Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Hum Mol Genet ; 22(2): 345-57, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23065701

RESUMEN

We discovered a new spontaneous mutant allele of Npr2 named peewee (pwe) that exhibits severe disproportionate dwarfism and female infertility. The pwe phenotype is caused by a four base-pair deletion in exon 3 that generates a premature stop codon at codon 313 (L313X). The Npr2(pwe/pwe) mouse is a model for the human skeletal dysplasia acromesomelic dysplasia, Maroteaux type (AMDM). We conducted a thorough analysis of the female reproductive tract and report that the primary cause of Npr2(pwe/pwe) female infertility is premature oocyte meiotic resumption, while the pituitary and uterus appear to be normal. Npr2 is expressed in chondrocytes and osteoblasts. We determined that the loss of Npr2 causes a reduction in the hypertrophic and proliferative zones of the growth plate, but mineralization of skeletal elements is normal. Mutant tibiae have increased levels of the activated form of ERK1/2, consistent with the idea that natriuretic peptide receptor type 2 (NPR2) signaling inhibits the activation of the MEK/ERK mitogen activated protein kinase pathway. Treatment of fetal tibiae explants with mitogen activated protein kinase 1 and 2 inhibitors U0126 and PD325901 rescues the Npr2(pwe/pwe) growth defect, providing a promising foundation for skeletal dysplasia therapeutics.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Mutación , Receptores del Factor Natriurético Atrial/genética , Reproducción/genética , Animales , Secuencia de Bases , Densidad Ósea/genética , Enfermedades del Desarrollo Óseo/tratamiento farmacológico , Huesos/metabolismo , Enanismo/genética , Femenino , Genotipo , Humanos , Infertilidad Femenina/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fenotipo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología
2.
Biol Reprod ; 82(6): 1129-38, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20130263

RESUMEN

The insulin-like growth factor (IGF) system plays an important role in regulating ovarian follicular development and steroidogenesis. IGF binding proteins (IGFBP) mostly inhibit IGF actions, and IGFBP proteolysis is a major mechanism for regulating IGF bioavailability. Pregnancy-associated plasma protein-A (PAPPA) is a secreted metalloprotease responsible for cleavage of IGFBP4 in the ovary. The aim of this study was to investigate whether PAPPA plays a role in regulating ovarian functions and female fertility by comparing the reproductive phenotype of wild-type (WT) mice with mice heterozygous or homozygous for a targeted Pappa gene deletion (heterozygous and PAPP-A knockout [KO] mice, respectively). When mated with WT males, PAPP-A KO females demonstrated an overall reduction in average litter size. PAPP-A KO mice had a reduced number of ovulated oocytes, lower serum estradiol levels following equine chorionic gonadotropin administration, lower serum progesterone levels after human chorionic gonadotropin injection, and reduced expression of ovarian steroidogenic enzyme genes, compared to WT controls. In PAPP-A KO mice, inhibitory IGFBP2, IGFBP3, and IGFBP4 ovarian gene expression was reduced postgonadotropin stimulation, suggesting some compensation within the ovarian IGF system. Expression levels of follicle-stimulating hormone receptor, luteinizing hormone receptor, and genes required for cumulus expansion were not affected. Analysis of preovulatory follicular fluid showed complete loss of IGFBP4 proteolytic activity in PAPP-A KO mice, demonstrating no compensation for loss of PAPPA proteolytic activity by other IGFBP proteases in vivo in the mouse ovary. Taken together, these data demonstrate an important role of PAPPA in modulating ovarian function and female fertility by control of the bioavailability of ovarian IGF.


Asunto(s)
Hormonas Esteroides Gonadales/biosíntesis , Infertilidad Femenina/enzimología , Ovario/enzimología , Proteína Plasmática A Asociada al Embarazo/fisiología , Animales , Gonadotropina Coriónica/administración & dosificación , Endopeptidasas/análisis , Estradiol/biosíntesis , Estradiol/sangre , Femenino , Tamaño de la Camada , Ratones , Ratones Noqueados , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Ovario/efectos de los fármacos , Proteína Plasmática A Asociada al Embarazo/genética , Progesterona/biosíntesis , Progesterona/sangre , Receptores de HFE/análisis , Receptores de HL/análisis
3.
Mol Cell Biol ; 27(5): 1914-24, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17194751

RESUMEN

In the preovulatory ovarian follicle, mammalian oocytes are maintained in prophase meiotic arrest until the luteinizing hormone (LH) surge induces reentry into the first meiotic division. Dramatic changes in the somatic cells surrounding the oocytes and in the follicular wall are also induced by LH and are necessary for ovulation. Here, we provide genetic evidence that LH-dependent transactivation of the epidermal growth factor receptor (EGFR) is indispensable for oocyte reentry into the meiotic cell cycle, for the synthesis of the extracellular matrix surrounding the oocyte that causes cumulus expansion, and for follicle rupture in vivo. Mice deficient in either amphiregulin or epiregulin, two EGFR ligands, display delayed or reduced oocyte maturation and cumulus expansion. In compound-mutant mice in which loss of one EGFR ligand is associated with decreased signaling from a hypomorphic allele of the EGFR, LH no longer signals oocyte meiotic resumption. Moreover, induction of genes involved in cumulus expansion and follicle rupture is compromised in these mice, resulting in impaired ovulation. Thus, these studies demonstrate that LH induction of epidermal growth factor-like growth factors and EGFR transactivation are essential for the regulation of a critical physiological process such as ovulation and provide new strategies for manipulation of fertility.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Hormona Luteinizante/farmacología , Ovulación/fisiología , Animales , Western Blotting , Células Cultivadas , Gonadotropina Coriónica/farmacología , Factor de Crecimiento Epidérmico/genética , Femenino , Inmunoprecipitación , Hibridación in Situ , Hormona Luteinizante/metabolismo , Meiosis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Oocitos/citología , Oocitos/metabolismo , Folículo Ovárico/citología , Ovario/citología , Transducción de Señal , Factores de Tiempo
4.
Biol Reprod ; 81(3): 595-604, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19474061

RESUMEN

It is well established that cAMP signaling is an important regulator of the oocyte meiotic cell cycle. Conversely, the function of cGMP during oocyte maturation is less clear. Herein, we evaluated the expression of cGMP-hydrolyzing phosphodiesterases (PDEs) in the somatic and germ cell compartments of the mouse ovarian follicle and demonstrate that PDE5 is preferentially expressed in somatic cells. Cyclic GMP is a potent inhibitor of cAMP hydrolysis from oocyte extracts, with a 50% inhibitory concentration of 97 nM. Luteinizing hormone (LH) stimulation of cultured preovulatory follicles results in a marked decrease in cGMP content, and a nadir is reached in 1.5 h; similarly, oocyte cGMP levels decrease after gonadotropin stimulation in vivo. The LH-dependent decrease in cGMP requires activation of the epidermal growth factor network. Treatment of follicles with a PDE5 inhibitor increases cGMP in the follicle well above unstimulated levels. Although LH causes a decrease in cGMP in follicles preincubated with PDE5 inhibitors, the levels of this nucleotide remain above unstimulated levels. Under these conditions of elevated cGMP, LH stimulation does not cause oocyte maturation after 5 h of incubation. Microinjection of a cGMP-specific PDE into oocytes causes meiotic maturation of wild-type oocytes, suggesting that an intraoocyte pool of cGMP is involved in the maintenance of meiotic arrest. This effect is absent in PDE3A-deficient oocytes. Taken together, these findings provide evidence that cGMP and cAMP signaling cooperate in maintaining meiotic arrest via regulation of PDE3A and that a decrease in cGMP in the somatic compartment is one of the signals contributing to meiotic maturation.


Asunto(s)
GMP Cíclico/fisiología , Hormona Luteinizante/farmacología , Meiosis/efectos de los fármacos , Oocitos/efectos de los fármacos , Animales , Células Cultivadas , GMP Cíclico/metabolismo , GMP Cíclico/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Factores de Crecimiento Endotelial/metabolismo , Factores de Crecimiento Endotelial/fisiología , Femenino , Meiosis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/metabolismo , Oocitos/fisiología , Oogénesis/efectos de los fármacos , Oogénesis/genética , Folículo Ovárico/citología , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Ovulación/metabolismo , Ovulación/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología
5.
Mol Endocrinol ; 22(4): 924-36, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18187604

RESUMEN

LH activates a cascade of signaling events that are propagated throughout the ovarian preovulatory follicle to promote ovulation of a mature egg. Critical to LH-induced ovulation is the induction of epidermal growth factor (EGF)-like growth factors and transactivation of EGF receptor (EGFR) signaling. Because the timing of this transactivation has not been well characterized, we investigated the dynamics of LH regulation of the EGF network in cultured follicles. Preovulatory follicles were cultured with or without recombinant LH and/or specific inhibitors. EGFR and MAPK phosphorylation were examined by immunoprecipitation and Western blot analyses. By semiquantitative RT-PCR, increases in amphiregulin and epiregulin mRNAs were detected 30 min after recombinant LH stimulation of follicles and were maximal after 2 h. LH-induced EGFR phosphorylation also increased after 30 min and reached a maximum at 2 h. EGFR activation precedes oocyte maturation and is cAMP dependent, because forskolin similarly activated EGFR. LH-induced EGFR phosphorylation was sensitive to AG1478, an EGFR kinase inhibitor, and to inhibitors of matrix metalloproteases GM6001 and TNFalpha protease inhibitor-1 (TAPI-1), suggesting the involvement of EGF-like growth factor shedding. LH- but not amphiregulin-induced oocyte maturation and EGFR phosphorylation were sensitive to protein synthesis inhibition. When granulosa cells were cultured with a combination of neutralizing antibodies against amphiregulin, epiregulin, and betacellulin, EGFR phosphorylation and MAPK activation were inhibited. In cultured follicles, LH-induced MAPK activation was partially inhibited by AG1478 and GM6001, indicating that this pathway is regulated in part by the EGF network but also involves additional pathways. Thus, complex mechanisms are involved in the rapid amplification and propagation of the LH signal within preovulatory follicles and include the early activation of the EGF network.


Asunto(s)
Receptores ErbB/metabolismo , Hormona Luteinizante/farmacología , Folículo Ovárico/metabolismo , Anfirregulina , Animales , Western Blotting , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dipéptidos/farmacología , Familia de Proteínas EGF , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/fisiología , Epirregulina , Receptores ErbB/genética , Receptores ErbB/fisiología , Femenino , Glicoproteínas/genética , Ácidos Hidroxámicos/farmacología , Inmunohistoquímica , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intercelular/genética , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Oocitos/efectos de los fármacos , Oocitos/crecimiento & desarrollo , Folículo Ovárico/efectos de los fármacos , Fosforilación/efectos de los fármacos , Biosíntesis de Proteínas/genética , Biosíntesis de Proteínas/fisiología , Quinazolinas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Tirfostinos/farmacología
6.
Mol Endocrinol ; 20(4): 715-23, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16051667

RESUMEN

The LH surge causes major remodeling of the ovarian follicle in preparation for the ovulatory process. These changes include reprogramming of granulosa cells to differentiate into luteal cells, changes in cumulus cell secretory properties, and oocyte maturation. This review summarizes published data in support of the concept that LH stimulation of ovarian follicles involves activation of a local epidermal growth factor (EGF) network. A model describing this property of LH signaling and its branching to other signaling modules is discussed. According to this model, LH activation of mural granulosa cells stimulates cAMP signaling, which, in turn, induces the expression of the EGF-like growth factors epiregulin, amphiregulin, and betacellulin. These growth factors function by activating EGF receptors in either an autocrine/juxtacrine fashion within the mural layer, or they diffuse to act on cumulus cells. Activation of EGF receptor signaling in cumulus cells, together with cAMP priming, triggers oocyte nuclear maturation and acquisition of developmental competence as well as cumulus expansion. This model has important implications for ovarian physiology and for the development of new strategies for the pharmacological control of ovulation and for gamete maturation in vitro.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Folículo Ovárico/fisiología , Animales , AMP Cíclico/fisiología , Factor de Crecimiento Epidérmico/genética , Receptores ErbB/fisiología , Femenino , Expresión Génica , Células de la Granulosa/fisiología , Humanos , Hormona Luteinizante/fisiología , Modelos Biológicos , Ovulación/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal
7.
Cancer Res ; 65(20): 9206-15, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16230381

RESUMEN

Misregulation of the Wnt/beta-catenin signaling pathway is a hallmark of several forms of cancer. Components of the Wnt/beta-catenin pathway are expressed in ovarian granulosa cells; nevertheless, its potential involvement in granulosa cell tumorigenesis has not been examined. To this end, human (n = 6) and equine (n = 18) granulosa cell tumors (GCT) were analyzed for beta-catenin expression by immunohistochemistry. Unlike granulosa cells of normal ovaries, most (15 of 24) GCT samples showed nuclear localization of beta-catenin, suggesting that activation of the Wnt/beta-catenin pathway plays a role in the etiology of GCT. To confirm this hypothesis, Catnb(flox(ex3)/+); Amhr2(cre/+) mice that express a dominant stable beta-catenin mutant in their granulosa cells were generated. These mice developed follicle-like structures containing disorganized, pleiomorphic granulosa by 6 weeks of age. Even in older mice, these follicle-like lesions grew no larger than the size of antral follicles and contained very few proliferating cells. Similar to corpora lutea, the lesions were highly vascularized, although they did not express the luteinization marker Cyp11a1. Catnb(flox(ex3)/+); Amhr2(cre/+) females were also found to be severely subfertile, and fewer corpora lutea were found to form in response to exogenous gonadotropin compared with control mice. In older mice, the ovarian lesions often evolved into GCT, indicating that they represent a pretumoral intermediate stage. The GCT in Catnb(flox(ex3)/+); Amhr2(cre/+) mice featured many histopathologic similarities to the human disease, and prevalence of tumor development attained 57% at 7.5 months of age. Together, these studies show a causal link between misregulated Wnt/beta-catenin signaling and GCT development and provide a novel model system for the study of GCT biology.


Asunto(s)
Tumor de Células de la Granulosa/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas Wnt/biosíntesis , beta Catenina/biosíntesis , Animales , Modelos Animales de Enfermedad , Femenino , Tumor de Células de la Granulosa/genética , Tumor de Células de la Granulosa/patología , Caballos , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta , Transducción de Señal , Proteínas Wnt/genética , beta Catenina/genética
8.
Trends Endocrinol Metab ; 16(7): 320-6, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16054836

RESUMEN

The epidermal growth factor (EGF) network is composed of a complex array of growth factors synthesized as precursors and expressed on the cell surface. These latent growth factors are activated by cleavage and shedding from the cell surface and act by binding to various homo- and hetero-dimers of the EGF receptors (ErbBs). Although the exact molecular steps are poorly understood, ligand binding to G-protein-coupled receptors as diverse as the beta-adrenoceptors or the lysophosphatidic acid receptors leads to shedding of EGF growth factors and activation of EGF receptors. Recent observations from the pituitary and in the ovary are providing new insight into the role of this network in endocrine systems.


Asunto(s)
Sistema Endocrino/fisiología , Receptores ErbB/fisiología , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/fisiología , Animales , Humanos , Reproducción/fisiología
9.
Endocrinology ; 143(3): 898-908, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11861511

RESUMEN

Wnt ligands and Frizzled (Fz) G protein-coupled receptors impact cell fate, including embryonic development of the ovary. Because the role of these regulatory molecules during follicular development in the adult is not known, an RT-PCR survey was done. Wnt-4, Fz-4, and Fz-1 were among the transcripts detected, and each exhibited a specific pattern of expression. Fz-1 mRNA was low in preovulatory follicles of PMSG-treated mice but was increased within 4-12 h after an ovulatory surge of human CG. By in situ analysis, Fz-1 transcripts increased first in the theca cells and then in the granulosa cells of ovulating follicles but were low in corpora lutea. In contrast, Wnt-4, a critical factor in early ovarian development, was expressed in small preantral follicles. In addition, Wnt-4 was detected in preovulatory follicles and exhibited high levels in corpora lutea. A potential receptor for Wnt-4 in corpora lutea is Fz-4 that was also elevated in this tissue. Although Wnt-4 has been shown to function downstream of the PR in other tissues, Wnt-4 was not altered in follicles of PR-null mice that fail to ovulate. Rather expression of Fz-1 was lower in ovaries of PR knockout mice, compared with normal littermates. Thus, specific Wnt/Fz are expressed at distinct stages of follicular development, suggesting multiple functions for this signaling pathway in the ovary.


Asunto(s)
Folículo Ovárico/fisiología , Ovario/metabolismo , Biosíntesis de Proteínas , Proteínas/genética , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Receptores de Neurotransmisores/biosíntesis , Receptores de Neurotransmisores/genética , Proteínas de Pez Cebra , Animales , Western Blotting , Cuerpo Lúteo/metabolismo , Cartilla de ADN , Femenino , Receptores Frizzled , Células de la Granulosa/metabolismo , Hibridación in Situ , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/biosíntesis , ARN Mensajero/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular , Receptores Acoplados a Proteínas G , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Proteínas Wnt
10.
Endocrinology ; 144(3): 1020-31, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12586779

RESUMEN

During ovulation, production of a specialized hyaluronan (HA)-rich matrix cross-linked by associated HA binding factors causes expansion of the cumulus oocyte complex. Versican is a member of the hyalectan family that binds HA, provides structure and elasticity to tissues, and impacts cell motility and adhesion. In these studies, we sought to determine whether versican is synthesized by ovulating follicles and localizes along with HA in the expanded cumulus oocyte complex matrix in rodent ovaries and whether its expression and/or localization is altered in anovulatory mutant mice. Analysis of mRNA and protein identified isoforms V0, V1, and V3 versican in mouse and rat ovaries throughout follicular development. In situ hybridization localized versican mRNA most specifically to the granulosa cells. Expression was not significantly altered by estradiol or FSH treatment but was increased up to 10-fold during the periovulatory period after human chorionic gonadotropin treatment. In cultured granulosa cells, forskolin and phorbol 12 myristate 13-acetate or FSH + testosterone increased expression of versican. Immunohistochemical analyses verified versican protein in ovulating follicles localized to the expanded cumulus matrix as well as adjacent to the basement membrane. After ovulation, versican was localized around newly formed corpora lutea and vasculature. Unexpectedly, immunohistochemical analyses also demonstrated versican protein on granulosa cells in early primary and small antral follicles. Versican expression and localization were not altered in progesterone receptor or cyclooxygenase-2 null mice, suggesting that transcription of the versican gene is not a target of these two ovulatory mediators. These observations suggest that versican (V0, V1, and V3) is a matrix component of the granulosa layer throughout folliculogenesis and is enriched in remodeling matrices during ovulation and neovascularization of the corpora lutea.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato/análisis , Proteoglicanos Tipo Condroitín Sulfato/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hormonas/farmacología , Ovario/química , Animales , Células Cultivadas , Gonadotropina Coriónica/farmacología , Colforsina/farmacología , Ciclooxigenasa 2 , Estradiol/farmacología , Femenino , Hormona Folículo Estimulante/farmacología , Células de la Granulosa/química , Hibridación in Situ , Isoenzimas/deficiencia , Lectinas Tipo C , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Folículo Ovárico/crecimiento & desarrollo , Ovulación , Prostaglandina-Endoperóxido Sintasas/deficiencia , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Receptores de Progesterona/deficiencia , Transducción de Señal , Testosterona/farmacología , Acetato de Tetradecanoilforbol/farmacología , Versicanos
11.
Endocrinology ; 144(10): 4597-606, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12960062

RESUMEN

Secreted frizzled-related protein-4 (sFRP-4) belongs to a family of soluble proteins that have a Frizzled-like cysteine-rich domain and function as modulators of Wnt-Frizzled (Fz) signals. As several Wnts and Fz are expressed at defined stages of follicular development in rodent ovaries, these studies were undertaken to evaluate the hormone-regulated expression and localization of sFRP-4. In the mouse ovary, the expression of sFRP-4 mRNA was up-regulated in granulosa cells of large antral follicles after human chorionic gonadotropin administration and was also elevated in corpora lutea, as determined by RT-PCR and in situ hybridization analyses. In hypophysectomized rat ovaries, sFRP-4 expression was similarly induced by human chorionic gonadotropin and further up-regulated by PRL. PRL also stimulated the secretion of sFRP-4 protein from luteinized rat granulosa cells in culture. Therefore, regulation of sFRP-4 by LH and PRL may be important for modulating Fz-1, which is known to be expressed in periovulatory follicles, and Wnt-4/Fz-4, which are expressed in corpora lutea.


Asunto(s)
Ovario/metabolismo , Proteínas/metabolismo , Animales , Células Cultivadas , Gonadotropina Coriónica/farmacología , Cuerpo Lúteo/metabolismo , Cuerpo Lúteo/fisiología , Estradiol/farmacología , Femenino , Gonadotropinas Equinas/farmacología , Células de la Granulosa/metabolismo , Células de la Granulosa/fisiología , Hipofisectomía , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Folículo Ovárico/metabolismo , Ovulación/fisiología , Embarazo , Prolactina/farmacología , Proteínas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Progesterona/deficiencia , Distribución Tisular , Regulación hacia Arriba
12.
Mol Cell Endocrinol ; 356(1-2): 65-73, 2012 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-22101318

RESUMEN

During the peri-ovulatory period, the gonadotropin LH triggers major changes in both the somatic and germ cell compartments of the ovarian follicle. The oocyte completes the meiotic cell cycle to become a fertilizable egg, and dramatic changes in gene expression and secretion take place in the somatic compartment of the follicle in preparation for follicular rupture and oocyte release. The concerted changes are regulated by activation of intracellular signaling pathways as well as paracrine and autocrine regulatory loops. This review will provide a summary of the current knowledge of the molecular events triggered by LH focusing mostly on the signaling pathways required for oocyte maturation.


Asunto(s)
Oocitos/fisiología , Ovario/metabolismo , Ovulación/metabolismo , Transducción de Señal , Animales , Femenino , Uniones Comunicantes/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hormona Luteinizante/metabolismo , Hormona Luteinizante/fisiología , Meiosis , Nucleótidos Cíclicos/metabolismo , Nucleótidos Cíclicos/fisiología , Oocitos/metabolismo , Ovario/citología , Receptores Acoplados a Proteínas G/metabolismo
13.
PLoS One ; 6(6): e21574, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21738714

RESUMEN

Recent evidence that luteinizing hormone (LH) stimulation of ovulatory follicles causes transactivation of the epidermal growth factor receptor (EGFR) has provided insights into the mechanisms of ovulation. However, the complete array of signals that promote oocyte reentry into the meiotic cell cycle in the follicle are still incompletely understood. To elucidate the signaling downstream of EGFR involved in oocyte maturation, we have investigated the LH responses in granulosa cells with targeted ablation of EGFR. Oocyte maturation and ovulation is disrupted when EGFR expression is progressively reduced. In granulosa cells from mice with either global or granulosa cell-specific disruption of EGFR signaling, LH-induced phosphorylation of MAPK3/1, p38MAPK, and connexin-43 is impaired. Although the LH-induced decrease in cGMP is EGFR-dependent in wild type follicles, LH still induces a decrease in cGMP in Egfr(delta/f) Cyp19-Cre follicles. Thus compensatory mechanisms appear activated in the mutant. Spatial propagation of the LH signal in the follicle also is dependent on the EGF network, and likely is important for the control of signaling to the oocyte. Thus, multiple signals and redundant pathways contribute to regulating oocyte reentry into the cell cycle.


Asunto(s)
Receptores ErbB/metabolismo , Hormona Luteinizante/farmacología , Oocitos/efectos de los fármacos , Animales , Western Blotting , Células Cultivadas , Receptores ErbB/genética , Femenino , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Oocitos/citología , Oocitos/metabolismo , Oogénesis/efectos de los fármacos , Folículo Ovárico/citología
14.
Semin Reprod Med ; 27(1): 52-61, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19197805

RESUMEN

The growth and maturation of the ovarian follicle requires the coordinate function of somatic cells and the oocyte. Over the past three decades, numerous growth factors involved in the bidirectional signals between the somatic and germ cells have been identified. A possible function of epidermal growth factor (EGF) signaling at selected stages of follicle maturation had been proposed early on and is supported by many observations of in vitro effects of this growth factor on steroidogenesis, oocyte maturation, and cumulus expansion. However, attempts to link EGF levels in the follicular fluid with the state of follicle and oocyte maturation have been inconclusive. More recently, data generated using mouse genetic models perturbing ovulation and fertility indicate that EGF-like growth factors, rather than EGF itself, accumulate in the follicle at the time of ovulation. EGF-like growth factor mRNA is regulated by the luteinizing hormone surge, and corresponding proteins are detected in the follicle. The EGF-like growth factors amphiregulin, epiregulin, and betacellulin are potent stimulators of oocyte maturation and cumulus expansion, and perturbation of this EGF network in vivo impairs ovulation. Similar findings in species other than the mouse confirm an important physiological role for this network at the time of ovulation. Whether this network also plays a critical role in humans and whether it can be used as a biological marker of follicle development or for the improvement of fertility remains to be determined. This review summarizes the most recent findings on the EGF network during ovulation and the potential clinical applications of manipulating this intercellular communication pathway in the control of fertility.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Líquido Folicular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Oocitos/crecimiento & desarrollo , Oocitos/fisiología , Animales , Proliferación Celular , Células del Cúmulo/metabolismo , Células del Cúmulo/fisiología , Factor de Crecimiento Epidérmico/química , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Modelos Biológicos , Oogénesis/genética , Oogénesis/fisiología , Ovulación/genética , Ovulación/fisiología , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador alfa/metabolismo , Factor de Crecimiento Transformador alfa/fisiología
15.
Biol Reprod ; 73(6): 1135-46, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16093361

RESUMEN

Previous studies showed that transcripts encoding specific Wnt ligands and Frizzled receptors including Wnt4, Frizzled1 (Fzd1), and Frizzled4 (Fzd4) were expressed in a cell-specific manner in the adult mouse ovary. Overlapping expression of Wnt4 and Fzd4 mRNA in small follicles and corpora lutea led us to hypothesize that the infertility of mice null for Fzd4 (Fzd4-/-) might involve impaired follicular growth or corpus luteum formation. Analyses at defined stages of reproductive function indicate that immature Fzd4-/- mouse ovaries contain follicles at many stages of development and respond to exogenous hormone treatments in a manner similar to their wild-type littermates, indicating that the processes controlling follicular development and follicular cell responses to gonadotropins are intact. Adult Fzd4-/- mice also exhibit normal mating behavior and ovulate, indicating that endocrine events controlling these processes occur. However, Fzd4-/- mice fail to become pregnant and do not produce offspring. Histological and functional analyses of ovaries from timed mating pairs at Days 1.5-7.5 postcoitus (p.c.) indicate that the corpora lutea of the Fzd4-/- mice do not develop normally. Expression of luteal cell-specific mRNAs (Lhcgr, Prlr, Cyp11a1 and Sfrp4) is reduced, luteal cell morphology is altered, and markers of angiogenesis and vascular formation (Efnb1, Efnb2, Ephb4, Vegfa, Vegfc) are low in the Fzd4-/- mice. Although a recently identified, high-affinity FZD4 ligand Norrin (Norrie disease pseudoglioma homolog) is expressed in the ovary, adult Ndph-/- mice contain functional corpora lutea and do not phenocopy Fzd4-/- mice. Thus, Fzd4 appears to impact the formation of the corpus luteum by mechanisms that more closely phenocopy Prlr null mice.


Asunto(s)
Cuerpo Lúteo/fisiopatología , Infertilidad Femenina/genética , Receptores de Superficie Celular/genética , Animales , Biomarcadores/análisis , Vasos Sanguíneos/patología , Cuerpo Lúteo/patología , Implantación del Embrión , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Femenino , Receptores Frizzled , Regulación del Desarrollo de la Expresión Génica , Infertilidad Femenina/fisiopatología , Masculino , Ratones , Ratones Mutantes , Neovascularización Fisiológica/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Folículo Ovárico/fisiología , Ovario/irrigación sanguínea , Ovario/patología , Ovulación/genética , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G , Receptores de Prolactina/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/genética
16.
Recent Prog Horm Res ; 57: 195-220, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12017544

RESUMEN

The interactions of peptide and steroid hormone signaling cascades in the ovary are critical for follicular growth, ovulation, and luteinization. Although the pituitary gonadotropins follicle-stimulating hormone (FSH) and luteinizing hormone (LH) play key regulatory roles, their actions are also dependent on other peptide signaling pathways, including those stimulated by insulin-like growth factor-1 (IGF-1), transforming growth factor-beta (TGF-beta) family members (e.g., inhibin, activin, growth differentiation factor-9, bone morphogenic proteins), fibroblast growth factor, and Wnts (via Frizzled receptors). Each of these factors is expressed and acts in a cell-specific manner at defined stages of follicular growth. IGF-1, estrogen, and FSH comprise one major regulatory system. The Wnt/Frizzled pathways define other aspects relating to ovarian embryogenesis and possibly ovulation and luteinization. Likewise, the steroid receptors as well as orphan nuclear receptors and their ligands impact ovarian cell function. The importance of these multiple signaling cascades has been documented by targeted deletion of specific genes. For example, mice null for the LH-induced genes progesterone receptor (PR) and cyclo-oxygenase-2 (COX-2) fail to ovulate. Whereas PR appears to regulate the induction of novel proteases, COX-2 appears to regulate cumulus expansion. This review summarizes some new aspects of peptide and steroid hormone signaling in the rodent ovary.


Asunto(s)
Cuerpo Lúteo/fisiología , Folículo Ovárico/crecimiento & desarrollo , Ovario/crecimiento & desarrollo , Ovulación , Transducción de Señal , Animales , Estradiol/fisiología , Femenino , Hormona Folículo Estimulante/fisiología , Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/fisiología , Ovulación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA