Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Endocrinol ; 28(5): 706-21, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24650175

RESUMO

Neuregulin 1 (NRG1) is induced in granulosa cells by LH and acts on granulosa and cumulus cells during ovulation. In this study, we sought to determine the role of NRG1 in oocyte maturation by generating a granulosa cell-specific Nrg1 knockout mouse (Nrg1(flox/flox);Cyp19a1Cre mice [gcNrg1KO]). In the gcNrg1KO mice, meiosis was induced 2 hours earlier than in control mice. More than 60% of the oocytes in the mutant mice spontaneously re-resumed meiosis beyond the MII stage. The percentage of successful fertilization was comparable in oocytes of both genotypes collected at 14 or 16 hours after human chorionic gonadotropin injection but was significantly lower in oocytes of the gcNrg1KO mice at 18 or 20 hours. The number of pups per litter was significantly decreased in gcNrg1KO mice. To determine the molecular events associated with the abnormal progression of meiosis in the gcNrg1KO mouse oocytes, the defects of cumulus/granulosa cell functions were analyzed. The expression of genes involved in luteinization and cumulus expansion was significantly higher at 2 hours after human chorionic gonadotropin injection in the gcNrg1KO mice; this was related to abnormal activation of protein kinase C (PKC) and phosphorylation of connexin-43 in cumulus cells. Changes in connexin-43 by PKC might lead to early meiotic resumption of oocytes in gcNrg1KO mice. We conclude that NRG1 is induced by LH in mural granulosa cells and exerts an important regulatory role in oocyte meiotic maturation and competence by reducing PKC activation in cumulus cells and preventing premature progression to the MII stage that leads to abnormal fertilization and fertility.


Assuntos
Células do Cúmulo/metabolismo , Neuregulina-1/genética , Oócitos/fisiologia , Animais , Células Cultivadas , Feminino , Fertilização , Junções Comunicantes/metabolismo , Humanos , Luteinização , Masculino , Meiose , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuregulina-1/metabolismo , Folículo Ovariano/citologia , Regiões Promotoras Genéticas , Proteína Quinase C/metabolismo , Ativação Transcricional
2.
Mol Endocrinol ; 25(1): 104-16, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21047912

RESUMO

Epidermal growth factor (EGF)-like factors [amphiregulin (AREG), betacellulin, and epiregulin] are induced by LH and activate the EGF receptor (ERBB1)/ERK1/2 pathway in granulosa cells and cumulus cells of preovulatory follicles to impact ovulation. However, the expression and roles of other ERBB family members and their ligands have not been explored in detail. Herein, we document that two transcripts of the neuregulin (Nrg1) gene are expressed in granulosa cells, and that the type III Nrg1 is induced during ovulation in an ERK1/2 and C/EBPß-dependent manner. Western blotting shows that intact (75 kDa) and secreted (45 kDa) forms of neuregulin 1 (NRG1) are present in the ovary. NRG1 likely binds to ERBB3/ERBB2 complexes that are expressed in granulosa cells and cumulus cells. In cultured granulosa cells, NRG1 selectively stimulates the phosphorylation of AKT/PKB compared to ERK1/2. However, when granulosa cells were cultured with NRG1 and AREG, the phosphorylation of ERK1/2 was markedly enhanced as compared with that by AREG alone. Cotreatment with NRG1 and AREG also increased progesterone production. When cumulus-oocyte complexes (COCs) were cultured with both NRG1 and AREG, the matured oocytes exhibited significantly higher developmental competence as compared with that of oocytes cultured with AREG alone. Collectively, these results document that the expression of type III NRG1 is induced in granulosa cells during ovulation and that NRG1 enhances AREG-induced ERK1/2 phosphorylation in both granulosa cells and cumulus cells. The NRG1 pathway has two roles: one is to enhance AREG-induced progesterone production in granulosa cells, and the other is to regulate oocyte maturation by a cumulus cell-dependent mechanism.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células da Granulosa/citologia , Hormônio Luteinizante/farmacologia , Neuregulina-1/genética , Oócitos/citologia , Anfirregulina , Animais , Sequência de Bases , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Células do Cúmulo/citologia , Células do Cúmulo/efeitos dos fármacos , Células do Cúmulo/enzimologia , Família de Proteínas EGF , Desenvolvimento Embrionário/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/farmacologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/enzimologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Ligantes , Camundongos , Dados de Sequência Molecular , Neuregulina-1/metabolismo , Neuregulina-1/farmacologia , Oócitos/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Fatores de Tempo
3.
Development ; 135(12): 2127-37, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18506027

RESUMO

Activation of the RAS family of small G-proteins is essential for follicle stimulating hormone-induced signaling events and the regulation of target genes in cultured granulosa cells. To analyze the functions of RAS protein in granulosa cells during ovarian follicular development in vivo, we generated conditional knock-in mouse models in which the granulosa cells express a constitutively active KrasG12D. The KrasG12D mutant mice were subfertile and exhibited signs of premature ovarian failure. The mutant ovaries contained numerous abnormal follicle-like structures that were devoid of mitotic and apoptotic cells and cells expressing granulosa cell-specific marker genes. Follicles that proceeded to the antral stage failed to ovulate and expressed reduced levels of ovulation-related genes. The human chorionic gonadotropin-stimulated phosphorylation of ERK1/2 was markedly reduced in mutant cells. Reduced ERK1/2 phosphorylation was due, in part, to increased expression of MKP3, an ERK1/2-specific phosphatase. By contrast, elevated levels of phospho-AKT were evident in granulosa cells of immature KrasG12D mice, even in the absence of hormone treatments, and were associated with the progressive decline of FOXO1 in the abnormal follicle-like structures. Thus, inappropriate activation of KRAS in granulosa cells blocks the granulosa cell differentiation pathway, leading to the persistence of abnormal non-mitotic, non-apoptotic cells rather than tumorigenic cells. Moreover, those follicles that reach the antral stage exhibit impaired responses to hormones, leading to ovulation failure. Transient but not sustained activation of RAS in granulosa cells is therefore crucial for directing normal follicle development and initiating the ovulation process.


Assuntos
Expressão Gênica/fisiologia , Células da Granulosa/metabolismo , Folículo Ovariano/fisiologia , Ovário/citologia , Ovulação , Proteínas ras/metabolismo , Animais , Células Cultivadas , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Expressão Gênica/genética , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Mutantes , Modelos Biológicos , Folículo Ovariano/anormalidades , Ovário/metabolismo , Proteínas ras/genética
4.
Reproduction ; 136(1): 9-21, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18456902

RESUMO

In this study, we collected follicular fluid, granulosa cells, and cumulus cells from antral follicles at specific time intervals following equine chorionic gonadotropin (eCG) and human chorionic gonadotropin (hCG) treatment of gilts. The treatment with eCG increased the production of estrogen coordinately with up-regulated proliferation of granulosa and cumulus cells. eCG also induced the expression of LHCGR and PGR in cumulus cells and progesterone accumulation was detected in follicular fluid prior to the LH/hCG surge. Moreover, progesterone and progesterone receptor (PGR) were critical for FSH-induced LHCGR expression in cumulus cells in culture. The expression of LHCGR mRNA in cumulus cells was associated with the ability of LH to induce prostaglandin production, release of epidermal growth factor (EGF)-like factors, and a disintegrin and metalloprotease with thrombospondin-like repeats 1 expression, promoting cumulus cell oocyte complexes (COCs) expansion and oocyte maturation. Based on the unique expression and regulation of PGR and LHCGR in cumulus cells, we designed a novel porcine COCs culture system in which hormones were added sequentially to mimic changes observed in vivo. Specifically, COCs from small antral follicles were pre-cultured with FSH and estradiol for 10 h at which time progesterone was added for another 10 h. After 20 h, COCs were moved to fresh medium containing LH, EGF, and progesterone. The oocytes matured in this revised COC culture system exhibited greater developmental competence to blastocyst stage. From these results, we conclude that to achieve optimal COC expansion and oocyte maturation in culture the unique gene expression patterns in cumulus cells of each species need to be characterized and used to increase the effectiveness of hormone stimulation.


Assuntos
Células do Cúmulo/metabolismo , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hormônio Luteinizante/farmacologia , Oogênese/genética , Suínos/metabolismo , Proteínas ADAM/genética , Proteína ADAMTS1 , Animais , Western Blotting/métodos , Técnicas de Cultura de Células , Gonadotropina Coriônica/farmacologia , Células do Cúmulo/efeitos dos fármacos , Primers do DNA/genética , Relação Dose-Resposta a Droga , Estradiol/análise , Estradiol/farmacologia , Feminino , Líquido Folicular/química , Expressão Gênica/efeitos dos fármacos , Gonadotropinas Equinas/farmacologia , Células da Granulosa/metabolismo , Oogênese/efeitos dos fármacos , Progesterona/análise , Progesterona/farmacologia , RNA Mensageiro/análise , Receptores do FSH/genética , Receptores do LH/genética , Receptores de Progesterona , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superovulação , Testosterona/análise
5.
Development ; 135(11): 2001-11, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18434414

RESUMO

The toll-like receptor (TLR) system is expressed in cumulus cells of ovulated cumulus-oocyte complexes (COCs) and is activated by bacterial lipopolysaccharides (LPS). However, the endogenous ligand(s) for the TLRs and the physiological role(s) in ovulated COCs remain to be defined. Based on reports that hyaluronan fragments can activate TLR2 and TLR4 in macrophages, and that ovulated COCs are characterized by a hyaluronan-rich matrix, we cultured ovulated mouse COCs with purified hyaluronan fragments, treated them with purified hyaluronidase or exposed them to sperm as a physiologically relevant source of hyaluronidase. Hyaluronan fragments or hyaluronidase activated the NFkappaB pathway and induced Il6, Ccl4 and Ccl5 mRNA expression within 2 hours. Anti-TLR2 and anti-TLR4 neutralizing antibodies significantly suppressed hyaluronan fragment- and hyaluronidase-induced activation of the NFkappaB pathway and the expression of these genes. When ovulated COCs were cultured with sperm, the expression and secretion of cytokine/chemokine family members were induced in a time-dependent manner that could be blocked by TLR2/TLR4 antibodies or by a hyaluronan-blocking peptide (Pep-1). The chemokines secreted from TLR2/TLR4-stimulated COCs activated cognate chemokine receptors (CCRs) localized on sperm and induced sperm protein tyrosine phosphorylation, which was used as an index of capacitation. Significantly, in vitro fertilization of COC-enclosed oocytes was reduced by the TLR2/TLR4 neutralizing antibodies or by Pep-1. From these results, we propose that TLR2 and TLR4 present on cumulus cells were activated by the co-culture with sperm in a hyaluronan fragment-dependent manner, and that chemokines secreted from COCs induced sperm capacitation and enhanced fertilization, providing evidence for a regulatory loop between sperm and COCs during fertilization.


Assuntos
Células do Cúmulo/efeitos dos fármacos , Citocinas/metabolismo , Ácido Hialurônico/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Western Blotting , Células Cultivadas , Quimiocinas/metabolismo , Células do Cúmulo/citologia , Células do Cúmulo/metabolismo , Feminino , Fertilização/efeitos dos fármacos , Imunofluorescência , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/metabolismo , Hialuronoglucosaminidase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Ovulação/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Espermatozoides/enzimologia , Espermatozoides/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...