Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 102021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-34939930

RESUMEN

Gonadotropin-releasing hormone (GnRH) is the primary neuropeptide controlling reproduction in vertebrates. GnRH stimulates follicle-stimulating hormone (FSH) and luteinizing hormone (LH) synthesis via a G-protein-coupled receptor, GnRHR, in the pituitary gland. In mammals, GnRHR lacks a C-terminal cytosolic tail (Ctail) and does not exhibit homologous desensitization. This might be an evolutionary adaptation that enables LH surge generation and ovulation. To test this idea, we fused the chicken GnRHR Ctail to the endogenous murine GnRHR in a transgenic model. The LH surge was blunted, but not blocked in these mice. In contrast, they showed reductions in FSH production, ovarian follicle development, and fertility. Addition of the Ctail altered the nature of agonist-induced calcium signaling required for normal FSH production. The loss of the GnRHR Ctail during mammalian evolution is unlikely to have conferred a selective advantage by enabling the LH surge. The adaptive significance of this specialization remains to be determined.


Asunto(s)
Fertilidad , Hormona Luteinizante/metabolismo , Receptores LHRH/química , Receptores LHRH/fisiología , Animales , Pollos , Femenino , Hormona Folículo Estimulante/metabolismo , Ratones , Ratones Transgénicos , Folículo Ovárico/fisiología , Receptores Acoplados a Proteínas G/fisiología
2.
Endocrinology ; 160(7): 1561-1572, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30942852

RESUMEN

WNT signaling regulates a variety of ovarian processes, including follicle development, granulosa cell (GC) proliferation and differentiation, steroidogenesis, and ovulation. The secreted frizzled-related proteins (SFRPs) comprise a family of WNT signaling antagonists. Sfrp4 expression was previously reported to be induced in ovarian GCs and cumulus cells in vivo following human chorionic gonadotropin treatment, suggesting that it may play key roles in cumulus expansion, ovulation/luteinization, and corpus luteum (CL) function. In this study, we aimed to define the physiological roles of Sfrp4 in the ovary by gene targeting. Sfrp4-null female mice were found to produce larger litters than did their wild-type littermates. Although previous studies had suggested roles of Sfrp4 in luteal cell survival, no differences in CL formation, morphology, steroidogenesis, involution, or luteal cell apoptosis were found in Sfrp4-null mice. Likewise, cumulus expansion occurred normally in Sfrp4-null mice, with minimal changes in cumulus cell gene expression. Hyperfertility in the Sfrp4-null model was ultimately attributed to decreased antral follicle atresia, leading to an enhanced ovulatory rate. Increased expression of FSH- and LH-responsive genes was found in GCs from Sfrp4-null mice, and GCs isolated from Sfrp4-null mice were found to be hyperresponsive to FSH and LH in vitro. Although Sfrp2 was found to be overexpressed in the GCs of Sfrp4-null mice (suggesting a compensatory mechanism), Sfrp2-null mice had normal fertility and ovulatory rates, and Sfrp2/4 double knockout mice did not differ from Sfrp4-null mice. Taken together, our results suggest that SFRP4 acts to attenuate GC responsiveness to gonadotropins, thereby decreasing follicle survival, ovulatory rate, and fertility.


Asunto(s)
Fertilidad/fisiología , Folículo Ovárico/metabolismo , Ovulación/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Apoptosis/fisiología , Cuerpo Lúteo/metabolismo , Células del Cúmulo/metabolismo , Femenino , Hormona Folículo Estimulante/farmacología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Tamaño de la Camada , Hormona Luteinizante/farmacología , Ratones , Ratones Noqueados , Folículo Ovárico/crecimiento & desarrollo , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/fisiología
3.
FASEB J ; 30(4): 1534-47, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26667040

RESUMEN

Whereas the roles of the canonical wingless-type MMTV (mouse mammary tumor virus) integration site family (WNT) signaling pathway in the regulation of ovarian follicle growth and steroidogenesis are now established, noncanonical WNT signaling in the ovary has been largely overlooked. Noncanonical WNTs, including WNT5a and WNT11, are expressed in granulosa cells (GCs) and are differentially regulated throughout follicle development, but their physiologic roles remain unknown. Using conditional gene targeting, we found that GC-specific inactivation ofWnt5a(but notWnt11) results in the female subfertility associated with increased follicular atresia and decreased rates of ovulation. Microarray analyses have revealed that WNT5a acts to down-regulate the expression of FSH-responsive genesin vitro, and corresponding increases in the expression of these genes have been found in the GCs of conditional knockout mice. Unexpectedly, we found that WNT5a regulates its target genes not by signalingviathe WNT/Ca(2+)or planar cell polarity pathways, but rather by inhibiting the canonical pathway, causing both ß-catenin (CTNNB1) and cAMP responsive element binding (CREB) protein levels to decreaseviaa glycogen synthase kinase-3ß-dependent mechanism. We further found that WNT5a prevents follicle-stimulating hormone and luteinizing protein from up-regulating the CTNNB1 and CREB proteins and their target genes, indicating that WNT5a functions as a physiologic inhibitor of gonadotropin signaling. Together, these findings identify WNT5a as a key regulator of follicle development and gonadotropin responsiveness.-Abedini, A., Zamberlam, G., Lapointe, E., Tourigny, C., Boyer, A., Paquet, M., Hayashi, K., Honda, H., Kikuchi, A., Price, C., Boerboom, D. WNT5a is required for normal ovarian follicle development and antagonizes gonadotropin responsiveness in granulosa cells by suppressing canonical WNT signaling.


Asunto(s)
Gonadotropinas/farmacología , Células de la Granulosa/efectos de los fármacos , Folículo Ovárico/metabolismo , Proteínas Wnt/genética , Vía de Señalización Wnt/efectos de los fármacos , Animales , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Femenino , Células de la Granulosa/metabolismo , Immunoblotting , Inmunohistoquímica , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Folículo Ovárico/citología , Folículo Ovárico/crecimiento & desarrollo , Ovulación/efectos de los fármacos , Ovulación/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/genética , Proteína Wnt-5a
4.
Biol Reprod ; 90(6): 135, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24855100

RESUMEN

Recent evidence has suggested that vascular endothelial growth factor A (VEGFA) is an important regulator of ovarian follicle development and survival. Both LH and FSH regulate Vegfa expression in granulosa cells and signal via the transcription factor hypoxia inducible factor 1 (HIF1). To further study the mechanism of action of HIF1 in the regulation of Vegfa, we studied Vegfa(delta/delta) mice, which lack a hypoxia response element in the Vegfa promoter. Granulosa cells from Vegfa(delta/delta) mice failed to respond to FSH or LH with an increase in Vegfa mRNA expression in vitro, and granulosa cells isolated from eCG-treated immature Vegfa(delta/delta) mice had significantly lower Vegfa mRNA levels compared to controls. However, normal Vegfa mRNA levels were detected in the granulosa cells from immature Vegfa(delta/delta) mice following hCG treatment. Vegfa(delta/delta) females produced infrequent litters, and their pups died shortly after birth. Ovaries from Vegfa(delta/delta) mice were much smaller than controls and contained few antral follicles and corpora lutea. Antral follicles numbers were decreased by nearly 50% in ovaries from Vegfa(delta/delta) mice relative to controls, and 74% of antral follicles in Vegfa(delta/delta) ovaries were atretic. Serum progesterone levels in adult Vegfa(delta/delta) females were significantly lower, apparently reflecting reduced numbers of corpora lutea. This study demonstrates for the first time the requirement of HIF1 for FSH-regulated Vegfa expression in vivo and that HIF1 acts via a single hypoxia response element in the Vegfa promoter to exert its regulatory functions. Our findings also further define the physiological role of VEGFA in follicle development.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Folículo Ovárico/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Supervivencia Celular/fisiología , Células Cultivadas , Cuerpo Lúteo/fisiología , Femenino , Gonadotropinas/metabolismo , Células de la Granulosa/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Tamaño de los Órganos/fisiología , Folículo Ovárico/citología , Embarazo , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , Reproducción/fisiología , Factor A de Crecimiento Endotelial Vascular/genética
5.
Biol Reprod ; 87(5): 104, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22954793

RESUMEN

WNT4 is required for normal ovarian follicle development and female fertility in mice, but how its signal is transduced remains unknown. Fzd1 encodes a WNT receptor whose expression is markedly induced in both mural granulosa cells and cumulus cells during the preovulatory period, in a manner similar to Wnt4. To study the physiological roles of FZD1 in ovarian physiology and to determine whether it serves as receptor for WNT4, Fzd1-null mice were created by gene targeting. Whereas rare Fzd1(-/-) females were sterile because of uterine fibrosis and ovarian tubulostromal hyperplasia, most were subfertile, producing ≈1 fewer pup per litter on average relative to controls. Unlike WNT4-deficient mice, ovaries from Fzd1(-/-) mice had normal weights, numbers of follicles, steroid hormone production, and WNT4 target gene expression levels. Microarray analyses of granulosa cells from periovulatory follicles revealed few genes whose expression was altered in Fzd1(-/-) mice. However, gene expression analyses of cumulus-oocyte complexes (COCs) revealed a blunted response of both oocyte (Zp3, Dppa3, Nlrp5, and Bmp15) and cumulus (Btc, Ptgs2, Sema3a, Ptx3, Il6, Nts, Alcam, and Cspg2) genes to the ovulatory signal, whereas the expression of these genes was not altered in WNT4-deficient COCs from Wnt4(tm1.1Boer/tm1.1Boer);Tg (CYP19A1-cre)1Jri mice. Despite altered gene expression, cumulus expansion appeared normal in Fzd1(-/-) COCs both in vitro and in vivo. Together, these results indicate that Fzd1 is required for normal female fertility and may act in part to regulate oocyte maturation and cumulus cell function, but it is unlikely to function as the sole ovarian WNT4 receptor.


Asunto(s)
Células del Cúmulo/fisiología , Fertilidad/fisiología , Receptores Frizzled/fisiología , Regulación de la Expresión Génica/fisiología , Animales , Femenino , Receptores Frizzled/deficiencia , Receptores Frizzled/genética , Expresión Génica , Células de la Granulosa/metabolismo , Ratones , Ratones Noqueados , Análisis por Micromatrices , Folículo Ovárico/crecimiento & desarrollo , Ovario/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Vía de Señalización Wnt/fisiología
6.
Carcinogenesis ; 33(11): 2283-92, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22871496

RESUMEN

Few targeted therapies have been developed for ovarian granulosa cell tumor (GCT), even though it represents 5% of all malignant ovarian tumors in women. As misregulation of PI3K/AKT signaling has been implicated in GCT development, we hypothesized that the AKT signaling effector mammalian target of rapamycin (mTOR) may play a role in the pathogenesis of GCT and could represent a therapeutic target. Analyses of human GCT samples showed an increase in protein levels of mTOR and its downstream effectors RPS6KB1, RPS6, eIF4B and PPARG relative to normal granulosa cells, suggestive of an increase in mTOR pathway activity and increased translational activity and/or protein stability. We next sought to evaluate mTOR as a GCT therapeutic target using the Pten (tm1Hwu/tmiHwu);Ctnnb1 (tm1Mmt/+);Amhr2 (tm3(cre)Bhr/+) (PCA) mouse model, in which mTOR, RPS6KB1, eIF4B and PPARG are upregulated in tumor cells in a manner similar to human GCT. Treatment of PCA mice with the mTOR-specific inhibitor everolimus reduced tumor growth rate (1.5-fold; P < 0.05) and also reduced total tumor burden (4.7-fold; P < 0.05) and increased survival rate (78 versus 44% in the vehicle group) in a PCA surgical model of GCT peritoneal carcinomatosis. Everolimus decreased tumor cell proliferation and tumor cell volume relative to controls (P < 0.05), whereas apoptosis was unaffected. Phosphorylation of RPS6KB1 and RPS6 were decreased (P < 0.05) by everolimus, but RPS6KB1, RPS6, eIF4B and PPARG expressions were not affected. These results suggest that mTOR is a valid and clinically useful pharmacological target for the treatment of GCT, although its inhibition does not reverse all consequences of aberrant PI3K/AKT signaling in the PCA model.


Asunto(s)
Proliferación Celular , Tumor de Células de la Granulosa/prevención & control , Inmunosupresores/uso terapéutico , Neoplasias Peritoneales/prevención & control , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Adulto , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Everolimus , Femenino , Tumor de Células de la Granulosa/mortalidad , Tumor de Células de la Granulosa/patología , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Transgénicos , Fosfohidrolasa PTEN/fisiología , Neoplasias Peritoneales/mortalidad , Neoplasias Peritoneales/secundario , Fosforilación , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Sirolimus/uso terapéutico , Tasa de Supervivencia , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , beta Catenina/fisiología
7.
Dev Dyn ; 240(7): 1806-14, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21584905

RESUMEN

Adamts4 encodes a widely-expressed proteinase thought to be involved in processes ranging from cartilage metabolism to ovarian follicle development. To study its physiological roles, Adamts4-null mice were created by gene targeting. Unexpectedly, these were found to be phenotypically normal, suggesting that other gene(s) may compensate for its loss. Adamts4(-/-) mice were, therefore, crossed with a strain lacking Adamts1, whose pattern of expression and substrate specificity overlap that of Adamts4. Most (>95%) Adamts1(-/-) ;Adamts4(-/-) mice died within 72 hr after birth with a marked thinning of the renal medulla. The renal defect was not observed in embryonic Adamts1(-/-) ;Adamts4(-/-) kidneys, but became apparent around birth. The few (<5%) Adamts1(-/-) ;Adamts4(-/-) animals to reach adulthood had the same renal phenotype seen in newborns. This study is thus the first to report Adamts4 expression and function in the mammalian kidney, and to demonstrate that Adamts1 and Adamts4 play redundant and essential roles in perinatal kidney development.


Asunto(s)
Proteínas ADAM/metabolismo , Médula Renal/embriología , Médula Renal/metabolismo , Procolágeno N-Endopeptidasa/metabolismo , Proteínas ADAM/genética , Proteína ADAMTS1 , Proteína ADAMTS4 , Animales , Western Blotting , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Procolágeno N-Endopeptidasa/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Front Biosci (Schol Ed) ; 3(1): 276-85, 2011 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-21196376

RESUMEN

One of the major functions of the ovary is the biosynthesis of steroid hormones, which are essential for the development of secondary sexual characteristics at puberty, for subsequent ovarian function, and for the establishment and maintenance of pregnancy. Increases in our understanding of the molecular mechanisms governing the control of ovarian steroidogenesis have greatly improved our understanding of the female reproductive cycle, as well as the pathogenesis of reproductive disorders such as polycystic ovarian syndrome and premature ovarian failure. The pituitary gonadotropins follicle stimulating hormone (FSH) and luteinizing hormone (LH) are the main endocrine regulators of ovarian steroidogenesis, and act by directly or indirectly modulating the activity of a multitude of intracellular signaling pathways. The WNT/CTNNB1 pathway, which is now believed to be a significant contributor to the regulation of ovarian steroidogenesis, could be one of the pathways modulated by gonadotropin signaling. This review will focus on the emerging role of WNT/CTNNB1 signaling in the regulation of steroidogenesis, with emphasis on potential mechanisms of interaction with FSH/LH signaling in ovarian granulosa and luteal cells.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Hormonas Esteroides Gonadales/biosíntesis , Hormona Luteinizante/metabolismo , Folículo Ovárico/fisiología , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Femenino , Humanos , Proteína Oncogénica v-akt/metabolismo , Folículo Ovárico/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Embarazo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factor Esteroidogénico 1/metabolismo
9.
FASEB J ; 24(8): 3010-25, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20371632

RESUMEN

To study the role of WNT4 in the postnatal ovary, a mouse strain bearing a floxed Wnt4 allele was created and mated to the Amhr2(tm3(cre)Bhr) strain to target deletion of Wnt4 to granulosa cells. Wnt4(flox/-);Amhr2(tm3(cre)Bhr/+) mice had reduced ovary weights and produced smaller litters (P<0.05). Serial follicle counting demonstrated that Wnt4(flox/-);Amhr2(tm3(cre)Bhr/+) mice were born with a normal ovarian reserve and maintained normal numbers of small follicles until puberty but had only 25.2% of the normal number of healthy antral follicles. Some Wnt4(flox/-);Amhr2(tm3(cre)Bhr/+) mice had no antral follicles or corpora lutea and underwent premature follicle depletion. RT-PCR analyses of Wnt4(flox/-);Amhr2(tm3(cre)Bhr/+) granulosa cells and cultured granulosa cells that overexpress WNT4 demonstrated that WNT4 regulates the expression of Star, Cyp11a1, and Cyp19, steroidogenic genes previously identified as downstream targets of the WNT signaling effector CTNNB1. Decreased serum progesterone levels were found in immature, gonadotropin-treated Wnt4(flox/-);Amhr2(tm3(cre)Bhr/+) mice (P<0.05). WNT4- and CTNNB1-overexpressing cultured granulosa cells were analyzed by microarray for alterations in gene expression, which showed that WNT4 regulates additional genes involved in late follicle development via the WNT/CTNNB1 signaling pathway. Together, these data indicate that WNT4 is required for normal antral follicle development and may act by regulating granulosa cell functions including steroidogenesis.


Asunto(s)
Fertilidad , Regulación de la Expresión Génica , Folículo Ovárico , Proteínas Wnt/fisiología , Animales , Femenino , Perfilación de la Expresión Génica , Células de la Granulosa/citología , Ratones , Ratones Mutantes , Transducción de Señal , Esteroide Hidroxilasas/genética , Esteroides/biosíntesis , Proteína Wnt4 , beta Catenina/metabolismo
10.
Can J Physiol Pharmacol ; 82(3): 191-9, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15052285

RESUMEN

The efficacy of angiotensin-converting enzyme inhibitors (ACEIs) in the treatment of chronic aortic regurgitation (AR) is not well established and remains controversial. The mechanisms by which ACEIs may protect against left-ventricular (LV) volume overload are not well understood, and clinical trials performed until now have yielded conflicting results. This study was therefore performed to assess the effectiveness of two different doses of the ACEI captopril in a rat model of chronic AR. We compared the effects of a 6-month low-dose (LD) (25 mg/kg) or higher dose (HD) (75 mg/kg) treatment with captopril on LV function and hypertrophy in Wistar rats with severe AR. Untreated animals developed LV eccentric hypertrophy and systolic dysfunction. LD treatment did not prevent hypertrophy and provided modest protection against systolic dysfunction. HD treatment preserved LV systolic function and dimensions and tended to slow hypertrophy. The cardiac index remained high and similar among all AR groups, treated or not. Tissue renin-angiotensin system (RAS) analysis revealed that ACE activity was increased in the LVs of AR animals and that only HD treatment significantly decreased angiotensin II receptor mRNA levels. Fibronectin expression was increased in the LV or AR animals, but HD treatment almost completely reversed this increase. The ACE inhibitor captopril was effective at high doses in this model of severe AR. These effects might be related to the modulation of tissue RAS and the control of fibrosis.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Insuficiencia de la Válvula Aórtica/tratamiento farmacológico , Captopril/uso terapéutico , Cardiomiopatía Dilatada/prevención & control , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Insuficiencia de la Válvula Aórtica/complicaciones , Captopril/farmacología , Gasto Cardíaco/efectos de los fármacos , Gasto Cardíaco/fisiología , Cardiomiopatía Dilatada/complicaciones , Cardiomiopatía Dilatada/tratamiento farmacológico , Enfermedad Crónica , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Fibronectinas/efectos de los fármacos , Fibronectinas/genética , Fibronectinas/metabolismo , Expresión Génica , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Hipertrofia Ventricular Izquierda/genética , Masculino , ARN Mensajero/química , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Volumen Sistólico/efectos de los fármacos , Volumen Sistólico/fisiología , Factores de Tiempo , Función Ventricular Izquierda/efectos de los fármacos , Función Ventricular Izquierda/fisiología , Remodelación Ventricular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...