Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Biol ; 13: 39, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26076587

RESUMEN

BACKGROUND: The early stages of ovarian follicle formation-beginning with the breakdown of germ cell cysts and continuing with the formation of primordial follicles and transition to primary and secondary follicles-are critical in determining reproductive life span and fertility. Previously, we discovered that global knockouts of germ cell-specific transcriptional co-regulators Sohlh1, Sohlh2, Lhx8, and Nobox, cause rapid oocyte loss and ovarian failure. Also factors such as Nobox and Sohlh1 are associated with human premature ovarian failure. In this study, we developed a conditional knockout of Lhx8 to study oocyte-specific pathways in postnatal folliculogenesis. RESULTS: The conditional deficiency of Lhx8 in the oocytes of primordial follicles leads to massive primordial oocyte activation, in part, by indirectly interacting with the PI3K-AKT pathway, as shown by synergistic effects on FOXO3 nucleocytoplasmic translocation and rpS6 activation. However, LHX8 does not directly regulate members of the PI3K-AKT pathway; instead, we show that LHX8 represses Lin28a expression, a known regulator of mammalian metabolism and of the AKT/mTOR pathway. LHX8 can bind to the Lin28a promoter, and the depletion of Lin28a in Lhx8-deficient oocytes partially suppresses primordial oocyte activation. Moreover, unlike the PI3K-AKT pathway, LHX8 is critical beyond primordial follicle activation, and blocks the primary to secondary follicle transition. CONCLUSIONS: Our results indicate that the LHX8-LIN28A pathway is essential in the earliest stages of primordial follicle activation, and LHX8 is an important oocyte-specific transcription factor in the ovary for regulating postnatal folliculogenesis.


Asunto(s)
Proteínas con Homeodominio LIM/metabolismo , Oocitos/citología , Folículo Ovárico/fisiología , Folículo Ovárico/ultraestructura , Factores de Transcripción/metabolismo , Animales , Femenino , Proteínas con Homeodominio LIM/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Mapas de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Regulación hacia Arriba
2.
Mol Cell Endocrinol ; 356(1-2): 31-9, 2012 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-21856374

RESUMEN

Oocyte differentiation into a totipotent cell requires initial germ cell cyst breakdown to form primordial follicles, recruitment of primordial follicles for development into primary follicles and remarkable growth of the ovarian follicle which culminates in ovulation. During oogenesis, the oocyte undergoes dynamic alterations in gene expression which are regulated by a set of well-coordinated transcription factors active in the germ line and soma. A number of germ cell specific as well as somatic expressed transcriptional regulators are critical in ovarian formation and folliculogenesis. These transcriptional regulators include: Foxo3, Foxl2, Figla, Lhx8, Nobox, Sohlh1 and Sohlh2. A subset of these transcriptional regulators is mutated in women with ovarian insufficiency and infertility. Studies on transcriptional regulators preferentially expressed in the ovary are important to develop a better understanding of the mechanisms of activation and survival of ovarian follicles, as well as an understanding of ovary specific pathways that can be modulated in the future to regulate fertility and protect against external insults such as chemotherapy.


Asunto(s)
Oogénesis/genética , Factores de Transcripción/fisiología , Animales , Fenómenos Fisiológicos Celulares/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Folículo Ovárico/embriología , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Folículo Ovárico/fisiología
3.
Mol Cell Endocrinol ; 315(1-2): 1-10, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19643165

RESUMEN

Premature ovarian failure (POF) is a complex disorder that affects approximately 1% of women. POF is characterized by the depletion of functional ovarian follicles before the age of 40 years, and clinically, patients may present with primary amenorrhea or secondary amenorrhea. Although some genes have been hypothesized to be candidates responsible for POF, the etiology of most of the cases is idiopathic, with the underlying causes still unidentified because of the heterogeneity of the disease. In this review, we consider some mutant mouse models that exhibit phenotypes which are comparable to human POF, and we suggest that the use of these mouse models may help us to gain a better understanding of the molecular mechanisms underlying POF in humans.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Noqueados , Insuficiencia Ovárica Primaria , Animales , Femenino , Humanos , Ratones , Folículo Ovárico/anatomía & histología , Folículo Ovárico/fisiología , Fenotipo , Insuficiencia Ovárica Primaria/etiología , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/fisiopatología
4.
PLoS One ; 4(7): e6186, 2009 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-19587782

RESUMEN

Immature ovarian primordial follicles are essential for maintenance of the reproductive lifespan of female mammals. Recently, it was found that overactivation of the phosphatidylinositol 3-kinase (PI3K) signaling in oocytes of primordial follicles by an oocyte-specific deletion of Pten (phosphatase and tensin homolog deleted on chromosome ten), the gene encoding PI3K negative regulator PTEN, results in premature activation of the entire pool of primordial follicles, indicating that activation of the PI3K pathway in oocytes is important for control of follicular activation. To investigate whether PI3K signaling in oocytes of primary and further developed follicles also plays a role at later stages in follicular development and ovulation, we conditionally deleted the Pten gene from oocytes of primary and further developed follicles by using transgenic mice expressing zona pellucida 3 (Zp3) promoter-mediated Cre recombinase. Our results show that Pten was efficiently deleted from oocytes of primary and further developed follicles, as indicated by the elevated phosphorylation of the major PI3K downstream component Akt. However, follicular development was not altered and oocyte maturation was also normal, which led to normal fertility with unaltered litter size in the mutant mice. Our data indicate that properly controlled PTEN/PI3K-Akt signaling in oocytes is essential for control of the development of primordial follicles whereas overactivation of PI3K signaling in oocytes does not appear to affect the development of growing follicles. This suggests that there is a stage-specific function of PTEN/PI3K signaling in mouse oocytes that controls follicular activation.


Asunto(s)
Oocitos/metabolismo , Folículo Ovárico/fisiología , Fosfohidrolasa PTEN/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Animales , Femenino , Fertilidad , Fertilización , Eliminación de Gen , Meiosis , Ratones , Ratones Transgénicos , Ovulación , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo
5.
Science ; 319(5863): 611-3, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18239123

RESUMEN

In the mammalian ovary, progressive activation of primordial follicles from the dormant pool serves as the source of fertilizable ova. Menopause, or the end of female reproductive life, occurs when the primordial follicle pool is exhausted. However, the molecular mechanisms underlying follicle activation are poorly understood. We provide genetic evidence that in mice lacking PTEN (phosphatase and tensin homolog deleted on chromosome 10) in oocytes, a major negative regulator of phosphatidylinositol 3-kinase (PI3K), the entire primordial follicle pool becomes activated. Subsequently, all primordial follicles become depleted in early adulthood, causing premature ovarian failure (POF). Our results show that the mammalian oocyte serves as the headquarters of programming of follicle activation and that the oocyte PTEN-PI3K pathway governs follicle activation through control of initiation of oocyte growth.


Asunto(s)
Oocitos/fisiología , Folículo Ovárico/fisiología , Fosfohidrolasa PTEN/fisiología , Animales , Femenino , Atresia Folicular , Ratones , Ratones Transgénicos , Oocitos/citología , Oocitos/crecimiento & desarrollo , Tamaño de los Órganos , Folículo Ovárico/citología , Ovario/anatomía & histología , Ovario/fisiología , Ovulación , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Insuficiencia Ovárica Primaria/fisiopatología , Proteínas Quinasas/metabolismo , Proteína S6 Ribosómica/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR
6.
Mol Endocrinol ; 21(9): 2189-202, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17565040

RESUMEN

In humans, the molecular mechanisms underlying ovarian follicle endowment and activation, which are closely related to the control of female reproduction, occurrence of menopause, and related diseases such as premature ovarian failure, are poorly understood. In the current study, we provide several lines of genetic evidence that the cyclin-dependent kinase (Cdk) inhibitor 1B (commonly known as p27(kip1) or p27) controls ovarian development in mice by suppressing follicle endowment and activation, and by promoting follicle death. In p27-deficient (p27(-/-)) mice, postnatal follicle assembly was accelerated, and the number of endowed follicles was doubled as compared with p27(+/+) mice. Moreover, in p27(-/-) ovaries the primordial follicle pool was prematurely activated once it was endowed, and at the same time the massive follicular death that occurs before sexual maturity was rescued by loss of p27. In early adulthood, however, the overactivated follicular pool in p27(-/-) ovaries was largely depleted, causing premature ovarian failure. Furthermore, we have extensively studied the molecular mechanisms underlying the above-mentioned phenotypes seen in p27(-/-) ovaries and have found that p27 controls follicular development by several distinct mechanisms at different stages of development of the ovary. For example, p27 controls oocyte growth by suppressing the functions of Cdk2/Cdc2-cyclin A/E1 in oocytes that are arrested at the diplotene stage of meiosis I. This function of p27 is distinct from its well-known role as a suppressor of cell cycle progression. In addition, we have found that p27 activates the caspase-9-caspase-3-caspase-7-poly (ADP-ribose) polymeraseapoptotic cascade by inhibiting Cdk2/Cdc2-cyclin A/B1 kinase activities in follicles, thereby inducing follicle atresia. Our results suggest that the p27 gene is important in determining mammalian ovarian development. This study therefore provides insight into ovary-borne genetic aberrations that cause defects in folliculogenesis and infertility in humans.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Folículo Ovárico/metabolismo , Ovario/embriología , Ovario/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Conejos
7.
J Mol Endocrinol ; 38(1-2): 137-46, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17242176

RESUMEN

Communication between mammalian oocytes and their surrounding granulosa cells through the Kit-Kit ligand (KL, or stem cell factor, SCF) system has been shown to be crucial for follicular development. Our previous studies (Reddy et al. 2005, Liu et al. 2006) have indicated that the intra-oocyte KL-Kit-PI3 kinase (PI3K)-Akt-Foxo3a cascade may play an important role in follicular activation and early development. In the present study, using in situ hybridization and in vitro culture of growing oocytes from 8-day-old postnatal mice, we have demonstrated that another Akt substrate, glycogen synthase kinase-3 (GSK-3), is expressed in growing oocytes. Also, treatment of cultured mouse oocytes with soluble KL not only leads to increased Akt kinase activity in the oocytes, which can phosphorylate recombinant GSK-3 in vitro, but also leads to phosphorylation of oocyte GSK-3alpha and GSK-3beta, which can result in the inactivation of GSK-3 function in oocytes. In addition, we have shown that the regulation of GSK-3alpha and GSK-3beta in cultured oocytes by soluble KL is accomplished through PI3K, since the PI3K-specific inhibitor LY294002 completely abolished the KL-induced phosphorylation of GSK-3alpha and GSK-3beta. Moreover, blockage of the Kit signaling pathway by a Kit function-blocking antibody, ACK2, resulted in reduced phosphorylation of GSK-3. Taken together, our data suggest that the cascade from granulosa cell-derived KL to Kit-PI3K-Akt-GSK-3 in oocytes may take part in regulation of oocyte growth and early ovarian follicular development.


Asunto(s)
Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Oocitos/enzimología , Folículo Ovárico/enzimología , Factor de Células Madre/fisiología , Animales , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Proto-Oncogénicas c-akt/fisiología
8.
Development ; 134(1): 199-209, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17164425

RESUMEN

In recent years, mammalian oocytes have been proposed to have important roles in the orchestration of ovarian follicular development and fertility. To determine whether intra-oocyte Foxo3a, a component of the phosphatidylinositol 3-kinase (PI3K) signaling pathway, influences follicular development and female fertility, a transgenic mouse model was generated with constitutively active Foxo3a expressed in oocytes. We found that the female transgenic mice were infertile, which was caused by retarded oocyte growth and follicular development, and anovulation. Further mechanistic studies revealed that the constitutively active Foxo3a in oocytes caused a dramatic reduction in the expression of bone morphogenic protein 15 (Bmp15), connexin 37 and connexin 43, which are important molecules for the establishment of paracrine and gap junction communications in follicles. Foxo3a was also found to facilitate the nuclear localization of p27(kip1) in oocytes, a cyclin-dependent kinase (Cdk) inhibitor that may serve to inhibit oocyte growth. The results from the current study indicate that Foxo3a is an important intra-oocyte signaling molecule that negatively regulates oocyte growth and follicular development. Our study may therefore give some insight into oocyte-borne genetic aberrations that cause defects in follicular development and anovulation in human diseases, such as premature ovarian failure.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Infertilidad Femenina , Oocitos/metabolismo , Folículo Ovárico/embriología , Folículo Ovárico/fisiología , Animales , Femenino , Proteína Forkhead Box O3 , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Transgénicos
9.
Dev Biol ; 299(1): 1-11, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16970938

RESUMEN

A large amount of information has accumulated over the past decade on how gonadotropins, steroid hormones and growth factors regulate development of the mammalian ovarian follicle. Moreover, the bi-directional communication between mammalian oocytes and their surrounding somatic (granulosa) cells has also been shown to be crucial for this process. The intra-ovarian factors, or more specifically, the intra-oocyte signaling pathways that control oocyte growth and early follicular development are largely unknown, however. Based on both in vitro studies and in vivo functional studies using gene-modified mouse models, this review focuses on the key features of the phosphatidylinositol 3 kinase (PI3K) pathway in growing mouse oocytes and on the novel functions of the oocyte PI3K pathway in controlling mammalian oocyte growth and follicular development that have come to light only recently. We propose that the PI3K pathway in the oocyte, which is activated by granulosa cell-produced Kit ligand (KL) via the oocyte-surface receptor Kit, may serve as an intra-oocyte network that regulates both oocyte growth and the early development of ovarian follicles.


Asunto(s)
Oocitos/enzimología , Oocitos/crecimiento & desarrollo , Folículo Ovárico/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Femenino , Células de la Granulosa/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...