Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Cell Sci ; 129(15): 2983-96, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27335427

RESUMEN

Preovulatory granulosa cells express the low-molecular-mass MAP2D variant of microtubule-associated protein 2 (MAP2). Activation of the luteinizing hormone choriogonadotropin receptor by human choriogonadotropin (hCG) promotes dephosphorylation of MAP2D on Thr256 and Thr259. We sought to evaluate the association of MAP2D with the cytoskeleton, and the effect of hCG on this association. MAP2D partially colocalized, as assessed by confocal immunofluorescence microscopy, with the vimentin intermediate filament and microtubule cytoskeletons in naive cells. In vitro binding studies showed that MAP2D bound directly to vimentin and ß-tubulin. Phosphorylation of recombinant MAP2D on Thr256 and Thr259, which mimics the phosphorylation status of MAP2D in naive cells, reduces binding of MAP2D to vimentin and tubulin by two- and three-fold, respectively. PKA-dependent phosphorylation of vimentin (Ser32 and Ser38) promoted binding of vimentin to MAP2D and increased contraction of granulosa cells with reorganization of vimentin filaments and MAP2D from the periphery into a thickened layer surrounding the nucleus and into prominent cellular extensions. Chemical disruption of vimentin filament organization increased progesterone production. Taken together, these results suggest that hCG-stimulated dephosphorylation of MAP2D at Thr256 and Thr259, phosphorylation of vimentin at Ser38 and Ser72, and the resulting enhanced binding of MAP2D to vimentin might contribute to the progesterone synthetic response required for ovulation.


Asunto(s)
Células de la Granulosa/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Ovulación , Vimentina/metabolismo , Animales , Gonadotropina Coriónica/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células de la Granulosa/efectos de los fármacos , Humanos , Filamentos Intermedios/efectos de los fármacos , Filamentos Intermedios/metabolismo , Microscopía Confocal , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Persona de Mediana Edad , Proteínas Mutantes/metabolismo , Ovulación/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosfotreonina/metabolismo , Progesterona/farmacología , Unión Proteica/efectos de los fármacos , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Solubilidad
2.
J Biol Chem ; 291(23): 12145-60, 2016 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-27080258

RESUMEN

Within the ovarian follicle, immature oocytes are surrounded and supported by granulosa cells (GCs). Stimulation of GCs by FSH leads to their proliferation and differentiation, events that are necessary for fertility. FSH activates multiple signaling pathways to regulate genes necessary for follicular maturation. Herein, we investigated the role of Y-box-binding protein-1 (YB-1) within GCs. YB-1 is a nucleic acid binding protein that regulates transcription and translation. Our results show that FSH promotes an increase in the phosphorylation of YB-1 on Ser(102) within 15 min that is maintained at significantly increased levels until ∼8 h post treatment. FSH-stimulated phosphorylation of YB-1(Ser(102)) is prevented by pretreatment of GCs with the PKA-selective inhibitor PKA inhibitor (PKI), the MEK inhibitor PD98059, or the ribosomal S6 kinase-2 (RSK-2) inhibitor BI-D1870. Thus, phosphorylation of YB-1 on Ser(102) is PKA-, ERK-, and RSK-2-dependent. However, pretreatment of GCs with the protein phosphatase 1 (PP1) inhibitor tautomycin increased phosphorylation of YB-1(Ser(102)) in the absence of FSH; FSH did not further increase YB-1(Ser(102)) phosphorylation. This result suggests that the major effect of RSK-2 is to inhibit PP1 rather than to directly phosphorylate YB-1 on Ser(102) YB-1 coimmunoprecipitated with PP1ß catalytic subunit and RSK-2. Transduction of GCs with the dephospho-adenoviral-YB-1(S102A) mutant prevented the induction by FSH of Egfr, Cyp19a1, Inha, Lhcgr, Cyp11a1, Hsd17b1, and Pappa mRNAs and estradiol-17ß production. Collectively, our results reveal that phosphorylation of YB-1 on Ser(102) via the ERK/RSK-2 signaling pathway is necessary for FSH-mediated expression of target genes required for maturation of follicles to a preovulatory phenotype.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hormona Folículo Estimulante/farmacología , Células de la Granulosa/efectos de los fármacos , Proteína 1 de Unión a la Caja Y/metabolismo , Animales , Aromatasa/genética , Western Blotting , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptores ErbB/genética , Estradiol/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Femenino , Expresión Génica/efectos de los fármacos , Células de la Granulosa/metabolismo , Mutación , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Serina/genética , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Proteína 1 de Unión a la Caja Y/genética
3.
J Biol Chem ; 291(9): 4547-60, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26702053

RESUMEN

The ubiquitous phosphatidylinositol 3-kinase (PI3K) signaling pathway regulates many cellular functions. However, the mechanism by which G protein-coupled receptors (GPCRs) signal to activate PI3K is poorly understood. We have used ovarian granulosa cells as a model to investigate this pathway, based on evidence that the GPCR agonist follicle-stimulating hormone (FSH) promotes the protein kinase A (PKA)-dependent phosphorylation of insulin receptor substrate 1 (IRS1) on tyrosine residues that activate PI3K. We report that in the absence of FSH, granulosa cells secrete a subthreshold concentration of insulin-like growth factor-1 (IGF-1) that primes the IGF-1 receptor (IGF-1R) but fails to promote tyrosine phosphorylation of IRS1. FSH via PKA acts to sensitize IRS1 to the tyrosine kinase activity of the IGF-1R by activating protein phosphatase 1 (PP1) to promote dephosphorylation of inhibitory Ser/Thr residues on IRS1, including Ser(789). Knockdown of PP1ß blocks the ability of FSH to activate PI3K in the presence of endogenous IGF-1. Activation of PI3K thus requires both PKA-mediated relief of IRS1 inhibition and IGF-1R-dependent tyrosine phosphorylation of IRS1. Treatment with FSH and increasing concentrations of exogenous IGF-1 triggers synergistic IRS1 tyrosine phosphorylation at PI3K-activating residues that persists downstream through protein kinase B (AKT) and FOXO1 (forkhead box protein O1) to drive synergistic expression of genes that underlies follicle maturation. Based on the ability of GPCR agonists to synergize with IGFs to enhance gene expression in other cell types, PP1 activation to relieve IRS1 inhibition may be a more general mechanism by which GPCRs act with the IGF-1R to activate PI3K/AKT.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteína Fosfatasa 1/metabolismo , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Activación Enzimática , Femenino , Células de la Granulosa/citología , Humanos , Proteínas Sustrato del Receptor de Insulina/agonistas , Proteínas Sustrato del Receptor de Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/genética , Mutación , Fosfatidilinositol 3-Quinasa/química , Fosforilación , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/química , Proteína Fosfatasa 1/genética , Procesamiento Proteico-Postraduccional , Interferencia de ARN , Ratas Sprague-Dawley , Receptor IGF Tipo 1/agonistas , Receptor IGF Tipo 1/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Tirosina/metabolismo
4.
J Biol Chem ; 291(37): 19701-12, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27422819

RESUMEN

Within the ovarian follicle, granulosa cells (GCs) surround and support immature oocytes. FSH promotes the differentiation and proliferation of GCs and is essential for fertility. We recently reported that ERK activation is necessary for FSH to induce key genes that define the preovulatory GC. This research focused on the phosphoregulation by FSH of ERK within GCs. FSH-stimulated ERK phosphorylation on Thr(202)/Tyr(204) was PKA-dependent, but MEK(Ser(217)/Ser(221)) phosphorylation was not regulated; rather, MEK was already active. However, treatment of GCs with the EGF receptor inhibitor AG1478, a dominant-negative RAS, an Src homology 2 domain-containing Tyr phosphatase inhibitor (NSC 87877), or the MEK inhibitor PD98059 blocked FSH-dependent ERK(Thr(202)/Tyr(204)) phosphorylation, demonstrating the requirement for upstream pathway components. We hypothesized that FSH via PKA enhances ERK phosphorylation by inhibiting the activity of a protein phosphatase that constitutively dephosphorylates ERK in the absence of FSH, allowing MEK-phosphorylated ERK to accumulate in the presence of FSH because of inactivation of the phosphatase. GCs treated with different phosphatase inhibitors permitted elimination of both Ser/Thr and Tyr phosphatases and implicated dual specificity phosphatases (DUSPs) in the dephosphorylation of ERK. Treatment with MAP kinase phosphatase (MKP3, DUSP6) inhibitors increased ERK(Thr(202)/Tyr(204)) phosphorylation in the absence of FSH to levels comparable with ERK phosphorylated in the presence of FSH. ERK co-immunoprecipitated with Myc-FLAG-tagged MKP3(DUSP6). GCs treated with MKP3(DUSP6) inhibitors blocked and PKA inhibitors enhanced dephosphorylation of recombinant ERK2-GST in an in vitro phosphatase assay. Together, these results suggest that FSH-stimulated ERK activation in GCs requires the PKA-dependent inactivation of MKP3(DUSP6).


Asunto(s)
Fosfatasa 6 de Especificidad Dual/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/enzimología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfatasa 6 de Especificidad Dual/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Femenino , Flavonoides/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley
5.
J Biol Chem ; 291(53): 27160-27169, 2016 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-27856640

RESUMEN

G protein-coupled receptors (GPCRs) activate PI3K/v-AKT thymoma viral oncoprotein (AKT) to regulate many cellular functions that promote cell survival, proliferation, and growth. However, the mechanism by which GPCRs activate PI3K/AKT remains poorly understood. We used ovarian preantral granulosa cells (GCs) to elucidate the mechanism by which the GPCR agonist FSH via PKA activates the PI3K/AKT cascade. Insulin-like growth factor 1 (IGF1) is secreted in an autocrine/paracrine manner by GCs and activates the IGF1 receptor (IGF1R) but, in the absence of FSH, fails to stimulate YXXM phosphorylation of IRS1 (insulin receptor substrate 1) required for PI3K/AKT activation. We show that PKA directly phosphorylates the protein phosphatase 1 (PP1) regulatory subunit myosin phosphatase targeting subunit 1 (MYPT1) to activate PP1 associated with the IGF1R-IRS1 complex. Activated PP1 is sufficient to dephosphorylate at least four IRS1 Ser residues, Ser318, Ser346, Ser612, and Ser789, and promotes IRS1 YXXM phosphorylation by the IGF1R to activate the PI3K/AKT cascade. Additional experiments indicate that this mechanism also occurs in breast cancer, thyroid, and preovulatory granulosa cells, suggesting that the PKA-dependent dephosphorylation of IRS1 Ser/Thr residues is a conserved mechanism by which GPCRs signal to activate the PI3K/AKT pathway downstream of the IGF1R.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Células Cultivadas , Femenino , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Humanos , Folículo Ovárico/citología , Folículo Ovárico/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología
6.
Cell Microbiol ; 16(9): 1441-55, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24779413

RESUMEN

Bacterial pathogens can induce an inflammatory response from epithelial tissues due to secretion of the pro-inflammatory chemokine interleukin-8 (IL-8). Many bacterial pathogens manipulate components of the focal complex (FC) to induce signalling events in host cells. We examined the interaction of several bacterial pathogens with host cells, including Campylobacter jejuni, to determine if the FC is required for induction of chemokine signalling in response to bacterial pathogens. Our data indicate that secretion of IL-8 is triggered by C. jejuni, Helicobacter pylori and Salmonella enterica serovar Typhimurium in response to engagement of ß1 integrins. Additionally, we found that the secretion of IL-8 from C. jejuni infected epithelial cells requires FAK, Src and paxillin, which in turn are necessary for Erk 1/2 recruitment and activation. Targeting the FC component paxillin with siRNA prevented IL-8 secretion from cells infected with several bacterial pathogens, including C. jejuni, Helicobacter pylori, Salmonella enterica serovar Typhimurium, Staphylococcus aureus, Pseudomonas aeruginosa, and Vibrio parahaemolyticus. Our findings indicate that maximal IL-8 secretion from epithelial cells in response to bacterial infection is dependent on the FC. Based on the commonality of the host response to bacterial pathogens, we propose that the FC is a signalling platform for an epithelial cell response to pathogenic organisms.


Asunto(s)
Campylobacter jejuni/inmunología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Infecciones por Bacterias Gramnegativas/inmunología , Interleucina-8/inmunología , Células CACO-2 , Línea Celular , Infecciones por Bacterias Gramnegativas/microbiología , Helicobacter pylori/fisiología , Humanos , Cadenas beta de Integrinas/metabolismo , Staphylococcus aureus/fisiología
7.
Proc Natl Acad Sci U S A ; 109(44): E2979-88, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23045700

RESUMEN

Controlled maturation of ovarian follicles is necessary for fertility. Follicles are restrained at an immature stage until stimulated by FSH secreted by pituitary gonadotropes. FSH acts on granulosa cells within the immature follicle to inhibit apoptosis, promote proliferation, stimulate production of steroid and protein hormones, and induce ligand receptors and signaling intermediates. The phosphoinositide 3-kinase (PI3K)/AKT (protein kinase B) pathway is a pivotal signaling corridor necessary for transducing the FSH signal. We report that protein kinase A (PKA) mediates the actions of FSH by signaling through multiple targets to activate PI3K/AKT. PKA uses a route that promotes phosphorylation of insulin receptor substrate-1 (IRS-1) on Tyr(989), a canonical binding site for the 85-kDa regulatory subunit of PI3K that allosterically activates the catalytic subunit. PI3K activation leads to activation of AKT through phosphorylation of AKT on Thr(308) and Ser(473). The adaptor growth factor receptor bound protein 2-associated binding protein 2 (GAB2) is present in a preformed complex with PI3K heterodimer and IRS-1, it is an A-kinase anchoring protein that binds the type I regulatory subunit of PKA, and it is phosphorylated by PKA on Ser(159). Overexpression of GAB2 enhances FSH-stimulated AKT phosphorylation. GAB2, thus, seems to coordinate signals from the FSH-stimulated rise in cAMP that leads to activation of PI3K/AKT. The ability of PKA to commandeer IRS-1 and GAB2, adaptors that normally integrate receptor/nonreceptor tyrosine kinase signaling into PI3K/AKT, reveals a previously unrecognized route for PKA to activate a pathway that promotes proliferation, inhibits apoptosis, enhances translation, and initiates differentiation of granulosa cells.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hormona Folículo Estimulante/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales , Regulación Alostérica , Animales , Dominio Catalítico , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Activación Enzimática , Femenino , Fosforilación , Ratas , Ratas Sprague-Dawley
8.
Mol Endocrinol ; 22(7): 1695-710, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18467524

RESUMEN

The actions of LH to induce ovulation and luteinization of preovulatory follicles are mediated principally by activation of cAMP-dependent protein kinase (PKA) in granulosa cells. PKA activity is targeted to specific locations in many cells by A kinase-anchoring proteins (AKAPs). We previously showed that FSH induces expression of microtubule-associated protein (MAP) 2D, an 80-kDa AKAP, in rat granulosa cells, and that MAP2D coimmunoprecipitates with PKA-regulatory subunits in these cells. Here we report a rapid and targeted dephosphorylation of MAP2D at Thr256/Thr259 after treatment with human chorionic gonadotropin, an LH receptor agonist. This event is mimicked by treatment with forskolin or a cAMP analog and is blocked by the PKA inhibitor myristoylated-PKI, indicating a role for cAMP and PKA signaling in phosphoregulation of granulosa cell MAP2D. Furthermore, we show that Thr256/Thr259 dephosphorylation is blocked by the protein phosphatase 2A (PP2A) inhibitor, okadaic acid, and demonstrate interactions between MAP2D and PP2A by coimmunoprecipitation and microcystin-agarose pull-down. We also show that MAP2D interacts with glycogen synthase kinase (GSK) 3beta and is phosphorylated at Thr256/Thr259 by this kinase in the basal state. Increased phosphorylation of GSK3beta at Ser9 and the PP2A B56delta subunit at Ser566 is observed after treatment with human chorionic gonadotropin and appears to result in LH receptor-mediated inhibition of GSK3beta and activation of PP2A, respectively. Taken together, these results show that the phosphorylation status of the AKAP MAP2D is acutely regulated by LH receptor-mediated modulation of kinase and phosphatase activities via PKA.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células de la Granulosa/citología , Proteínas Asociadas a Microtúbulos/química , Ovario/citología , Receptores de HL/metabolismo , Animales , Catálisis , Dominio Catalítico , Femenino , Humanos , Hormona Luteinizante/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Ovario/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal
9.
Cell Signal ; 18(9): 1351-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16616457

RESUMEN

Follicle-stimulating hormone (FSH) is necessary and sufficient to induce maturation of ovarian follicles to a mature, preovulatory phenotype in the intact animal, resulting in the generation of mature eggs and production of estrogen. FSH accomplishes these actions by inducing a complex pattern of gene expression in target granulosa cells that is regulated by input from many different signaling cascades, including those for the extracellular regulated kinases (ERKs), p38 mitogen-activated protein kinases (MAPKs), and phosphatidylinositol-3 kinase (PI3K). The upstream kinase that appears to be responsible for initiating all of the signaling that regulates gene expression in these epithelial cells is protein kinase A (PKA). PKA not only signals to directly phosphorylate transcription factors like cAMP response element binding protein and to promote chromatin remodeling by phosphorylating histone H3, this versatile kinase also enhances the activity of the p38 MAPK, ERK, and PI3K pathways. Additionally, accumulating evidence suggests that activation of a single signaling cascade downstream of PKA is not sufficient to activate target gene expression. Rather, cross-talk between and among signaling cascades is required. We will review the signaling cascades activated by FSH in granulosa cells and how these cascades contribute to the regulation of select target gene expression.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hormona Folículo Estimulante/metabolismo , Regulación de la Expresión Génica , Células de la Granulosa/fisiología , Transducción de Señal/fisiología , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Activación Enzimática , Femenino , Factores de Transcripción Forkhead/metabolismo , Histonas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Endocrinology ; 158(7): 2043-2051, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28460125

RESUMEN

Protein kinase A (PKA) has recently been shown to mimic the actions of follicle-stimulating hormone (FSH) by activating signaling pathways that promote granulosa cell (GC) differentiation, such as phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK). We sought to elucidate the mechanism by which PKA, a Ser/Thr kinase, intersected the PI3K/AKT and MAPK/ERK pathways that are canonically activated by receptor tyrosine kinases (RTKs). Our results show that for both of these pathways, the RTK is active in the absence of FSH yet signaling down the pathways to commence transcriptional responses requires FSH-stimulated PKA activation. For both pathways, PKA initiates signaling by regulating the activity of a protein phosphatase (PP). For the PI3K/AKT pathway, PKA activates the Ser/Thr PP1 complexed with the insulinlike growth factor 1 receptor (IGF-1R) and insulin receptor substrate 1 (IRS1) to dephosphorylate Ser residues on IRS1, authorizing phosphorylation of IRS1 by the IGF-1R to activate PI3K. Treatment of GCs with FSH and exogenous IGF-1 initiates synergistic IRS1 Tyr phosphorylation and resulting gene activation. The mechanism by which PKA activates PI3K is conserved in preovulatory GCs, MCF7 breast cancer cells, and FRTL thyroid cells. For the MAPK/ERK pathway, PKA promotes inactivation of the MAPK phosphatase (MKP) dual specificity phosphatase (DUSP) MKP3/DUSP6 to permit MEK-phosphorylated ERK to accumulate downstream of the epidermal growth factor receptor. Thus, for the two central signaling pathways that regulate gene expression in GCs, FSH via PKA intersects canonical RTK-regulated signaling by modulating the activity of PPs.


Asunto(s)
Diferenciación Celular , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Células de la Granulosa/fisiología , Proteínas Tirosina Quinasas/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática , Femenino , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Células MCF-7 , Fosforilación , Ratas , Transducción de Señal/genética
11.
Sci Rep ; 6: 28132, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27324437

RESUMEN

Activation of protein kinase A (PKA) by follicle stimulating hormone (FSH) transduces the signal that drives differentiation of ovarian granulosa cells (GCs). An unresolved question is whether PKA is sufficient to initiate the complex program of GC responses to FSH. We compared signaling pathways and gene expression profiles of GCs stimulated with FSH or expressing PKA-CQR, a constitutively active mutant of PKA. Both FSH and PKA-CQR stimulated the phosphorylation of proteins known to be involved in GC differentiation including CREB, ß-catenin, AKT, p42/44 MAPK, GAB2, GSK-3ß, FOXO1, and YAP. In contrast, FSH stimulated the phosphorylation of p38 MAP kinase but PKA-CQR did not. Microarray analysis revealed that 85% of transcripts that were up-regulated by FSH were increased to a comparable extent by PKA-CQR and of the transcripts that were down-regulated by FSH, 76% were also down-regulated by PKA-CQR. Transcripts regulated similarly by FSH and PKA-CQR are involved in steroidogenesis and differentiation, while transcripts more robustly up-regulated by PKA-CQR are involved in ovulation. Thus, PKA, under the conditions of our experimental approach appears to function as a master upstream kinase that is sufficient to initiate the complex pattern of intracellular signaling pathway and gene expression profiles that accompany GC differentiation.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hormona Folículo Estimulante Humana/metabolismo , Células de la Granulosa/fisiología , Ovario/citología , Animales , Diferenciación Celular , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Femenino , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Mutación/genética , Ratas , Ratas Endogámicas , Transducción de Señal , Esteroides/metabolismo , beta Catenina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Mol Cell Endocrinol ; 434: 116-26, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27328024

RESUMEN

FSH promotes maturation of ovarian follicles. One pathway activated by FSH in granulosa cells (GCs) is phosphatidylinositol-3 kinase/AKT. The AKT target FOXO1 is reported to function primarily as a repressor of FSH genes, including Ccnd2 and Inha. Based on its broad functions in other tissues, we hypothesized that FOXO1 may regulate many more GC genes. We transduced GCs with empty adenovirus or constitutively active FOXO1 followed by treatment with FSH for 24 h, and conducted RNA deep sequencing. Results show that FSH regulates 3772 genes ≥2.0-fold; 60% of these genes are activated or repressed by FOXO1. Pathway Studio Analysis revealed enrichment of genes repressed by FOXO1 in metabolism, signaling, transport, development, and activated by FOXO1 in signaling, cytoskeletal functions, and apoptosis. Gene regulation was verified by q-PCR (eight genes) and ChIP analysis (two genes). We conclude that FOXO1 regulates the majority of FSH target genes in GCs.


Asunto(s)
Hormona Folículo Estimulante/farmacología , Redes Reguladoras de Genes/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Animales , Células Cultivadas , Femenino , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/citología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Proteínas del Tejido Nervioso/metabolismo , Ratas , Análisis de Secuencia de ARN/métodos
13.
Mol Endocrinol ; 16(2): 221-33, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11818496

RESUMEN

Pulsatile secretion of GnRH is the major regulator of gonadotropin (LH, FSH) gene expression and secretion. Recently, GnRH has been shown to rapidly stimulate the expression of early growth response protein-1 (Egr-1), a transcription factor that is essential for LHbeta gene expression in the pituitary. In this study, we examined the regulatory elements and signal transduction pathways by which GnRH regulates Egr-1 transcription. Deletion analysis of the murine Egr-1 promoter identified two regions (-370 to -342 and -116 to -73) that are critical for GnRH responsiveness in alphaT3 pituitary gonadotrope cells. The first region, which contains two serum response elements (SREs), contributed about 70-80% of GnRH inducibility, whereas the second region, which contains two SREs and one Ets binding site, conferred an additional 20-30% of activity. Mutations that abolish protein binding to these SREs and Ets binding sites completely eliminated GnRH-mediated transcriptional activation of the Egr-1 promoter. Mutation of cAMP response element reduced promoter activity by 40%. Using specific protein kinase inhibitors, GnRH stimulation of Egr-1 expression was found to be dependent on PKC/ERK pathways. In addition, GnRH activated p90 ribosomal S6 kinase, which has the potential to phosphorylate serum response factor and cAMP response element binding protein. We conclude that GnRH stimulation of Egr-1 gene expression requires several distinct SREs/Ets elements and a cAMP response element and is mediated via activation of PKC/ERK signaling pathways.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/farmacología , Proteínas Inmediatas-Precoces , Regiones Promotoras Genéticas/genética , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Animales , Sitios de Unión , Western Blotting , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz , Ensayo de Cambio de Movilidad Electroforética , Activación Enzimática/efectos de los fármacos , Indoles/farmacología , Maleimidas/farmacología , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Hipófisis/citología , Hipófisis/efectos de los fármacos , Hipófisis/enzimología , Hipófisis/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets , Proteínas Quinasas S6 Ribosómicas/metabolismo , Eliminación de Secuencia/genética , Elemento de Respuesta al Suero/genética , Factor de Respuesta Sérica/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/genética , Proteína Elk-1 con Dominio ets
14.
Endocrinology ; 143(8): 2986-94, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12130564

RESUMEN

LH receptor activation leads to the phosphorylation/activation of p42/44 MAPK in preovulatory granulosa cells. As the LH receptor can activate both adenylyl cyclase and phospholipase C, we hypothesized that the LH receptor could elicit phosphorylation of p42/44 MAPK through activation of protein kinase A (PKA) and/or protein kinase C (PKC). Preovulatory granulosa cells in serum-free primary cultures were treated with ovulatory concentrations of human chorionic gonadotropin (hCG), an LH receptor agonist, with or without various inhibitors. The PKA inhibitor H89 as well as the myristoylated PKA inhibitor peptide PKI strongly inhibited hCG-stimulated p42/44 MAPK phosphorylation, whereas the PKC inhibitor GF109203X had no effect on p42/44 MAPK phosphorylation. LH receptor-stimulated phosphorylation of cAMP response element-binding protein (CREB), histone H3, and MAPK kinase (MEK) was also strongly inhibited by H89 and not by GF109203X. The extent of PKC activation was assessed in preovulatory granulosa cells using three criteria: translocation of PKC isoforms to the membrane fraction, phosphorylation of a known PKC substrate, and autophosphorylation of PKC delta on an activation-related site. By all three criteria PKCs were partially activated before hCG stimulation, and hCG treatment failed to elicit further PKC activation, in vitro or in vivo. Taken together, these results indicate that, under primary culture conditions where physiological levels of signaling proteins are present, hCG signals to activate MEK, p42/44 MAPK, CREB, and histone H3 in a predominantly PKA-dependent and PKC-independent manner. Unexpectedly, PKCs were partially activated in the absence of LH receptor activation, and LH receptor activation did not elicit further detectable PKC activation.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas de la Membrana , Proteína Quinasa C/fisiología , Receptores de HL/fisiología , Animales , Células Cultivadas , Gonadotropina Coriónica/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Activación Enzimática , Glucosidasas , Histonas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Gene ; 323: 149-55, 2003 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-14659888

RESUMEN

Relaxin, a hormone in the insulin superfamily, is synthesized by the corpus luteum of the rat ovary. Expression of relaxin precursor mRNA in rats is sharply induced after day 10 of pregnancy and plateaus on days 15 to 20 (parturition occurs on day 23). In an effort to understand this induction, we cloned the gene and carried out promoter analyses by transient transfection and chromatin immunoprecipitation methods. The single gene is 2.9 kilobases and is composed of two exons and one intron. There are alternative splice acceptor sites, 3 base pairs apart, which account for the inclusion of an extra codon in about 10% of the transcripts. The induction of transcription by day 15 was observed by the binding of polymerase II and histone H3 acetylation at the promoter region. There is a functional STAT binding site, about 3.8 kb upstream from the transcriptional start site, that is occupied by STAT3 on day 6 of pregnancy, when relaxin expression is minimal; on day 15, when expression is maximal, STAT3 is replaced by STAT5a. These data are consistent with STAT5 playing a role in the induction of relaxin expression.


Asunto(s)
Genes/genética , Relaxina/genética , Animales , Secuencia de Bases , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Clonación Molecular , ADN/química , ADN/genética , Exones , Femenino , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Humanos , Intrones , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Datos de Secuencia Molecular , Pruebas de Precipitina/métodos , Embarazo , Precursores de Proteínas/genética , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Relaxina/metabolismo , Análisis de Secuencia de ADN , Sitio de Iniciación de la Transcripción , Transfección
16.
FEBS Lett ; 521(1-3): 3-8, 2002 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-12067715

RESUMEN

The luteinizing hormone/choriogonadotropin hormone receptor (LH/CG R) signals to regulate ovulation, corpus luteum formation, and fetal survival during pregnancy. Agonist binding to the LH/CG R is poorly reversible, emphasizing the importance of a cellular mechanism to temper signaling by a potentially persistently active receptor. Like other G protein-coupled receptors (GPCRs), signaling by this receptor is modulated by its binding of an arrestin. We have identified ADP ribosylation factor 6 (ARF6) as a protein whose activation state is regulated by the LH/CG R and which functions to regulate the availability of plasma membrane-docked arrestin 2 to this receptor. We hypothesize that ARF6 might also serve GPCRs other than the LH/CG R to regulate the availability of arrestin 2 for receptor desensitization.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas de Unión al GTP/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de HL/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/fisiología , Animales , Arrestinas/metabolismo , Gonadotropina Coriónica/metabolismo , Humanos , Hormona Luteinizante/metabolismo , Fosfoproteínas/metabolismo , Receptores de Superficie Celular/fisiología
17.
Mol Endocrinol ; 27(8): 1295-310, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23754802

RESUMEN

Ovarian follicles lacking FSH or FSH receptors fail to progress to a preovulatory stage, resulting in infertility. One hallmark of the preovulatory follicle is the presence of luteinizing hormone/choriogonadotropin receptors (LHCGR) on granulosa cells (GCs). However, the mechanisms by which FSH induces Lhcgr gene expression are poorly understood. Our results show that protein kinase A (PKA) and phosphoinositide 3-kinase (PI3K)/AKT pathways are required for FSH to activate both the murine Lhcgr-luciferase reporter and expression of Lhcgr mRNA in rat GCs. Based on results showing that an adenovirus (Ad) expressing a steroidogenic factor 1 (SF1) mutant that cannot bind ß-catenin abolished FSH-induced Lhcgr mRNA, we evaluated the role of ß-catenin in the regulation of Lhcgr gene expression. FSH promoted the PKA-dependent, PI3K-independent phosphorylation of ß-catenin on Ser552 and Ser665. FSH activated the ß-catenin/T-cell factor (TCF) artificial promoter-reporter TOPFlash via a PKA-dependent, PI3K-independent pathway, and dominant-negative (DN) TCF abolished FSH-activated Lhcgr-luciferase reporter and induction of Lhcgr mRNA. Microarray analysis of GCs treated with Ad-DN-TCF and FSH identified the Lhcgr as the most down-regulated gene. Chromatin immunoprecipitation results placed ß-catenin phosphorylated on Ser552 and Ser675 and SF1 on the Lhcgr promoter in FSH-treated GCs; TCF3 was constitutively associated with the Lhcgr promoter. Transduction with an Ad-phospho-ß-catenin mutant (Ser552/665/Asp) enhanced Lhcgr mRNA expression in FSH-treated cells greater than 3-fold. Finally, we identified a recognized PI3K/AKT target, forkhead box O1, as a negative regulator of Lhcgr mRNA expression. These results provide new understanding of the complex regulation of Lhcgr gene expression in GCs.


Asunto(s)
Células de la Granulosa/metabolismo , Folículo Ovárico/metabolismo , Receptores de Gonadotropina/metabolismo , Receptores de HL/metabolismo , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo , Femenino , Hormona Folículo Estimulante/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptores de Gonadotropina/biosíntesis , Receptores de Gonadotropina/genética , Receptores de HL/biosíntesis , Receptores de HL/genética , Transducción de Señal , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Proteína 1 Similar al Factor de Transcripción 7/metabolismo , Transfección , beta Catenina/metabolismo
18.
Mol Endocrinol ; 24(9): 1765-81, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20610540

RESUMEN

Activation of the LH receptor (LHR) on preovulatory granulosa cells stimulates the cAMP/protein kinase A (PKA) pathway to regulate expression of genes required for ovulation and luteinization. LHR signaling also initiates rearrangement of the actin cytoskeleton. Because disruption of the actin cytoskeleton has been causally linked to steroidogenesis in various cell models, we sought to identify the cellular mechanisms that may modulate reorganization of the actin cytoskeleton and to determine whether cytoskeletal reorganization is required for steroidogenesis. Herein we report that LHR signaling in preovulatory granulosa cells promotes rapid dephosphorylation of the actin-depolymerizing factor cofilin at Ser3 that is dependent on PKA. The LHR-stimulated dephosphorylation of cofilin(Ser3) switches on cofilin activity to bind actin filaments and enhance their dynamics. Basal phosphorylation of cofilin(Ser3) is mediated by active/GTP-bound Rho and downstream protein kinases; LHR signaling promotes a decrease in active/GTP-bound Rho by a PKA-dependent mechanism. LHR-dependent Rho inactivation and subsequent activation of cofilin does not involve ERK, epidermal growth factor receptor, or phosphatidylinositol 3-kinase pathways downstream of PKA. To understand the biological significance of cofilin activation, preovulatory granulosa cells were transduced with a mutant cofilin adenoviral vector in which Ser3 was mutated to Glu (S-E cofilin). Inactive S-E cofilin abolished LHR-mediated reorganization of the actin cytoskeleton and caused a 70% decrease in LHR-stimulated progesterone that is obligatory for ovulation. Taken together, these results show that LHR signaling via PKA activates a cofilin-regulated rearrangement of the actin cytoskeleton and that active cofilin is required to initiate progesterone secretion by preovulatory granulosa cells.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fase Folicular/metabolismo , Células de la Granulosa/enzimología , Progesterona/biosíntesis , Receptores de HL/metabolismo , Animales , Gonadotropina Coriónica/farmacología , AMP Cíclico/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Fase Folicular/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Humanos , Modelos Biológicos , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteínas de Unión al GTP rho/metabolismo
20.
Biol Reprod ; 80(6): 1282-92, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19176883

RESUMEN

Follicle-stimulating hormone (FSH) regulation of aromatase gene expression in vitro requires the transcriptional coactivator beta-catenin. To ascertain the physiological significance of beta-catenin in granulosa cells during folliculogenesis, mice homozygous for floxed alleles of beta-catenin were intercrossed with Amhr2cre mice. Conditional deletion of beta-catenin in 8-wk-old females occurred in derivatives of the Müllerian duct, granulosa cells and, surprisingly, in brain, pituitary, heart, liver, and tail. Female mice deficient for beta-catenin were infertile, despite reaching puberty and ovulating at the expected age, indications of apparently normal ovarian function. In contrast, their oviducts were grossly distended, with fewer but healthy oocytes. In addition, their uteri lacked implantation sites. Together, these two phenotypes could explain the complete loss of fertility. Nevertheless, although the ovary appeared normal, with serum estradiol concentrations in the normal range, there was marked animal-to-animal variation of mRNAs encoding beta-catenin and aromatase. Similarly, inhibin-alpha and luteinizing hormone receptor mRNAs varied considerably in whole ovaries, whereas pituitary Fshb mRNA was significantly reduced. Collectively, these features suggested cyclization recombination (CRE)-mediated recombination of beta-catenin may be unstable in proliferating granulosa cells, and therefore may mask the suspected steroidogenic requirement for beta-catenin. We tested this possibility by transducing primary cultures of granulosa cells from mice homozygous for floxed alleles of beta-catenin with a CRE-expressing adenovirus. Reduction of beta-catenin significantly compromised FSH stimulation of aromatase mRNA and subsequent production of estradiol. Collectively, these data suggest that FSH regulation of steroidogenesis requires beta-catenin, a role that remains hidden when tested through Amhr2cre-mediated recombination in vivo.


Asunto(s)
Trompas Uterinas/crecimiento & desarrollo , Infertilidad Femenina/metabolismo , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Útero/crecimiento & desarrollo , beta Catenina/metabolismo , Animales , Aromatasa/metabolismo , Proliferación Celular , Células Cultivadas , AMP Cíclico/metabolismo , Femenino , Hormona Folículo Estimulante/metabolismo , Hormonas Esteroides Gonadales/biosíntesis , Integrasas , Ratones , Ratones Transgénicos , Conductos Paramesonéfricos/crecimiento & desarrollo , Ovario/fisiología , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Recombinación Genética , Elementos de Respuesta , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA