Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biol Reprod ; 94(3): 62, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26843449

RESUMEN

Parturition in rodents is highly dependent on the engagement of the luteal prostaglandin F2 alpha receptor, which, through activation of the Gq/11 family of G proteins, increases the expression of Akr1c18, leading to an increase in progesterone catabolism. To further understand the involvement of Gq/11 on luteolysis and parturition, we used microarray analysis to compare the ovarian transcriptome of mice with a granulosa/luteal cell-specific deletion of Galphaq/11 with their control littermates on Day 18 of pregnancy, when mice from both genotypes are pregnant, and on Day 22, when mice with a granulosa/luteal cell-specific deletion of Galphaq/11 are still pregnant but their control littermates are 1-2 days postpartum. Ovarian genes up-regulated at the end of gestation in a Galphaq/11 -dependent fashion include genes involved in focal adhesion and extracellular matrix interactions. Genes down-regulated at the end of gestation in a Galphaq/11-dependent manner include Serpina6 (which encodes corticosteroid-binding globulin); Enpp2 (which encodes autotaxin, the enzyme responsible for the synthesis of lysophosphatidic acid); genes involved in protein processing and export; reproductive genes, such as Lhcgr; the three genes needed to convert progesterone to estradiol (Cyp17a1, Hsd17b7, and Cyp19a1); and Inha. Activation of ovarian Gq/11 by engagement of the prostaglandin F2 alpha receptor on Day 18 of pregnancy recapitulated the regulation of many but not all of these genes. Thus, although the ovarian transcriptome at the end of gestation is highly dependent on the activation of Gq/11, not all of these changes are dependent on the actions of prostaglandin F2 alpha.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Regulación de la Expresión Génica/fisiología , Transcriptoma , Animales , Cloprostenol/farmacología , Regulación hacia Abajo , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Genotipo , Células Lúteas , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Embarazo , Regulación hacia Arriba
2.
Biol Reprod ; 95(1): 27, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27335071

RESUMEN

The Frontiers in Reproduction (FIR) course has been held annually since 1998 at the Marine Biological Laboratories in Woods Hole, MA. The primary purpose of the course is to train young reproductive biologists in cutting-edge techniques that would strengthen their career opportunities. An initial evaluation of the FIR course was conducted by surveying the participants who took the course between 1998 and 2002. The findings of this survey were published in Biology of Reproduction in 2006, which highlighted the overall positive impact the course had on the training and upward career trajectory of the participants during the first 5 yr. The current study was designed to access the continued impact of FIR at the 10-yr mark by evaluating the participants who took the course between 1998 and 2008 using two different survey mechanisms. Based on these evaluations and feedback from the participants, it was evident that 1) FIR continues to have a significant positive impact on the careers of the participants, 2) the majority of the participants continue to be involved in research or administration related to the reproductive sciences, 3) nearly 90% of the attendees have been successful in obtaining funding for their research, and 4) most alumni have published at least five manuscripts in higher impact journals since they took the course. Therefore, it is evident that FIR participants are highly successful and continue to significantly impact the advances in the reproductive sciences worldwide.


Asunto(s)
Reproducción , Ciencia/educación , Movilidad Laboral , Humanos , Evaluación de Programas y Proyectos de Salud , Investigación
3.
J Assist Reprod Genet ; 32(5): 737-45, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25682117

RESUMEN

PURPOSE: Cathepsin L and ADAMTS-1 are known to play critical roles in follicular rupture, ovulation, and fertility in mice. Similar studies in humans are limited; however, both are known to increase during the periovulatory period. No studies have examined either protease in the follicular fluid of women with unexplained infertility or infertility related to advanced maternal age (AMA). We sought to determine if alterations in cathepsin L and/or ADAMTS-1 existed in these infertile populations. METHODS: Patients undergoing in vitro fertilization (IVF) for unexplained infertility or AMA-related infertility were prospectively recruited for the study; patients with tubal or male factor infertility were recruited as controls. Follicular fluid was collected to determine gene expression (via quantitative polymerase chain reaction), enzyme concentrations (via enzyme-linked immunosorbent assays), and enzymatic activities (via fluorogenic enzyme cleavage assay or Western blot analysis) of cathepsin L and ADAMTS-1. RESULTS: The analysis included a total of 42 patients (14 per group). We found no statistically significant difference in gene expression, enzyme concentration, or enzymatic activity of cathepsin L or ADAMTS-1 in unexplained infertility or AMA-related infertility as compared to controls. We also found no statistically significant difference in expression or concentration with advancing age. CONCLUSIONS: Cathepsin L and ADAMTS-1 are not altered in women with unexplained infertility or AMA-related infertility undergoing IVF, and they do not decline with advancing age. It is possible that differences exist in natural cycles, contributing to infertility; however, our findings do not support a role for protease alterations as a common cause of infertility.


Asunto(s)
Proteínas ADAM/metabolismo , Catepsina L/metabolismo , Líquido Folicular/enzimología , Infertilidad/diagnóstico , Infertilidad/enzimología , Proteínas ADAM/genética , Proteína ADAMTS1 , Adulto , Animales , Western Blotting , Estudios de Casos y Controles , Catepsina L/genética , Femenino , Fertilización In Vitro , Líquido Folicular/química , Humanos , Técnicas para Inmunoenzimas , Masculino , Edad Materna , Ratones , Pronóstico , Estudios Prospectivos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
BMC Med Genet ; 14: 77, 2013 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-23889750

RESUMEN

BACKGROUND: Preterm birth (PTB) is a complex disorder associated with significant neonatal mortality and morbidity and long-term adverse health consequences. Multiple lines of evidence suggest that genetic factors play an important role in its etiology. This study was designed to identify genetic variation associated with PTB in oxytocin pathway genes whose role in parturition is well known. METHODS: To identify common genetic variants predisposing to PTB, we genotyped 16 single nucleotide polymorphisms (SNPs) in the oxytocin (OXT), oxytocin receptor (OXTR), and leucyl/cystinyl aminopeptidase (LNPEP) genes in 651 case infants from the U.S. and one or both of their parents. In addition, we examined the role of rare genetic variation in susceptibility to PTB by conducting direct sequence analysis of OXTR in 1394 cases and 1112 controls from the U.S., Argentina, Denmark, and Finland. This study was further extended to maternal triads (maternal grandparents-mother of a case infant, N=309). We also performed in vitro analysis of selected rare OXTR missense variants to evaluate their functional importance. RESULTS: Maternal genetic effect analysis of the SNP genotype data revealed four SNPs in LNPEP that show significant association with prematurity. In our case-control sequence analysis, we detected fourteen coding variants in exon 3 of OXTR, all but four of which were found in cases only. Of the fourteen variants, three were previously unreported novel rare variants. When the sequence data from the maternal triads were analyzed using the transmission disequilibrium test, two common missense SNPs (rs4686302 and rs237902) in OXTR showed suggestive association for three gestational age subgroups. In vitro functional assays showed a significant difference in ligand binding between wild-type and two mutant receptors. CONCLUSIONS: Our study suggests an association between maternal common polymorphisms in LNPEP and susceptibility to PTB. Maternal OXTR missense SNPs rs4686302 and rs237902 may have gestational age-dependent effects on prematurity. Most of the OXTR rare variants identified do not appear to significantly contribute to the risk of PTB, but those shown to affect receptor function in our in vitro study warrant further investigation. Future studies with larger sample sizes are needed to confirm the findings of this study.


Asunto(s)
Cistinil Aminopeptidasa/genética , Estudios de Asociación Genética , Variación Estructural del Genoma , Nacimiento Prematuro/genética , Receptores de Oxitocina/genética , Alelos , Animales , Argentina , Células COS , Estudios de Casos y Controles , Chlorocebus aethiops , Cistinil Aminopeptidasa/metabolismo , Dinamarca , Femenino , Finlandia , Predisposición Genética a la Enfermedad , Edad Gestacional , Haplotipos , Humanos , Patrón de Herencia , Fosfatos de Inositol/metabolismo , Mutación Missense , Oxitocina/genética , Oxitocina/metabolismo , Polimorfismo de Nucleótido Simple , Embarazo , Unión Proteica , Receptores de Oxitocina/metabolismo , Factores de Riesgo
5.
Cell Signal ; 20(10): 1822-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18647647

RESUMEN

Recent studies showed that Fyn is a mediator of the LHR-induced activation of the ERK1/2 cascade in MA-10 cells. Since the LHR is a G protein-coupled receptor and the Src family of kinases can be activated by some Galpha subunits and by the non-visual arrestins we investigated the role of these signaling molecules in the LHR-provoked activation of Fyn. Small interfering RNAs (siRNAs) that target two Galpha subunits that participate in LHR signaling (Galpha(s) and Galpha(11)) and one that targets arrestin-3 were co-transfected with the hLHR in MA-10 cells. We then determined the effects of these siRNAs on the LHR-provoked activation of Fyn, the phosphorylation of FAK (a prominent Fyn substrate) and the release of EGF-like growth factors (a Fyn-mediated process). Expression of the siRNA against Galpha(s) decreased the level of Galpha(s) and LHR-stimulated cAMP production by approximately 50% but did not affect LHR-stimulated Fyn activation or FAK phosphorylation. Likewise, expression of the siRNA against Galpha(11) decreased the level of Galpha(11) and LHR-stimulated inositol phosphate production by approximately 50% but did not affect LHR-stimulated Fyn activation or FAK phosphorylation. Expression of the siRNA against arrestin-3 decreased the level of arrestin-3 and the rate of internalization of hCG by approximately 50% and it also inhibited the LHR-provoked stimulation of Fyn, the phosphorylation of FAK and the release of EGF-like growth factors. These results show that, in MA-10 cells, the hLHR activates Fyn through an arrestin-3-dependent pathway and that this pathway is a mediator of the hLHR-provoked release of EGF-like growth factors.


Asunto(s)
Arrestinas/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de HL/metabolismo , Animales , Gonadotropina Coriónica/farmacología , Activación Enzimática/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular , Quinasa 1 de Adhesión Focal/metabolismo , Proteínas de Unión al GTP/metabolismo , Silenciador del Gen/efectos de los fármacos , Humanos , Ratones , Modelos Biológicos , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/metabolismo
6.
Endocrinology ; 149(11): 5549-56, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18653716

RESUMEN

We examine the pathways involved in the luteinizing hormone receptor (LHR)-dependent activation of the epidermal growth factor (EGF) network using cocultures of LHR-positive granulosa cells and LHR-negative test cells expressing an EGF receptor (EGFR)-green fluorescent protein fusion protein. Activation of the LHR in granulosa cells results in the release of EGF-like growth factors that are detected by measuring the phosphorylation of the EGFR-green fluorescent protein expressed only in the LHR-negative test cells. Using neutralizing antibodies and real-time PCR, we identified epiregulin as the main EGF-like growth factor produced upon activation of the LHR expressed in immature rat granulosa cells, and we show that exclusive inhibition or activation of the ERK1/2 cascade in granulosa cells prevents or enhances epiregulin release, respectively, with little or no effect on epiregulin expression. These results show that the LHR-stimulated ERK1/2 pathway stimulates epiregulin release.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Células de la Granulosa/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Receptores de HL/fisiología , Animales , Células Cultivadas , Activación Enzimática , Epirregulina , Receptores ErbB/metabolismo , Femenino , Células de la Granulosa/enzimología , Metaloproteasas/fisiología , Fosforilación , Ratas , Receptores de HL/genética , Transfección
7.
Endocr Rev ; 23(2): 141-74, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11943741

RESUMEN

Reproduction cannot take place without the proper functioning of the lutropin/choriogonadotropin receptor (LHR). When the LHR does not work properly, ovulation does not occur in females and Leydig cells do not develop normally in the male. Also, because the LHR is essential for sustaining the elevated levels of progesterone needed to maintain pregnancy during the first trimester, disruptions in the functions of the LHR during pregnancy have catastrophic consequences. As such, a full understanding of the biology of the LHR is essential to the survival of our species. In this review we summarize our current knowledge of the structure, functions, and regulation of this important receptor.


Asunto(s)
Receptores de HL/fisiología , Reproducción/fisiología , Secuencia de Aminoácidos/genética , Animales , Gonadotropina Coriónica/metabolismo , Femenino , Expresión Génica , Regulación de la Expresión Génica , Humanos , Hormona Luteinizante/metabolismo , Masculino , Datos de Secuencia Molecular , Embarazo , ARN Mensajero/metabolismo , Receptores de HL/genética , Transducción de Señal/fisiología , Relación Estructura-Actividad
8.
Mol Cell Endocrinol ; 285(1-2): 62-72, 2008 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-18313839

RESUMEN

Using primary cultures of immature rat granulosa cells and adenoviral infections we expressed two mutants of the human lutropin receptor (hLHR) that do not activate the phosphoinositide cascade. One mutant (hLFF) has the extracellular domain of the hLHR and the transmembrane and intracellular domains of the hFSHR. The other (hLHR-L457D) has a leucine to aspartate mutation in residue 457 of transmembrane helix 3. When expressed in immature rat granulosa cells the hLHR stimulates cAMP and inositol phosphate accumulation, transactivates the epidermal growth factor receptor (EGFR), elicits a transient increase in Akt phosphorylation, and a sustained increase in ERK1/2 phosphorylation but aromatase expression is not enhanced. When expressed at comparable densities, hLFF and hLHR-L457D support cAMP accumulation and transient Akt phosphorylation but do not support inositol phosphate accumulation, EGFR transactivation or a sustained phosphorylation of ERK1/2. Cells expressing either of these two mutants respond to hCG with increased aromatase expression. We also show that addition of hCG to cells expressing the hLHR antagonizes the effects of hFSH on aromatase expression whereas addition of hCG to cells expressing the hLHR-L457D mutant does not. These results show that activation of the phosphoinositide cascade is upstream of EGFR transactivation and ERK1/2 phosphorylation and that this pathway is a negative regulator of aromatase expression in granulosa cells.


Asunto(s)
Aromatasa/metabolismo , Gonadotropina Coriónica/metabolismo , Células de la Granulosa/fisiología , Mutación , Fosfatidilinositoles/metabolismo , Receptores de HL/metabolismo , Sistemas de Mensajero Secundario/fisiología , Animales , Aromatasa/genética , Células Cultivadas , Activación Enzimática , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Células de la Granulosa/citología , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
9.
Mol Cell Endocrinol ; 470: 179-187, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29107092

RESUMEN

Towards the end of gestation prostaglandin F2α (PGF2α) stimulates the expression of Akr1c18 in the murine corpus luteum. Akr1c18 codes for 20α-hydroxysteroid dehydrogenase, an enzyme that precipitates parturition by catabolizing progesterone. Previous results from our laboratory have shown that this effect of PGF2α is mediated by the activation of Gαq/11, but the downstream effector(s) of Gαq/11 that elicit the increase in Akr1c18 expression have not been identified. The physiological effects of Gαq/11 are mediated by its ability to interact with phospholipase Cß, p63RhoGEF, and PKCζ. In the experiments described herein we used biochemical and pharmacological approaches, as well as adenoviral-mediated expression of a constitutively active form of Gαq and mutants thereof, to examine the role of each of these effectors as potential mediators of the increased expression of luteal Akr1c18. By measuring the effects of PGF2α on the activation of RhoA (activated by p63RhoGEF) and the effects of activators and inhibitors of RhoA on the PGF2α-induced expression of luteal Akr1c18, we determined that RhoA is neither activated by PGF2α or involved in the PGF2α-induced expression of luteal Akr1c18. The potential involvement of PKCζ was ruled out by the inability of a mutant of a constitutively active Gαq that prevents PKCζ binding to block the increased expression of Akr1c18. Furthermore, PGF2α does not increase the phosphorylation of ERK-5, the only known downstream target of PKCζ. On the other hand, three different mutants of a constitutively active Gαq that prevent phospholipase C activation blocked the induction of luteal Akr1c18. We conclude that the induction of luteal Akr1c18 by Gαq/11 is mediated by the activation of phospholipase C.


Asunto(s)
Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células Lúteas/metabolismo , Fosfolipasas de Tipo C/metabolismo , Adenoviridae/metabolismo , Animales , Dinoprost/farmacología , Femenino , Humanos , Fosfatos de Inositol/metabolismo , Células Lúteas/efectos de los fármacos , Masculino , Ratones Transgénicos , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Mutación/genética , Fosforilación/efectos de los fármacos , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
10.
Trends Endocrinol Metab ; 29(5): 313-325, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29602523

RESUMEN

The central role of luteinizing hormone (LH) and its receptor (LHCGR) in triggering ovulation has been recognized for decades. Because the LHCGR is present in the mural (outermost) granulosa cell layer of preovulatory follicles (POFs), the LH-initiated signal has to be transmitted to another somatic cell type (cumulus granulosa cells) and the oocyte to release a fertilizable oocyte. Recent studies have shown that activation of the LHCGR initiates vectorial transfer of information among the two somatic cell types and the oocyte and the molecules and signaling pathways involved are now better understood. This review summarizes the newer developments on the complex signaling pathways that regulate ovulation.


Asunto(s)
Comunicación Autocrina/fisiología , Receptores ErbB/metabolismo , Ovulación/fisiología , Comunicación Paracrina/fisiología , Animales , Comunicación Autocrina/genética , Receptores ErbB/genética , Femenino , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Ovulación/genética , Comunicación Paracrina/genética , Receptores de HL/genética , Receptores de HL/metabolismo
11.
Endocrinology ; 148(7): 3214-25, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17412805

RESUMEN

Primary cultures of progenitor and immature rat Leydig cells were established from the testes of 21- and 35-d-old rats, respectively. The cell population remained homogeneous after 4-6 d in culture as judged by staining for 3beta-hydroxysteroid dehydrogenase, but the cells were unable to bind 125I-human chorionic gonadotropin (hCG) or to respond to hCG with classical LH receptor (LHR)-mediated responses, including cAMP and inositol phosphate accumulation, steroid biosynthesis, or the phosphorylation of ERK1/2. Infection of primary cultures with recombinant adenovirus coding for beta-galactosidase showed that approximately 65% of the cells are infected. Infection with adenovirus coding for the human LHR (hLHR) allowed for expression of the hLHR at a density of approximately 25,000 receptors per cell and allowed the cells to respond to hCG with increases in cAMP and inositol phosphate accumulation, steroid biosynthesis, and the phosphorylation of ERK1/2. Although progenitor and immature cells were able to respond to hCG with an increase in progesterone, only the immature cells responded with an increase in testosterone. In addition to these classical LHR-mediated responses, the primary cultures of progenitor or immature rat Leydig cells expressing the recombinant hLHR proliferated robustly when incubated with hCG, and this proliferative response was sensitive to an inhibitor of ERK1/2 phosphorylation. These studies establish a novel experimental paradigm that can be used to study the proliferative response of Leydig cells to LH/CG. We conclude that activation of the LHR-provoked Leydig cell proliferation requires activation of the ERK1/2 cascade.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Gonadotropina Coriónica/farmacología , Células Intersticiales del Testículo/efectos de los fármacos , Hormona Luteinizante/farmacología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Animales , Western Blotting , Butadienos/farmacología , Células Cultivadas , AMP Cíclico/metabolismo , Dipéptidos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Flavonoides/farmacología , Inmunohistoquímica , Fosfatos de Inositol/metabolismo , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Quinazolinas , Ratas , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Esteroides/metabolismo , Tirfostinos/farmacología
12.
Mol Endocrinol ; 20(12): 3308-20, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16973759

RESUMEN

Human chorionic gonadotropin and human FSH (hFSH) elicit a transient increase in ERK1/2 phosphorylation lasting less than 60 min in immature granulosa cells expressing a low density of gonadotropin receptors. In cells expressing a high density of receptors, human chorionic gonadotropin and human FSH elicit this fast transient increase in ERK1/2 phosphorylation and also a delayed and more sustained increase that is detectable after 6-9 h. Both the early and delayed increases in ERK1/2 phosphorylation can be blocked with inhibitors of protein kinase A, the epidermal growth factor receptor kinase, metalloproteases, and MAPK kinase. The delayed effect, but not the early effect, can also be blocked with an inhibitor of protein kinase C. Because the delayed increase in ERK1/2 phosphorylation correlates with low aromatase expression in response to gonadotropins, we tested the effects of these inhibitors on aromatase expression. These inhibitors had little or no effect on gonadotropin-induced aromatase expression in cells expressing a low density of receptors, but they enhanced gonadotropin-induced aromatase expression in cells expressing a high density of receptors. Phorbol esters also induced a prolonged increase in ERK1/2 phosphorylation and, when added together with hFSH, blocked the induction of aromatase expression by hFSH in cells expressing a low density of hFSH receptor. A MAPK kinase inhibitor reversed the inhibitory effect of the phorbol ester on aromatase induction. We conclude that the effects of gonadotropins on ERK1/2 phosphorylation are mediated by epidermal growth factor-like growth factors and that the delayed effect is partially mediated by protein kinase C and acts as a negative regulator of aromatase expression.


Asunto(s)
Aromatasa/metabolismo , Gonadotropina Coriónica Humana de Subunidad beta/farmacología , Hormona Folículo Estimulante Humana/farmacología , Células de la Granulosa/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Animales , Activación Enzimática , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Epirregulina , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Células de la Granulosa/enzimología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Ratas , Receptores de Gonadotropina/metabolismo , Acetato de Tetradecanoilforbol/farmacología
13.
Mol Endocrinol ; 20(11): 2931-45, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16803865

RESUMEN

Using biochemical and imaging approaches, we examined the postendocytotic fate of the complex formed by human choriogonadotropin (hCG) and a constitutively active mutant of the human lutropin receptor (hLHR-L457R) found in a boy with precocious puberty and Leydig cell hyperplasia. After internalization, some of the complex formed by the hLHR-wild type (hLHR-wt) and hCG recycles to the cell surface, and some is found in lysosomes where the hormone is degraded. In contrast, the complex formed by the hLHR-L457R and hCG is not routed to the lysosomes, most of it is recycled to the cell surface and hormone degradation is barely detectable. For both, hLHR-wt and -L457R, there is an hCG-induced loss of cell surface receptors that accompanies internalization but this loss cannot be prevented by leupeptin. The removal of recycling motifs of the hLHR by truncation of the C-terminal tail at residue 682 greatly enhances the lysosomal accumulation of the hormone-receptor complexes formed by the hLHR-wt or the L457R mutant, the degradation of the internalized hormone, and the loss of cell surface receptors. The degradation of the hormone internalized by these mutants as well as the loss of cell surface receptors is largely prevented by leupeptin. These results highlight a previously unrecognized complexity in the postendocytotic trafficking of the hLHR and document a clear difference between the properties of the constitutively active mutant and the agonist-activated hLHR-wt. This lack of lysosomal degradation of the L457R mutant could contribute to its constitutive activity by prolonging the duration of signaling.


Asunto(s)
Lisosomas/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , Sustitución de Aminoácidos , Animales , Antígenos de Superficie/metabolismo , Células Cultivadas , Gonadotropina Coriónica/farmacocinética , Endocitosis/fisiología , Humanos , Ratones , Proteínas Mutantes/metabolismo , Desnaturalización Proteica , Transporte de Proteínas , Transfección
14.
Mol Endocrinol ; 20(3): 619-30, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16293639

RESUMEN

We show that activation of the endogenous or recombinant lutropin/choriogonadotropin receptor (LHR) in mouse Leydig tumor cells (MA-10 cells) leads to the tyrosine phosphorylation of the focal adhesion kinase (FAK) and one of its substrates (paxillin). Using specific antibodies to the five tyrosine residues of FAK that become phosphorylated, we show that activation of the LHR increases the phosphorylation of Tyr576 and Tyr577, but it does not affect the phosphorylation of Tyr397, Tyr861, or Tyr925. Because FAK is a prominent substrate for the Src family of tyrosine kinases (SFKs) we tested for their involvement in the LHR-mediated phosphorylation of FAK-Tyr576. Src is not detectable in MA-10 cells, but two other prominent members of this family (Fyn and Yes) are present. The LHR-mediated phosphorylation of FAK-Tyr576 is readily inhibited by PP2 (a pharmacological inhibitor of SFKs) and by dominant-negative mutants of SKFs. Moreover, activation of the LHR in MA-10 cells results in the stimulation of the activity of Fyn and Yes, and overexpression of either of these two tyrosine kinases enhances the LHR-mediated phosphorylation of FAK-Tyr576. Studies involving activation of other G protein-coupled receptors, overexpression of the different Galpha-subunits, and the use of second messenger analogs suggest that the LHR-induced phosphorylation of FAK-Tyr576 in MA-10 cells is mediated by SFKs, and that this family of kinases is, in turn, independently or cooperatively activated by the LHR-induced stimulation of Gs and Gq/11-mediated pathways.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Receptores de HL/metabolismo , Tirosina/metabolismo , Familia-src Quinasas/metabolismo , Animales , Gonadotropina Coriónica/farmacología , Inhibidores Enzimáticos/farmacología , Quinasa 1 de Adhesión Focal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/genética , Proteínas de Unión al GTP/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Humanos , Tumor de Células de Leydig/metabolismo , Masculino , Ratones , Paxillin/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas Proto-Oncogénicas c-yes/metabolismo , Pirimidinas/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Neoplasias Testiculares/metabolismo , Células Tumorales Cultivadas , Familia-src Quinasas/antagonistas & inhibidores
15.
Endocrinology ; 147(7): 3419-27, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16614081

RESUMEN

We show that activation of the recombinant lutropin/choriogonadotropin receptor (LHR) in mouse Leydig tumor cells (MA-10 cells) leads to the tyrosine phosphorylation of Shc (Src homology and collagen homology) and the formation of complexes containing Shc and Sos (Son of sevenless), a guanine nucleotide exchange factor for Ras. Because a dominant-negative mutant of Shc inhibits the LHR-mediated activation of Ras and the phosphorylation of ERK1/2, we conclude that the LHR-mediated phosphorylation of ERK1/2 is mediated, at least partially, by the classical pathway used by growth factor receptors. We also show that the endogenous epidermal growth factor receptor (EGFR) present in MA-10 cells is phosphorylated upon activation of the LHR. The LHR-mediated phosphorylation of the EGFR and Shc, the activation of Ras, and the phosphorylation of ERK1/2 are inhibited by expression of a dominant-negative mutant of Fyn, a member of the Src family kinases (SFKs) expressed in MA-10 cells and by PP2, a pharmacological inhibitor of the SFKs. These are also inhibited, but to a lesser extent, by AG1478, an inhibitor of the EGFR kinase. We conclude that the SFKs are responsible for the LHR-mediated phosphorylation of the EGFR and Shc, the formation of complexes containing Shc and Sos, the activation of Ras, and the phosphorylation of ERK1/2.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores de HL/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Proteínas ras/metabolismo , Animales , Línea Celular , Gonadotropina Coriónica/metabolismo , Activación Enzimática , Genes Dominantes , Humanos , Ratones , Quinazolinas , Tirfostinos/farmacología
16.
Mol Endocrinol ; 19(5): 1263-76, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15677709

RESUMEN

The high degree of amino acid sequence homology and the divergent ligand binding affinities of the rat (r) and human (h) LH receptors (LHRs) allowed us to identify amino acid residues of their extracellular domain that are responsible for the different binding affinities of bovine (b) and hLH, and human choriogonadotropin (hCG) to the hLHR and rLHR. Because of the proposed importance of the beta-sheets of the leucine-rich repeats (LRRs) of the extracellular domain of the LHR on hormone binding, we examined 10 divergent residues present in these regions by analyzing two complementary sets of mutants in which hLHR residues were substituted with the corresponding rLHR residues and vice versa. These experiments resulted in the identification of a single residue (a Ile or Ser in the C-terminal end of LRR2 of the hLHR or rLHR, respectively) that is important for hLH binding affinity. Surprisingly, however, this residue does not affect hCG or for bLH binding affinity. In fact, the results obtained with bLH and hCG show that several of the divergent residues in the beta-sheets of LRR1-9 affect bLH binding affinity, but none of them affect hCG binding affinity. Importantly, our results also emphasize the involvement of residues outside of the beta-sheets of the LRRs of the LHR in ligand binding affinity. This finding has to be considered in future models of the interaction of LH/CG with the LHR.


Asunto(s)
Aminoácidos/metabolismo , Gonadotropina Coriónica/metabolismo , Hormona Luteinizante/metabolismo , Receptores de HL/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Mutación , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Receptores de HL/genética
17.
Endocrinology ; 146(9): 3907-16, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15919743

RESUMEN

Signaling pathways mediating the divergent effects of FSH and LH on aromatase in immature rat granulosa cells were studied by infecting cells with increasing amounts of adenoviral vectors for the human LH receptor (hLHR) or FSH receptor (hFSHR). Increasing amounts of Ad-hLHR, used at a multiplicity of infection (MOI) of 20 or 200 viable viral particles/cell, increased human chorionic gonadotropin (hCG) binding and hCG-induced cAMP and Akt phosphorylation, but inositol phosphates only increased in response to hCG in cells infected with 200 MOI Ad-hLHR. In contrast, hCG increased aromatase expression in cells infected with 20, but not in cells infected with 200, MOI Ad-hLHR. Cells infected with 20 or 200 MOI Ad-hFSHR showed increased hFSH binding and hFSH-induced Akt phosphorylation, but the hFSH-induced cAMP response was unchanged relative to control cells. However, hFSH was able to stimulate the inositol phosphate cascade in the Ad-hFSHR-infected cells, and the hFSH induction of aromatase was abolished. We also found that activation of C kinase or expression of a constitutively active form of Galphaq inhibited the induction of aromatase by hFSH or 8Br-cAMP. We conclude that the differential effects of FSH and LH on aromatase in immature granulosa cells are highly dependent on gonadotropin receptor density and on the signaling pathways activated. We propose that aromatase is induced by common signals generated by activation of the FSHR and LHR (possibly cAMP and Akt) and that the activation of the inositol phosphate cascade in cells expressing a high density of LHR or FSHR antagonizes this induction.


Asunto(s)
Aromatasa/genética , Células de la Granulosa/enzimología , Receptores de HFE/metabolismo , Receptores de HL/metabolismo , Adenoviridae/genética , Factores de Edad , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Vectores Genéticos , Células de la Granulosa/citología , Células de la Granulosa/fisiología , Fosfatos de Inositol/metabolismo , Radioisótopos de Yodo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Sprague-Dawley
18.
Mol Endocrinol ; 17(11): 2189-200, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12920236

RESUMEN

The pathways involved in activation of the ERK1/2 cascade in Leydig cells were examined in MA-10 cells expressing the recombinant human LH receptor (hLHR) and in primary cultures of rat Leydig cell precursors. In MA-10 cells expressing the recombinant hLHR, human choriogonadotropin-induced activation of ERK1/2 is effectively inhibited by overexpression of a cAMP phosphodiesterase (a manipulation that blunts the human choriogonadotropin-induced cAMP response), by addition of H89 (a selective inhibitor of protein kinase A), or by overexpression of the heat-stable protein kinase A inhibitor, but not by overexpression of an inactive mutant of this inhibitor. Stimulation of hLHR did not activate Rap1, but activated Ras in an H89-sensitive fashion. Addition of H89 to MA-10 cells that had been cotransfected with a guanosine triphosphatase-deficient mutant of Ras almost completely inhibited the hLHR-mediated activation of ERK1/2. We also show that 8-bromo-cAMP activates Ras and ERK1/2 in MA-10 cells and in primary cultures of rat Leydig cells precursors in an H89-sensitive fashion, whereas a cAMP analog 8-(4-chloro-phenylthio)-2'-O-methyl-cAMP (8CPT-2Me-cAMP) that is selective for cAMP-dependent guanine nucleotide exchange factor has no effect. Collectively, our results show that the hLHR-induced phosphorylation of ERK1/2 in Leydig cells is mediated by a protein kinase A-dependent activation of Ras.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Intersticiales del Testículo/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores de HL/metabolismo , Proteínas ras/metabolismo , Animales , Arginina Vasopresina/farmacología , Células Cultivadas , Gonadotropina Coriónica/metabolismo , Gonadotropina Coriónica/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Activación Enzimática , Humanos , Fosfatos de Inositol/metabolismo , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/enzimología , Masculino , Ratones , Fosforilación , Ratas , Sistemas de Mensajero Secundario/efectos de los fármacos , Transducción de Señal , Proteínas de Unión al GTP rap/metabolismo
19.
Mol Endocrinol ; 18(2): 434-46, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14605099

RESUMEN

Mutants of the human (h) lutropin receptor (LHR) were analyzed using a combination of biochemical and imaging approaches to define motifs that participate in the postendocytotic sorting of this G protein-coupled receptor (GPCR). We show that a substantial portion of the human chorionic gonadotropin internalized by the hLHR sorts to a recycling pathway, and the internalized hLHR accumulates in endosomes because of the C-terminal cysteine (Cys(699)) and an upstream Leu(683) present in the hLHR. The removal or simultaneous mutation of these two residues reroutes the internalized human chorionic gonadotropin to a degradation pathway and the internalized hLHR to lysosomes. We also show that grafting the 17 C-terminal residues of the hLHR into the C-terminal tail of two GPCRs that are routed to a lysosomal/degradation pathway (the rat LHR or the murine delta opioid receptor) reroutes them to an endosomal/recycling pathway. This is due to the Leu(683) and Cys(699) combination and another recycling motif (Gly(687)Thr(688)) that was previously identified in the hLHR. The importance of both motifs can be readily ascertained in the context of a murine delta opioid receptor/hLHR chimera. The importance of the Gly(687)Thr(688) motif is revealed mostly in the context of a rat LHR/hLHR chimera. These studies define a novel, noncontiguous, transferable motif that participates in the sorting of internalized GPCRs.


Asunto(s)
Secuencias de Aminoácidos , Cisteína , Endocitosis/fisiología , Leucina , Receptores de HL/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Gonadotropina Coriónica/metabolismo , Gonadotropina Coriónica/farmacología , Endosomas/metabolismo , Humanos , Lisosomas/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación , Transporte de Proteínas/efectos de los fármacos , Ratas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de HL/efectos de los fármacos , Receptores de HL/genética , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal
20.
Mol Endocrinol ; 17(11): 2162-76, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12907758

RESUMEN

Although the fates of the internalized hormone-receptor complexes formed by the lutropin/choriogonadotropin and the TSH receptors have been examined in some detail, much less is known about the fate of the internalized FSH-FSH receptor (FSHR) complex. Using biochemical and imaging approaches we show here that the majority of the internalized FSH-FSHR complex accumulates in endosomes and subsequently recycles back to the cell surface where the bound, intact hormone dissociates back into the medium. Only small amounts of FSH and the FSHR are routed to a lysosomal degradation pathway, and the extent of FSH-induced down-regulation of the cell surface and total FSHR is minimal. This pathway was detected in heterologous (human kidney 293T) cells transfected with the rat (r) or human (h) FSHR as well as in a mouse Sertoli cell line (MSC-1) or a mouse granulosa cell line (KK-1) transfected with the rFSHR.Additional experiments using a series of C-terminal deletions of the rFSHR and the hFSHR showed that the recycling of the internalized FSH-FSHR complex and the extent of hFSH-induced down-regulation is dictated by a short stretch of amino acids present at the extreme C-terminal end of the receptor.We conclude that most of the internalized FSH-FSHR complex is recycled back to the cell surface, that this recycling pathway is highly dependent on amino acid residues present near the C terminus of the FSHR, and that it is an important determinant of the extent of down-regulation of the FSHR.


Asunto(s)
Endocitosis , Hormona Folículo Estimulante/metabolismo , Receptores de HFE/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Gonadotropina Coriónica/metabolismo , Regulación hacia Abajo , Humanos , Riñón/citología , Riñón/metabolismo , Sustancias Macromoleculares , Masculino , Ratones , Datos de Secuencia Molecular , Unión Proteica , Transporte de Proteínas , Ratas , Receptores de HFE/agonistas , Receptores de HFE/química , Receptores de HFE/genética , Receptores de Gonadotropina/metabolismo , Eliminación de Secuencia , Células de Sertoli/citología , Células de Sertoli/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA