male pathway.


Assuntos
Feto/citologia , Mitose/efeitos dos fármacos , Espermatozoides/citologia , Espermatozoides/efeitos dos fármacos , Testículo/citologia , Testículo/efeitos dos fármacos , Tretinoína/farmacologia , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/enzimologia , Feminino , Feto/efeitos dos fármacos , Feto/enzimologia , Masculino , Meiose/efeitos dos fármacos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Gravidez , Transdução de Sinais/efeitos dos fármacos , Espermatozoides/enzimologia , Testículo/enzimologia , Tretinoína/administração & dosagem
3.
Reproduction ; 135(1): 3-12, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18159078

RESUMO

Female fertility in mammals is determined by the pool of primordial follicles and low doses of radiation induce a major loss of primordial follicles in the ovary. We investigated the expression of p53 and its homologues, p63 and p73, in the normal and irradiated neonatal ovary. p63 was the only member of the p53 family detected in oocyte nucleus. No p63 transcripts or protein were detected in the early foetal ovary. p63 production began in late pachytene-stage oocytes and peaked in diplotene oocytes in mice and humans. The production of p63 was correlated with meiotic DNA double-strand break repair. Only transactivation (TA) isoforms were present in the ovary, with TAp63 alpha by far the most abundant in terms of mRNA and protein levels. Complete p63 null mutation did not affect normal ovary development. Irradiation rapidly triggered p63 phosphorylation. p63 null mutation prevented the cleavage of caspases-9 and -3 and the follicle loss induced by ionising radiation. Thus, our results evidence that irradiation-induced depletion of the primordial follicle pool results from the activation of p63 in quiescent oocytes.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Mutação , Oócitos/metabolismo , Ovário/embriologia , Ovário/efeitos da radiação , Transativadores/genética , Proteínas Supressoras de Tumor/genética , Animais , Apoptose , Western Blotting/métodos , Contagem de Células , Núcleo Celular/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Feminino , Genes p53 , Prófase Meiótica I , Camundongos , Camundongos Endogâmicos , Oócitos/citologia , Oócitos/efeitos da radiação , Técnicas de Cultura de Órgãos , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/metabolismo , Fatores de Transcrição , Proteínas Supressoras de Tumor/metabolismo , Irradiação Corporal Total
4.
J Cell Physiol ; 210(1): 87-98, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16998800

RESUMO

The fetal and neonatal development of male germ cells (gonocytes) is a poorly understood but crucial process for establishing fertility. In rodents, gonocytes go through two phases of proliferation accompanied by apoptosis and separated by a quiescent period during the end of fetal development. P63 is a member of the P53 gene family that yields six isoforms. We detected only the p63 protein and no p53 and p73 in the nucleus of the gonocytes of mouse testes. We report for the first time the ontogeny of each p63 mRNA isoform during testis development. We observed a strong expression of p63gamma mRNA and protein when gonocytes are in the quiescent period. In vitro treatment with retinoic acid prevented gonocytes from entering the quiescent period and was correlated with a reduced production of p63gamma isoform mRNA. We investigated the function of p63 by studying the testicular phenotype of P63-null mice. P63 invalidation slightly, but significantly increased the number of gonocytes counted during the quiescent period. As P63-null animals die at birth we used an original organ culture that mimicked neonatal in vivo development to study further the testicular development. P63 invalidation resulted in a sharply increased number of gonocytes during the culture period due to a decrease in spontaneous apoptosis with no change in proliferation. P63 invalidation also caused abnormal morphologies in the germ cells that were also found in P63(+/-) adult male mice. Thus, p63 appears as an important regulator of germ cell development.


Assuntos
Apoptose , Regulação da Expressão Gênica no Desenvolvimento , Fosfoproteínas/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Testículo/metabolismo , Transativadores/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Diferenciação Celular , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Idade Gestacional , Imuno-Histoquímica , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitose/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Técnicas de Cultura de Órgãos , Fosfoproteínas/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogênese/efeitos dos fármacos , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Testículo/citologia , Testículo/efeitos dos fármacos , Testículo/embriologia , Transativadores/genética , Tretinoína/farmacologia , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA