Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Biol Chem ; 288(4): 2641-54, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23233674

RESUMEN

In mammals, the receptor of the neuropeptide gonadotropin-releasing hormone (GnRHR) is unique among the G protein-coupled receptor (GPCR) family because it lacks the carboxyl-terminal tail involved in GPCR desensitization. Therefore, mechanisms involved in the regulation of GnRHR signaling are currently poorly known. Here, using immunoprecipitation and GST pull-down experiments, we demonstrated that SET interacts with GnRHR and targets the first and third intracellular loops. We delineated, by site-directed mutagenesis, SET binding sites to the basic amino acids (66)KRKK(69) and (246)RK(247), located next to sequences required for receptor signaling. The impact of SET on GnRHR signaling was assessed by decreasing endogenous expression of SET with siRNA in gonadotrope cells. Using cAMP and calcium biosensors in gonadotrope living cells, we showed that SET knockdown specifically decreases GnRHR-mediated mobilization of intracellular cAMP, whereas it increases its intracellular calcium signaling. This suggests that SET influences signal transfer between GnRHR and G proteins to enhance GnRHR signaling to cAMP. Accordingly, complexing endogenous SET by introduction of the first intracellular loop of GnRHR in αT3-1 cells significantly reduced GnRHR activation of the cAMP pathway. Furthermore, decreasing SET expression prevented cAMP-mediated GnRH stimulation of Gnrhr promoter activity, highlighting a role of SET in gonadotropin-releasing hormone regulation of gene expression. In conclusion, we identified SET as the first direct interacting partner of mammalian GnRHR and showed that SET contributes to a switch of GnRHR signaling toward the cAMP pathway.


Asunto(s)
Calcio/metabolismo , AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Gonadotrofos/metabolismo , Receptores LHRH/química , Animales , Señalización del Calcio , Proteínas de Unión al GTP/metabolismo , Humanos , Cinética , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Ratas , Receptores LHRH/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal
2.
J Biol Chem ; 288(12): 8505-8518, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23362264

RESUMEN

Hormone-sensitive lipase (HSL) catalyzes the hydrolysis of cholesteryl esters in steroidogenic tissues and, thus, facilitates cholesterol availability for steroidogenesis. The steroidogenic acute regulatory protein (StAR) controls the rate-limiting step in steroid biosynthesis. However, the modes of action of HSL in the regulation of StAR expression remain obscure. We demonstrate in MA-10 mouse Leydig cells that activation of the protein kinase A (PKA) pathway, by a cAMP analog Bt2cAMP, enhanced expression of HSL and its phosphorylation (P) at Ser-660 and Ser-563, but not at Ser-565, concomitant with increased HSL activity. Phosphorylation and activation of HSL coincided with increases in StAR, P-StAR (Ser-194), and progesterone levels. Inhibition of HSL activity by CAY10499 effectively suppressed Bt2cAMP-induced StAR expression and progesterone synthesis. Targeted silencing of endogenous HSL, with siRNAs, resulted in increased cholesteryl ester levels and decreased cholesterol content in MA-10 cells. Depletion of HSL affected lipoprotein-derived cellular cholesterol influx, diminished the supply of cholesterol to the mitochondria, and resulted in the repression of StAR and P-StAR levels. Cells overexpressing HSL increased the efficacy of liver X receptor (LXR) ligands on StAR expression and steroid synthesis, suggesting HSL-mediated steroidogenesis entails enhanced oxysterol production. Conversely, cells deficient in LXRs exhibited decreased HSL responsiveness. Furthermore, an increase in HSL was correlated with the LXR target genes, steroid receptor element-binding protein 1c and ATP binding cassette transporter A1, demonstrating HSL-dependent regulation of steroidogenesis predominantly involves LXR signaling. LXRs interact/cooperate with RXRs and result in the activation of StAR gene transcription. These findings provide novel insight and demonstrate the molecular events by which HSL acts to drive cAMP/PKA-mediated regulation of StAR expression and steroidogenesis in mouse Leydig cells.


Asunto(s)
Células Intersticiales del Testículo/enzimología , Fosfoproteínas/genética , Progesterona/biosíntesis , Esterol Esterasa/fisiología , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Bucladesina/farmacología , Carbamatos/farmacología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Colesterol/sangre , Ésteres del Colesterol/sangre , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , AMP Cíclico/metabolismo , AMP Cíclico/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Intersticiales del Testículo/metabolismo , Receptores X del Hígado , Masculino , Ratones , Ácidos Nicotínicos/farmacología , Receptores Nucleares Huérfanos/agonistas , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , Oxadiazoles/farmacología , Perilipina-1 , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Receptores X Retinoide/agonistas , Receptores X Retinoide/metabolismo , Receptores Depuradores de Clase B/genética , Receptores Depuradores de Clase B/metabolismo , Sistemas de Mensajero Secundario , Esterol Esterasa/genética , Esterol Esterasa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Tetrahidronaftalenos/farmacología
3.
Neuroendocrinology ; 97(2): 115-31, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22414758

RESUMEN

BACKGROUND AND AIMS: In mammals, activation of pituitary GnRH receptor (GnRHR) by hypothalamic GnRH increases the synthesis and secretion of LH and FSH, which, in turn, regulate gonadal functions. However, GnRHR gene (Gnrhr) expression is not restricted to the pituitary. METHODS: To gain insight into the extrapituitary expression of Gnrhr, a transgenic mouse model that expresses the human placental alkaline phosphatase reporter gene driven by the rat Gnrhr promoter was created. RESULTS: This study shows that the rat Gnrhr promoter is operative in two functionally related organs, the pineal gland, as early as embryonic day (E) 13.5, and the retina where activity was only detected at E17.5. Accordingly, Gnrhr mRNA were present in both tissues. Transcription factors known to regulate Gnrhr promoter activity such as the LIM homeodomain factors LHX3 and ISL1 were also detected in the retina. Furthermore, transient transfection studies in CHO and gonadotrope cells revealed that OTX2, a major transcription factor in both pineal and retina cell differentiation, is able to activate the Gnrhr promoter together with either CREB or PROP1, depending on the cell context. CONCLUSION: Rather than using alternate promoters, Gnrhr expression is directed to diverse cell lineages through specific associations of transcription factors acting on distinct response elements along the same promoter. These data open new avenues regarding GnRH-mediated control of seasonal and circadian rhythms in reproductive physiology.


Asunto(s)
Glándula Pineal/metabolismo , Regiones Promotoras Genéticas/genética , Receptores LHRH/genética , Retina/metabolismo , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Embrión de Mamíferos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Transgénicos , Especificidad de Órganos/genética , Glándula Pineal/embriología , Ratas , Ratas Sprague-Dawley , Retina/embriología
4.
Biol Reprod ; 82(6): 1170-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20181617

RESUMEN

Previous in vivo studies have established that pituitary nitric oxide synthase type 1 (NOS1) is regulated by gonadotropin-releasing hormone (GnRH). The aim of our study was to elucidate the mechanisms of NOS1 regulation by GnRH in rat pituitary cells. Using a perifused cell system, we demonstrated that NOS1 induction was sensitive to GnRH pulse frequency and was maximally induced under continuous GnRH stimulation. In primary cultures of rat pituitary cells, sustained stimulation with the GnRH agonist triptorelin (GnRHa) increased NOS1 protein levels, whereas NOS2 and NOS3 levels were unaffected. NOS1 up-regulation occurred in gonadotroph cells only, in a time-dependent and concentration-dependent manner (maximum increase, 2.5-fold; half-maximal concentration, 0.17 nM). GnRHa effect was mimicked by cAMP pathway activators and, most importantly, was blocked by disruption of the protein kinase A (PKA) pathway using pharmacological inhibitors such as Rp-cAMP or drug phosphatase technology-protein kinase inhibitor (DPT-PKI), a cell-permeant PKI peptide. In contrast, modulation of the PKC pathway and inhibition of the MAPK cascade were ineffective. Overall, these experiments demonstrated that GnRH-induced up-regulation of pituitary NOS1 is mediated notably by the cAMP/PKA pathway. Last, in vivo administration of a GnRH antagonist markedly inhibited the pituitary cAMP rise at proestrus in addition to suppressing NOS1 increase. Altogether, our data suggest that the cAMP/PKA signaling pathway is preferentially recruited under sustained GnRH stimulation in vivo during proestrus, allowing the expression of a specific set of PKA-regulated proteins, including NOS1, in gonadotroph cells.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Óxido Nítrico Sintasa/metabolismo , Hipófisis/enzimología , Proestro/metabolismo , Animales , Técnicas de Cultivo de Célula , AMP Cíclico/análisis , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Femenino , Hormona Liberadora de Gonadotropina/farmacología , Óxido Nítrico Sintasa/análisis , Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico Sintasa de Tipo II/análisis , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/análisis , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proestro/efectos de los fármacos , Ratas , Ratas Wistar
5.
Endocrinology ; 149(12): 6389-98, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18755795

RESUMEN

Gonadotrope cells are primarily regulated by GnRH but are also targets of the pituitary adenylyl cyclase-activating polypeptide (PACAP). Although it has been reported that reciprocal interactions between both neuropeptides contribute to regulation of gonadotrope function, the underlying mechanisms remain poorly understood. In this study, we reevaluated PACAP coupling to the cAMP pathway in LbetaT2 gonadotrope cells and analyzed GnRH effect on PACAP signaling. We established that PACAP38 markedly increases intracellular cAMP levels (EC50 of 4.7 +/- 1.3 nm) through the PACAP type 1 receptor (PAC1-R), as evidenced by pharmacological and RT-PCR studies. Interestingly, although GnRH couples to cAMP pathway in LbetaT2 cells, the effects of both neuropeptides were not synergistic. Instead, the GnRH agonist (GnRHa) triptorelin rapidly and strongly inhibited (70% inhibition as early as 5 min) PACAP38-induced cAMP production. Inhibition was calcium independent, mimicked by the phorbol ester phorbol 12-myristate 13-acetate, and blocked by the protein kinase C (PKC) inhibitor bisindoylmaleimide, indicating that GnRHa inhibitory action relies on PKC. Selective down-regulation of both conventional and novel PKC prevented a GnRHa effect, whereas pharmacological inhibition of conventional PKC only was ineffective, strongly suggesting the involvement of novel PKC isoforms. GnRHa did not inhibit forskolin- or cholera toxin-stimulated cAMP accumulation, suggesting that PAC1-R is the predominant target of GnRH. Accordingly, we demonstrated for the first time that GnRH increases PAC1-R phosphorylation through PKC, providing a potential molecular mechanism which may account for GnRH inhibitory effect.


Asunto(s)
AMP Cíclico/metabolismo , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Proteína Quinasa C/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Animales , Western Blotting , Calcio/metabolismo , Línea Celular , Gonadotrofos/citología , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/agonistas , Ratones , Fosforilación/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Unión Proteica/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pamoato de Triptorelina/farmacología
6.
Endocrinology ; 148(3): 1099-107, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17185372

RESUMEN

GnRH regulates the reproductive system by stimulating synthesis and release of gonadotropins. GnRH acts through a receptor coupled to multiple intracellular events including a rapid phosphoinositide turnover. Although the cAMP pathway is essential for gonadotrope function, the ability of GnRH to induce cAMP, as well as the coupling mechanisms involved, remain controversial. In this study, we established that GnRH increases intracellular cAMP levels in a concentration-dependent manner in LbetaT2 gonadotrope cells (maximal increase, 2.5-fold; EC(50), 0.30 nm), and this was further evidenced by GnRH activation of a cAMP-sensitive reporter gene. The GnRH effect was Ca(2+) independent, mimicked by the phorbol ester phorbol 12-myristate 13-acetate, and blocked by the protein kinase C (PKC) inhibitor bisindolylmaleimide, indicating that the GnRH effect was mediated by PKC. Pharmacological inhibition of conventional PKC isoforms with Gö6976 did not prevent GnRH-induced cAMP production, whereas down-regulation of novel PKCdelta, -epsilon, and -theta by a long-term treatment with GnRH markedly reduced it. Expression of dominant-negative (DN) mutants of PKCdelta or -epsilon but not PKCtheta impaired GnRH activation of a cAMP-sensitive promoter, demonstrating that PKCdelta and -epsilon are the two endogenous isoforms mediating GnRH activation of the adenylyl cyclase (AC) pathway in LbetaT2 cells. Accordingly, we identified by RT-PCR and immunocytochemical analysis, two PKC-sensitive AC isoforms, i.e. AC5 and AC7 as potential targets for GnRH. Lastly, we showed that only sustained stimulation of GnRH receptor significantly increased cAMP, suggesting that in vivo, the cAMP signaling pathway may be selectively recruited under intense GnRH release such as the preovulatory GnRH surge.


Asunto(s)
AMP Cíclico/metabolismo , Gonadotrofos , Hormona Liberadora de Gonadotropina/fisiología , Proteína Quinasa C-delta/fisiología , Proteína Quinasa C-epsilon/fisiología , Adenilil Ciclasas/metabolismo , Animales , Calcio/fisiología , Línea Celular , Gonadotrofos/efectos de los fármacos , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/agonistas , Isoenzimas/fisiología , Ratones , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Receptores LHRH/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Peptides ; 28(9): 1797-804, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17601637

RESUMEN

Strong evidence in favor of a direct action of hypothalamic PACAP at the pituitary to modulate gonadotrope function has been acquired mainly by in vitro studies using cultured pituitary cells or gonadotrope cell lines. In particular, PACAP has been shown to cooperate with GnRH, the primary regulator of gonadotropes, to regulate/modulate gonadotropin subunit gene expression, gonadotropin release as well as gonadotrope responsiveness. These effects of PACAP appear to be due essentially to its high potent stimulatory action on the cAMP/protein kinase pathway. Ensuing mechanisms include signaling cross-talk and/or enhanced gene expression within gonadotropes. PACAP may also indirectly operate on these cells through paracrine mechanisms. While PACAP has long been viewed as a hypophysiotropic factor, a locally produced PACAP has also been described. Interestingly, both appear similarly up-regulated at proestrus of the reproductive cycle in female rats. Further in vivo investigation is now necessary to ascertain the physiological relevance of the observed pituitary PACAP effects and especially to evaluate the respective contribution of hypothalamic and pituitary PACAP in the dynamic control of gonadotrope function.


Asunto(s)
Gonadotrofos/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Animales , AMP Cíclico/metabolismo , Femenino , Gonadotrofos/citología , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Modelos Biológicos , Hipófisis/citología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
8.
Mol Endocrinol ; 20(9): 2093-108, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16613990

RESUMEN

The GnRH receptor (GnRH-R) plays a central role in mammalian reproductive function throughout adulthood. It also appears as an early marker gene of the presumptive gonadotrope lineage in developing pituitary. Here, using transient transfections combined with DNA/protein interaction assays, we have delineated cis-acting elements within the rat GnRH-R gene promoter that represent targets for the LIM-homeodomain (LIM-HD) proteins, Isl-1 and Lhx3. These factors, critical in early pituitary development, are thus also crucial for gonadotrope-specific expression of the GnRH-R gene. In heterologous cells, the expression of Isl-1 and Lhx3, together with steroidogenic factor 1 (SF-1), culminates in the activation of both the rat as well as human GnRH-R promoter, suggesting that this combination is evolutionarily conserved among mammals. The specificity of these LIM-HD factors is attested by the inefficiency of related proteins, including Lhx5 and Lhx9, to activate the GnRH-R gene promoter, as well as by the repressive capacity of a dominant-negative derivative of Lhx3. Accordingly, targeted deletion of the LIM response element decreases promoter activity. In addition, experiments with Gal4-SF-1 fusion proteins suggest that LIM-HD protein activity in gonadotrope cells is dependent upon SF-1 binding. Finally, using a transgenic model that allows monitoring of in vivo promoter activity, we show that the overlapping expression of Isl-1 and Lhx3 in the developing pituitary correlates with promoter activity. Collectively, these data suggest the occurrence of a specific LIM-HD pituitary code and designate the GnRH-R gene as the first identified transcriptional target of Isl-1 in the anterior pituitary.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Hipófisis/metabolismo , Regiones Promotoras Genéticas/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores LHRH/genética , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cricetinae , ADN/genética , ADN/metabolismo , Proteínas de Homeodominio/genética , Humanos , Ratones , Especificidad de Órganos , Unión Proteica , Ratas , Receptores Citoplasmáticos y Nucleares/genética , Elementos de Respuesta/genética , Factor Esteroidogénico 1 , Factores de Transcripción/genética , Activación Transcripcional
9.
Mol Endocrinol ; 20(2): 362-78, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16166197

RESUMEN

Growth factors are known to play diverse roles in steroidogenesis, a process regulated by the mitochondrial steroidogenic acute regulatory (StAR) protein. The mechanism of action of one such growth factor, IGF-I, was investigated in mouse Leydig tumor (mLTC-1) cells to determine its potential role in the regulation of StAR expression. mLTC-1 cells treated with IGF-I demonstrated temporal and concentration-dependent increases in StAR expression and steroid synthesis. However, IGF-I had no effect on cytochrome P450 side-chain cleavage or 3beta-hydroxysteroid dehydrogenase protein levels. IGF-I was capable of augmenting N,O'-dibutyrl-cAMP-stimulated steroidogenic responsiveness in these cells. The steroidogenic potential of IGF-I was also confirmed in primary cultures of isolated mouse Leydig cells. IGF-I increased phosphorylation of ERK1/2, an event inhibited by the MAPK/ERK inhibitors, PD98059 and U0126. Interestingly, inhibition of ERK activity enhanced IGF-I-mediated StAR protein expression, but phosphorylation of StAR was undetectable, an observation in contrast to that seen with N,O'-dibutyrl-cAMP signaling. Further studies demonstrated that these events were tightly correlated with the expression of dosage-sensitive sex reversal, adrenal hypoplasia congenita, critical region on the X chromosome, gene 1 and scavenger receptor class B type 1. Whereas both protein kinase A and protein kinase C signaling were involved in the IGF-I-mediated steroidogenic response, the majority of the effects of IGF-I were found to be mediated by the protein kinase C pathway. Transcriptional activation of the StAR gene by IGF-I was influenced by several transcription factors, its up-regulation being dependent on phosphorylation of the cAMP response element-binding protein (CREB) and the activator protein 1 family member, c-Jun. Conversely, StAR gene transcription was markedly inhibited by expression of nonphosphorylatable CREB (Ser(133)Ala), dominant negative A-CREB, and dominant negative c-Jun (TAM-67) mutants. Collectively, the present studies identify molecular events in IGF-I signaling that may influence testicular growth, development, and the Leydig cell steroidogenic machinery through autocrine/paracrine regulation.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/fisiología , Células Intersticiales del Testículo/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Esteroides/biosíntesis , Activación Transcripcional , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Animales , Bucladesina/farmacología , Células Cultivadas , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/farmacología , Regulación de la Expresión Génica , Insulina/fisiología , Masculino , Ratones , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Fosforilación , Proteína Quinasa C/farmacología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transcripción Genética
10.
J Chem Neuroanat ; 31(1): 51-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16185839

RESUMEN

This study was designed to investigate the histochemical effects of maternal deprivation on the adrenocorticotrophic and gonadotrophic axes in the hypothalamo-pituitary unit of preweanling lambs. Twelve-week-old female lambs were divided into either the control (lambs reared under undisturbed maternal conditions; n=3) or the maternally deprived group (lambs separated for three days from their dams; n=3). The corticotrophin-releasing hormone (CRH) and gonadotrophin-releasing hormone (GnRH) in the median eminence and the adenohypophyseal adrenocorticotrophin (ACTH), gonadotrophins (LH and FSH) and mRNAs for their beta-subunits were investigated using the immunohistochemistry or hybridohistochemistry. In maternally deprived lambs, the percentage of the area occupied by immunoreactive (ir)-CRH nerve terminals was lower (P<0.05) and the percentage of the adenohypophyseal area (PAA) occupied by ir-ACTH cells was higher (P<0.05) compared with the control lambs. In the hypothalamo-gonadotrophic axis of maternally deprived lambs the percentage of area occupied by ir-GnRH nerve terminals was higher (P<0.05) and the PAA occupied by ir-FSHbeta cells was lower (P<0.05) in comparison with controls. The PAA occupied by gonadotrophs detected using hybridohistochemistry was higher (P<0.05) for LHbeta-mRNA in contrast to a lower (P<0.05) percentage for FSHbeta-mRNA in maternally deprived lambs compared with those staying with dams. In conclusion, maternal deprivation affected the accumulation of CRH and ACTH. The different and more striking alterations in FSH synthesis and storage in comparison with those concerning LH were observed in maternally deprived lambs. Thus, rupture of the preweanling young-mother social contact can affect the gonadotroph population activity, especially that relating to FSH-producing cells in the infantile female sheep.


Asunto(s)
Hormona Adrenocorticotrópica/metabolismo , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hormona Luteinizante de Subunidad beta/metabolismo , Privación Materna , Animales , Animales Lactantes , Hormona Liberadora de Corticotropina/metabolismo , Femenino , Hormona Folículo Estimulante de Subunidad beta/genética , Hormona Liberadora de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisario/citología , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Inmunohistoquímica , Hibridación in Situ , Hormona Luteinizante de Subunidad beta/genética , Eminencia Media/citología , Eminencia Media/crecimiento & desarrollo , Eminencia Media/metabolismo , Neuronas/metabolismo , ARN Mensajero/análisis , Ovinos
11.
Ann N Y Acad Sci ; 1070: 376-9, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16888195

RESUMEN

The effects and respective influence of pituitary adenylate cyclase-activating polypeptide (PACAP) and gonadotropin-releasing hormone (GnRH) on cyclic AMP (cAMP) production in pituitary gonadotropes were analyzed using the LbetaT2 cell line. Both hormones induced cAMP with, however, different intensity and time course. In addition, the GnRH effect was markedly reduced by PKC inhibitors. Despite its positive coupling to cAMP pathway, GnRH counteracted PACAP induction of cAMP and this effect was mimicked by the PKC activator phorbol 12-myristate 13-acetate (PMA). The data reveal major differences in the mechanisms by which PACAP and GnRH activate cAMP/PKA pathway in LbetaT2 cells and suggest that PKC activation serves GnRH not only to increase cAMP but also to counteract the PACAP stimulation of this signaling pathway.


Asunto(s)
AMP Cíclico/biosíntesis , Hormona Liberadora de Gonadotropina/farmacología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Animales , Línea Celular , Femenino , Ratones
12.
Brain Res Bull ; 65(5): 391-6, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15833593

RESUMEN

Metal complexes with GnRH were shown to interact with GnRH receptors in pituitary cells. In the present study we examined the effects of GnRH and its cobalt complex form (Co-GnRH) on LH secretion and generation of second messengers, namely inositol phosphates (IPs) and cAMP, in porcine pituitary cells in vitro. The cells were obtained from gilt pituitary at the pre-ovulatory phase of estrous cycle and cultured for 72 h before challenge with GnRH or Co-GnRH. Both substances induced a significant increase in LH release that was detectable after 60 min (P<0.05) of treatment, with the Co-GnRH complex being more efficient than GnRH at 180 min (P<0.01). GnRH and Co-GnRH were equally effective at 10(-8)M (P<0.01), however, at the lowest (10(-9)M) as well as the highest (10(-7)M) concentrations tested, Co-GnRH was more potent than its native counterpart (P<0.01). Interestingly, Co-GnRH revealed twice more efficient than GnRH at stimulating cAMP production, an effect which was detectable in cells after 1h-incubation (P<0.001). In contrast, while native GnRH induced a rapid increase (P<0.05) in IPs no such effect of Co-GnRH was observed. These data demonstrate that Co-GnRH and GnRH differentially effect on the signaling pathway in porcine gonadotropes and suggest that in these cells, the releasing action of Co-GnRH results from the mediation via the cAMP/protein kinase A second messenger system.


Asunto(s)
Cobalto/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Adenohipófisis/citología , Transducción de Señal/efectos de los fármacos , Análisis de Varianza , Animales , Células Cultivadas , Colforsina/farmacología , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Técnicas para Inmunoenzimas/métodos , Técnicas In Vitro , Hormona Luteinizante/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Transducción de Señal/fisiología , Porcinos , Factores de Tiempo
13.
Neuro Endocrinol Lett ; 26(4): 377-82, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16136005

RESUMEN

Gonadotropin releasing hormone (GnRH) is an essential factor in the regulation of synthesis and release of pituitary gonadotropins. After binding to specific receptors and coupling with G proteins, it triggers the intracellular signaling involving the synthesis of inositol phosphates and diacylglycerol. Previously we have showed that certain metal complexes with GnRH, i.e. copper (Cu-GnRH) and nickel (Ni-GnRH) are able to bind to the GnRH receptors. The intracellular signalling of these complexes, however, has not been yet elucidated. In this experiment, the ability of the Cu-GnRH and Ni-GnRH complexes to modulate cAMP synthesis and phosphoinositols formation in the pig anterior pituitary cells in vitro was studied. The native GnRH and its metal complexes stimulated the luteinizing hormone (LH) release, but only the effect of Cu-GnRH was found to be a dose-dependent. The metal complexes did not significantly influence inositol phosphates accumulation, while their effect on cAMP synthesis was significantly more potent than that of GnRH alone. We conclude that the Cu-GnRH and Ni-GnRH complexes increase LH release in the porcine pituitary cells although their intracellular signaling is different from that of the native GnRH. It seems that metal complexes with GnRH deserve more attention in further studies.


Asunto(s)
Cobre/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Níquel/metabolismo , Adenohipófisis/metabolismo , Transducción de Señal/fisiología , Animales , AMP Cíclico/metabolismo , Femenino , Hidrólisis , Fosfatos de Inositol/metabolismo , Hormona Luteinizante/metabolismo , Fosfatidilinositoles/metabolismo , Sus scrofa
14.
Endocrinology ; 144(9): 3995-4007, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12933674

RESUMEN

The neuronal nitric oxide synthase (NOS I) is expressed and hormonally regulated in rat anterior pituitary gonadotropes. In the present study, we investigated the mechanisms that underlie the constitutive and GnRH up-regulated activity of the pituitary exon 1p promoter of the NOS I gene in these cells. Through the use of 5'-deletions and transient transfections in L beta T2, a gonadotrope-derived cell line, we delineated a NOS I cell-specific (NCS) enhancer region (-73/-59) that is required for constitutive activity. Independently of the NCS enhancer, GnRH responsiveness is supported by a bipartite regulatory domain referred to as the GnRH response element I and II located between -33/-10 and -4/+4, the latter consisting of a cAMP-like response element. By combining transient transfections, gel shift, and supershift assays, we demonstrate that Sp1 and LIM-homeodomain-related protein bind the NCS enhancer, whereas cAMP response element binding protein and cAMP regulatory element modulator-like factors bind the GnRH response element II motif. We further show that factors involved in GnRH regulation are also implicated in constitutive activity, suggesting intimate links between constitutive and regulated promoter activity. We speculate that specific expression of the NOS I gene in gonadotropes together with its regulation by GnRH is suggestive of a critical participation of NOS I in gonadotrope function.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Hormona Liberadora de Gonadotropina/metabolismo , Proteínas de Homeodominio/metabolismo , Óxido Nítrico Sintasa/genética , Regiones Promotoras Genéticas/genética , Proteínas Represoras , Factor de Transcripción Sp1/metabolismo , Animales , Secuencia de Bases , Células CHO , Cricetinae , Modulador del Elemento de Respuesta al AMP Cíclico , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas con Homeodominio LIM , Datos de Secuencia Molecular , Óxido Nítrico Sintasa de Tipo I , Ratas , Factores de Transcripción , Transcripción Genética/fisiología , Transfección
15.
Endocrinology ; 145(2): 983-93, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14592958

RESUMEN

Previous studies dealing with the mechanisms underlying the tissue-specific and regulated expression of the GnRH receptor (GnRH-R) gene led us to define several cis-acting regulatory sequences in the rat GnRH-R gene promoter. These include functional sites for steroidogenic factor 1, activator protein 1, and motifs related to GATA and LIM homeodomain response elements as demonstrated primarily in transient transfection assays in mouse gonadotrope-derived cell lines. To understand these mechanisms in more depth, we generated transgenic mice bearing the 3.3-kb rat GnRH-R promoter linked to the human placental alkaline phosphatase reporter gene. Here we show that the rat GnRH-R promoter drives the expression of the reporter gene in pituitary cells expressing the LHbeta and/or FSHbeta subunit but not in TSHbeta- or GH-positive cells. Furthermore, the spatial and temporal pattern of the transgene expression during the development of the pituitary was compatible with that characterizing the emergence of the gonadotrope lineage. In particular, transgene expression is colocalized with the expression of the glycoprotein hormone alpha-subunit at embryonic day 13.5 and with that of steroidogenic factor 1 at later stages of pituitary development. Transgene expression was also found in specific brain areas, such as the lateral septum and the hippocampus. A single promoter is thus capable of directing transcription in highly diverse tissues, raising the question of the different combinations of transcription factors that lead to such a multiple, but nevertheless cell-specific, expressions of the GnRH-R gene.


Asunto(s)
Fosfatasa Alcalina/genética , Genes Reporteros/genética , Hormona Liberadora de Gonadotropina/genética , Adenohipófisis/enzimología , Placenta/enzimología , Regiones Promotoras Genéticas/genética , Animales , Química Encefálica , Femenino , Hormona Folículo Estimulante de Subunidad beta/genética , Eliminación de Gen , Expresión Génica , Regulación de la Expresión Génica , Histocitoquímica , Humanos , Hormona Luteinizante de Subunidad beta/genética , Masculino , Ratones , Ratones Transgénicos , Adenohipófisis/embriología , Adenohipófisis/crecimiento & desarrollo , Embarazo , Ratas , Proteínas Recombinantes de Fusión , Secuencias Reguladoras de Ácidos Nucleicos
16.
J Chem Neuroanat ; 23(2): 133-42, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11841917

RESUMEN

Neuropeptide Y (NPY) is a putative neuroregulator of the reproductive axis in the central nervous system. In this study we evaluated the effects of central infusion of exogenous NPY on the secretory activity of pituitary gonadotrophic cells in prepubertal lambs. Immature female Merino sheep (n=12) were infused of Ringer solution (control) or 50 microg of NPY to the third ventricle for 5 min and then slaughtered 3 h later. Immunoreactive luteinizing hormone (LH) and follicle stimulating hormone (FSH) cells were localised by immunohistochemistry using antibody raised against LHbeta and FSHbeta. Messenger RNA analyses were performed by in situ hybridisation using sense and antisense riboprobes produced from beta subunits of LH and FSH cDNA clones. The results were generated by computer image analysis to determine the area fraction occupied by immunoreactive and/or hybridising cells and optical density for immunostaining and hybridisation signal. LH in the blood plasma was determined by radioimmunoassay. It was found, that in the lambs infused with NPY the area fraction and optical density for immunoreactive LH cells and mRNA LHbeta-expressing cells increased significantly (P<0.001), compared to the vehicle-infused animals. The concentration of LH in the blood plasma did not differ between control and treated groups. The NPY infusions had no effect on the immunoreactivity of FSH cells or on expression of mRNA for FSHbeta. In conclusion we suggest that NPY may be an important component of mechanisms stimulating the synthesis and storage but not the release of LH in the pituitary gonadotrophs from prepubertal female sheep. In addition, this effect is specific for LH, no such effect was apparent on FSH.


Asunto(s)
Hormona Folículo Estimulante/genética , Hormona Luteinizante/genética , Neuropéptido Y/farmacología , Hipófisis/fisiología , Factores de Edad , Animales , Anticuerpos , Femenino , Hormona Folículo Estimulante/análisis , Hormona Folículo Estimulante/inmunología , Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Hibridación in Situ , Inyecciones Intraventriculares , Hormona Luteinizante/análisis , Hormona Luteinizante/inmunología , Hipófisis/química , Hipófisis/efectos de los fármacos , ARN Mensajero/análisis , Ovinos
17.
Fertil Steril ; 77(6): 1288-91, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12057744

RESUMEN

OBJECTIVE: To report the occurrence of a spontaneous pregnancy in a patient who was homozygous for the Q106R mutation in the GnRH receptor (GnRHR) gene. DESIGN: Case report. SETTING: Reproductive endocrinology unit of an academic medical center. PATIENT(S): A 27-year-old woman who initially presented with partial idiopathic hypogonadotropic hypogonadism and who achieved a spontaneous pregnancy 3 months after oral contraceptive pill (OCP) withdrawal. INTERVENTION(S): Blood sampling for hormonal and genetic investigations, transvaginal ultrasound. MAIN OUTCOME MEASURE(S): LH, FSH, E2, and betahCG serum levels. Ultrasound examination of the uterine cavity. RESULT(S): Three months after OCP withdrawal, the patient was amenorrheic. However, the betahCG serum level was 149 IU/L. Transvaginal ultrasound 2 weeks later revealed the presence of one intrauterine sac containing two embryos with cardiac activity. At 9 weeks of gestation, no cardiac activity was found. A curettage was then performed, and the pathological examination indicated the presence of chorionic villi. CONCLUSION(S): OCP withdrawal might have induced a transient situation with optimal endogenous pulsatile GnRH secretion, thus overriding the GnRH resistance induced by the partially inactivating Q106R GnRHR gene mutation and allowing ovulation to occur.


Asunto(s)
Homocigoto , Mutación/fisiología , Embarazo/genética , Receptores LHRH/genética , Adulto , Secuencia de Bases/genética , Gonadotropina Coriónica Humana de Subunidad beta/sangre , Anticonceptivos Orales/administración & dosificación , Esquema de Medicación , Estradiol/sangre , Femenino , Hormona Folículo Estimulante/sangre , Hormona Liberadora de Gonadotropina/uso terapéutico , Humanos , Hormona Luteinizante/sangre , Inducción de la Ovulación , Linaje , Embarazo/fisiología , Ultrasonografía Prenatal , Útero/diagnóstico por imagen
18.
Brain Res Bull ; 58(5): 475-80, 2002 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12242100

RESUMEN

The effects of separate and simultaneous recombinant bovine (rb) activin A and buserelin administration on the FSH release and pituitary FSH beta subunit gene expression in vivo were examined in ovariectomised, estradiol pretreated rats. The animals received a single injection of either rb activin A (50 ng), buserelin (1 micro g) or activin/buserelin (50 ng+1 micro g/0.1 ml PBS) into the jugular vein and were killed 30 min, 1, 3 and 5h later. Activin A stimulated FSH release and effect appeared 1h after injection (168% increase of controls) reaching a maximum at 3h (437% of controls). Activin A and buserelin exerted their effects with a distinct time courses: activin's stimulation was not so rapid when compared with buserelin. The simultaneous administration of rb activin A and buserelin amplified FSH release (118, 309, 1006 and 779% of controls). The low dose of activin A was sufficient to elevate FSH beta mRNA level as early as 3 and 5h after administration (170 and 140%, respectively). Activin plus buserelin stimulation resulted in a higher (340 and 360% of controls) FSH beta gene expression than after their separate administration. These results suggest that activin and buserelin may act independently and synergistically in the regulation of FSH release and beta subunit mRNA level.


Asunto(s)
Activinas/farmacología , Buserelina/farmacología , Hormona Folículo Estimulante de Subunidad beta/biosíntesis , Hormona Folículo Estimulante/metabolismo , ARN Mensajero/análisis , Activinas/administración & dosificación , Animales , Buserelina/administración & dosificación , Células Cultivadas , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Hormona Folículo Estimulante/sangre , Hormona Folículo Estimulante/genética , Hormona Folículo Estimulante de Subunidad beta/genética , Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/agonistas , Ovariectomía , Adenohipófisis/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
19.
Neuro Endocrinol Lett ; 21(4): 277-281, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11455359

RESUMEN

OBJECTIVES: The purpose of this study was to examine whether in long-term ovariectomized rats direct pulsatile intracerebroventricular microinfusions of GnRH would result in frequency dependent biosynthesis of pituitary FSH beta-subunit mRNA. METHODS: Stainless steel cannula was stereotaxically implanted into the third ventricle of ovariectomized rats. 1 nM of GnRH microinjections were given at frequency 1, 2, 4 pulses/hour during 5 hours. Pituitary FSH beta-subunit mRNA level was determined by Northern-blot and serum FSH concentration was examined by RIA. RESULTS: Exogenous GnRH (1nM) induced a significant increase of pituitary content of FSH beta-subunit mRNA when administered 30 or 60 min intervals over 5 hours to ovariectomized rats. RESULTS: GnRH microinjections given at frequency of 1 pulse/hour were mostly effective resulting both in 85% increase of FSH beta-subunit mRNA level as compared to control as well as a significant (78%) stimulation of FSH release. CONCLUSIONS: our data show that in a long term ovariectomized rats (without steroid and gonadal peptides supplementation) a direct pulsatile intracerebroventricular microinjections of GnRH induce frequency-dependent pituitary FSH beta-subunit mRNA biosynthesis in the same mode as it was reported for gonadectomized and steroid replaced rats. It seems therefore that this method would represent an interesting alternative to the use of classical agents to disconnect, in vivo, the pituitary from hypothalamic GnRH influence. It may also provide a physiological data concerning regulatory aspects of gonadotropin subunits biosynthesis.

20.
Neuro Endocrinol Lett ; 21(6): 431-436, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11335862

RESUMEN

OBJECTIVES: The aim of the study was to examine susceptibility of the pituitary gland to estrogenic impulse in old, noncycling rats by measurement of steady state level of mRNAs encoding LH subunits a and b and mRNA for PRL. METHODS: 22-month-old rats were ovariectomized and after one week they were subcutaneously implanted with silastic tubing filled with oil or with estradiol 17-beta. Pituitary alpha, LHbeta and PRL mRNAs content and serum LH and PRL concentration was determined. RESULTS: The effect of E (2)treatment was manifested by the significant increase in the weight of the uterus and pituitary gland as well as by elevation of total pituitary RNA (109%, 60% and 78%, respectively; p<0.001). No significant changes (p>0.05) in serum LH concentration were observed, while levels of mRNAs encoding alpha and LH-beta subunits were lowered by 54% (p<0.05) and 96% (p<0.01), respectively, in the rats subjected to E(2) stimuli. No direct correlation between synthesis and release of LH in E(2) treated old rats was observed. The blood PRL concentration and the pituitary level of PRL mRNA increased up to 2,000% and 1,300%, respectively (p<0.001). Spontaneous pituitary adenoma was observed in about 30% of the rats, irrespective of treatment. CONCLUSIONS: These data show that in old rats estrogenic stimulus can effectively diminish both pituitary LH subunits mRNAs as well as stimulate pituitary PRL mRNA level indicating that the E(2)-dependent processes involved in the regulation of corresponding genes are still functional.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA