Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Am J Physiol Cell Physiol ; 297(3): C591-600, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19587220

RESUMEN

Gonadotropin-releasing hormone (GnRH) acts via seven transmembrane receptors to stimulate gonadotropin secretion. Sustained stimulation desensitizes GnRH receptor (GnRHR)-mediated gonadotropin secretion, and this underlies agonist use in hormone-dependent cancers. Since type I mammalian GnRHR do not desensitize, agonist-induced internalization and downregulation may underlie desensitization of GnRH-stimulated gonadotropin secretion; however, research focus has recently shifted to anterograde trafficking, with the finding that human (h)GnRHR are mostly intracellular. Moreover, there is little direct evidence for agonist-induced trafficking of hGnRHR, and whether or not type I mammalian GnRHR show agonist-induced internalization is controversial. Here we use automated imaging to monitor expression and internalization of hemagglutinin (HA)-tagged hGnRHRs, mouse (m) GnRHR, Xenopus (X) GnRHRs, and chimeric receptors (hGnRHR with added XGnRHR COOH tails, h.XGnRHR) expressed by adenoviral transduction in HeLa cells. We find that agonists stimulate downregulation and/or internalization of mGnRHR and XGnRHR, that GnRH stimulates trafficking of hGnRHR and can stimulate internalization or downregulation of hGnRHR when steps are taken to increase cell surface expression (addition of the XGnRHR COOH tail or pretreatment with pharmacological chaperone). Agonist effects on internalization (of h.XGnRHR) and downregulation (of hGnRHR and h.XGnRHR) were not mimicked by a peptide antagonist and were prevented by a mutation that prevents GnRHR signaling, demonstrating dependence on receptor signaling as well as agonist occupancy. Thus agonist-induced internalization and downregulation of type I mammalian GnRHR occurs in HeLa cells, and we suggest that the high throughput imaging systems described here will facilitate study of the molecular mechanisms involved.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Receptores LHRH/genética , Receptores LHRH/metabolismo , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Buserelina/farmacología , Regulación hacia Abajo , Regulación de la Expresión Génica/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Células HeLa , Hemaglutininas , Humanos , Indoles/farmacología , Ratones , Piridinas/farmacología , Proteínas Recombinantes , Transducción de Señal , Xenopus
2.
Trends Endocrinol Metab ; 17(7): 276-83, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16890451

RESUMEN

Vast numbers of extracellular signalling molecules exert effects on their target cells by activation of a relatively limited number of mitogen-activated protein kinase (MAPK) cascades, raising the question of how specificity is achieved. To a large extent, this appears to be attributable to differences in kinetics and compartmentalization of MAPK protein activation that are dictated by MAPK-associated proteins serving as scaffolds, anchors, activators or effectors. Here, we review spatiotemporal aspects of signalling via the Ras-Raf-extracellular signal-regulated kinase pathway, emphasizing recent work on roles of arrestins as scaffolds and transducers for seven transmembrane receptor signalling.


Asunto(s)
Membrana Celular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Arrestinas/metabolismo , Citoplasma/metabolismo , Humanos , Cinética , Transducción de Señal/fisiología
3.
Trends Endocrinol Metab ; 17(8): 308-13, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16919966

RESUMEN

Many hormones, neurotransmitters and growth factors influence their target cells by activation of mitogen-activated protein kinase cascades. The consequences of such activation reflect not only the magnitude, but also the kinetics and cellular compartmentalization of kinase activity. Gonadotropin-releasing hormone (GnRH) receptors are seven-transmembrane receptors that have undergone a period of rapidly accelerated molecular evolution in which the advent of type I mammalian GnRH receptors has been associated with the loss of the carboxyl-terminal tail, a structure present in all other seven-transmembrane receptors. Here, we review spatiotemporal aspects of extracellular-signal-regulated kinase activation by gonadotropin-releasing hormone receptors, emphasizing how the absence or presence of the carboxyl-terminal tail dictates the receptors' ability to engage and signal via arrestins.


Asunto(s)
Compartimento Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Receptores LHRH/fisiología , Animales , Humanos , Cinética , Modelos Biológicos , Estructura Terciaria de Proteína , Receptores LHRH/química , Transducción de Señal
4.
J Endocrinol ; 191(3): 625-36, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17170220

RESUMEN

Gonadotropin-releasing hormone receptors (GnRHRs) are expressed in gonadotropes and several extra-pituitary sites. They are assumed to be cell surface proteins but the human (h) GnRHR lacks features favoring plasma membrane localization and receptor location varies with cell type. When expressed in mammary (MCF7) cells, cell surface hGnRHR binding was much lower than that of mouse and sheep GnRHRs (type I GnRHRs without C-terminal tails), Xenopus (X) and marmoset type II GnRHRs (type II GnRHRs with C-tails) or chimeric receptors (type I GnRHRs with added XGnRHR C-tails). hGnRHR binding was higher in alphaT4 (gonadotrope-derived) cells and was increased less by C-tail addition. Whole cell levels of tagged human, Xenopus and chimeric GnRHRs were comparable (Western blotting) and confocal microscopy revealed that the hGnRHR is primarily intracellular (distribution similar to the endoplasmic reticulum marker, calreticulin), whereas most XGnRHR is at the plasma membrane, and adding the C-tail increased cell surface hGnRHR levels. A membrane-permeant antagonist increased cell surface hGnRHR number (>4-fold, t1/2 = 4 h) and also increased hGnRHR signaling and hGnRHR-mediated inhibition of proliferation. A more rapid increase in hGnRHR binding occurred when the temperature was raised from 4 to 37 degrees C (>5-fold, t1/2 = 15 min) and this effect was prevented by mutation to prevent signaling. Thus, cell surface GnRHR expression depends on receptor and cell type and the hGnRHR is primarily an intracellular protein that traffics to the cell surface for signaling in MCF7 cells. Manipulations favoring such trafficking may facilitate selective targeting of extra-pituitary GnRHRs.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Líquido Intracelular/metabolismo , Receptores LHRH/genética , Animales , Western Blotting/métodos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Indoles/farmacología , Ratones , Microscopía Confocal , Mutación , Unión Proteica , Transporte de Proteínas , Piridinas/farmacología , Ensayo de Unión Radioligante , Receptores LHRH/análisis , Especificidad de la Especie , Tirotrofos/metabolismo , Transfección/métodos , Xenopus
5.
Endocrinology ; 145(8): 3594-602, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15059960

RESUMEN

Desensitization and internalization of G protein-coupled receptors can be mediated by phosphorylation within the C-terminal tail, facilitating beta-arrestin binding and targeting the receptor for internalization. Type II GnRH receptors (GnRH-Rs) show such regulation, but type I GnRH-Rs lack C-tails and are not rapidly desensitized or internalized. Here we show contrasting susceptibility of type I (human and sheep) and II (Xenopus) GnRH-Rs to regulation by protein kinase C (PKC). When human (h) or Xenopus (X) GnRH-Rs were expressed using recombinant adenovirus, PKC activation increased radioligand binding to XGnRH-Rs but not to hGnRH-Rs. A dominant-negative dynamin mutant (K44A) inhibited internalization of XGnRH-Rs (but not hGnRH-Rs) without influencing PKC regulation of XGnRH-R binding. PKC activation increased the affinity of XGnRH-Rs for the type II GnRH ligand and increased effects of low concentrations of GnRH-II on the [Ca(2+)](i) but had no effect on type I ligand binding to hGnRH-Rs, sGnRH-Rs or XGnRH-Rs, or to chimeric receptors with the XGnRH-R C-tail added to a type I receptor. Binding of type II ligand to human or sheep receptors was also unaffected but was increased in the chimeras. Mutation of both PKC-phosphorylation consensus sites in the XGnRH-R tail did not prevent the PKC-mediated increases in binding or alter agonist-induced translocation of beta-arrestin2/green fluorescent protein or inhibition of inositol phosphate accumulation by beta-arrestin2/green fluorescent protein. Thus, it appears that there are two distinct active conformations of XGnRH-Rs (differing in affinity for type I and II ligands) and that these cells exhibit a novel form of inside-out signaling in which PKC feeds back to influence receptor affinity.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Proteína Quinasa C/fisiología , Receptores LHRH/metabolismo , Transducción de Señal , Sitios de Unión , Células HeLa , Humanos , Fosforilación , Conformación Proteica , Receptores LHRH/química , Acetato de Tetradecanoilforbol/farmacología
6.
J Clin Endocrinol Metab ; 89(4): 1823-32, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15070951

RESUMEN

GnRH receptors (GnRH-Rs) mediate direct antiproliferative effects on hormone-dependent cancer cells. GnRH-Rs can be grouped according to ligand specificity (for GnRH-I and -II), and there is evidence that type II GnRH ligands and/or receptors can inhibit proliferation. Type I GnRH-Rs (e.g. human and sheep) lack the C-terminal tails found in other G protein-coupled receptors including type II GnRH-Rs (e.g. Xenopus; XGnRH-R). This underlies the remarkable resistance of type I GnRH-Rs to desensitization and may be important for chronic effects on proliferation. To test this, we have compared the antiproliferative effects of GnRH-Rs expressed in MCF7 breast cancer cells using recombinant adenovirus (Ad). Endogenous GnRH-Rs were not detected, but infection with Ad-expressing sheep GnRH-Rs (sGnRH-R) facilitated proliferation inhibition by Buserelin, and maximum inhibition required only 10,000-20,000 sGnRH-Rs. XGnRH-Rs were much less efficient at inhibiting proliferation and were internalized faster than sGnRH-Rs. Thus, the type II GnRH-R is less efficient at inhibiting proliferation, presumably because it is rapidly desensitized and/or internalized. Moreover, comparisons of human GnRH-R, sGnRH-R, and XGnRH-R, as well as chimeric receptors (type I GnRH-Rs with C-terminal tails from XGnRH-Rs), revealed that C-terminal tail addition increases receptor expression and thereby increases the efficiency with which the vector facilitates the antiproliferative effect.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Receptores LHRH/metabolismo , Transducción de Señal , Animales , Antineoplásicos Hormonales/farmacología , Buserelina/farmacología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Quimera , Femenino , Técnicas de Transferencia de Gen , Humanos , Conformación Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Receptores LHRH/genética , Ovinos , Especificidad de la Especie , Xenopus laevis
7.
Cell Signal ; 24(5): 1002-11, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22245064

RESUMEN

We have explored the possible role of dual specificity phosphatases (DUSPs) on acute EGF-mediated ERK signalling using high content imaging and a delayed MEK inhibition protocol to distinguish direct and indirect effects of the phosphatases on ERK activity. Using siRNAs, we were unable to find evidence that any of the MAPK phosphatases (MKPs) expressed in HeLa cells acts directly to dephosphorylate ppERK1/2 (dual phosphorylated ERKs 1 and/or 2) in the acute time-frame tested (0-14 min). Nevertheless, siRNAs against two p38/JNK MKPs (DUSPs 10 and 16) inhibited acute EGF-stimulated ERK activation. No such effect was seen for acute effects of the protein kinase C activator PDBu (phorbol 12,13 dibutyrate) on ERK activity, although effects of EGF and PDBu on ERK-dependent transcription (Egr-1 luciferase activity) were both reduced by siRNA targeting DUSPs 10 and 16. Inhibition of EGF-stimulated ERK activity by these siRNAs was reversed by pharmacological inhibition of p38 MAPK and single cell analysis revealed that the siRNAs did not influence the nuclear-cytoplasmic distribution of ppERK1/2. Thus, DUSPs 10 and 16 are positive regulators of activation, apparently acting by modulating cross-talk between the p38 and ERK pathways. A simplified mathematical model of this scenario accurately predicted the experimental data, supporting the conclusion that the major mechanism by which MKPs influence acute EGF-stimulated ERK responses is the negative regulation of p38, resulting in the positive regulation of ERK phosphorylation and activity.


Asunto(s)
Fosfatasas de Especificidad Dual/fisiología , Factor de Crecimiento Epidérmico/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Activación Enzimática , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional , Interferencia de ARN , Receptor Cross-Talk
8.
Mol Endocrinol ; 24(2): 423-35, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20009083

RESUMEN

Gonadotropin-releasing hormone acts via cell surface receptors but most human (h) GnRH receptors (GnRHRs) are intracellular. A membrane-permeant nonpeptide antagonist [(2S)-2-[5-[2-(2-axabicyclo[2.2.2]oct-2-yl)-1,1-dimethy-2-oxoethyl]-2-(3,5-dimethylphenyl)-1H-indol-3-yl]-N-(2-pyridin-4-ylethyl)propan-1-amine (IN3)] increases hGnRHR expression at the surface, apparently by facilitating its exit from the endoplasmic reticulum. Here we have quantified GnRHR by automated imaging in HeLa cells transduced with adenovirus expressing hemagglutinin-tagged GnRHR. Consistent with an intracellular site of action, IN3 increases cell surface hGnRHR, and this effect is not blocked or mimicked by membrane-impermeant peptide antagonists [Ac-D2Nal-D4Cpa-D3Pal-Ser-Tyr-d-Cit-Leu-Arg-Pro-d-Ala-NH(2) (cetrorelix) and antide]. However, when the C-terminal tail of a Xenopus (X) GnRHR was added (h.XGnRHR) to increase expression, both peptides further increased cell surface GnRHR. Cetrorelix also synergized with IN3 to increase expression of hGnRHR and a G-protein coupling-deficient mutant (A261K-hGnRHR). Cetrorelix also increased cell surface expression of hGnRHR, h.XGnRHR, and mouse GnRHR in gonadotrope-lineage LbetaT2 cells, and in HeLa cells it slowed h.XGnRHR internalization (measured by receptor-mediated antihemagglutinin uptake). Thus cetrorelix has effects other than GnRHR blockade; it acts as an inverse agonist in internalization assays, supporting the potential importance of ligand-biased efficacy at GnRHR. We also developed an imaging assay for GnRH function based on Ca(2+)-dependent nuclear translocation of a nuclear factor of activated T cells reporter. Using this in HeLa and LbetaT2 cells, IN3 and cetrorelix behaved as competitive antagonists when coincubated with GnRH, and long-term pretreatment (16 h) with IN3 reduced its effectiveness as an inhibitor whereas pretreatment with cetrorelix increased its inhibitory effect. This distinction between peptide and nonpeptide antagonists may prove important for therapeutic applications of GnRH antagonists.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Membrana Celular/metabolismo , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Antagonistas de Hormonas/farmacología , Indoles/farmacología , Péptidos/farmacología , Piridinas/farmacología , Receptores LHRH/antagonistas & inhibidores , Receptores LHRH/metabolismo , Animales , Buserelina/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Células HeLa , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Ligandos , Ratones , Microscopía Fluorescente , Oligopéptidos/farmacología , Transporte de Proteínas/efectos de los fármacos , Receptores LHRH/agonistas , Receptores LHRH/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Especificidad de la Especie
9.
J Endocrinol ; 196(2): 353-67, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18252959

RESUMEN

In heterologous expression systems, human GnRH receptors (hGnRHRs) are poorly expressed at the cell surface and this may reflect inefficient exit from the endoplasmic reticulum. Here, we have defined the proportion of GnRHRs at the cell surface using a novel assay based on adenoviral transduction with epitope-tagged GnRHRs followed by staining and semi-automated imaging. We find that in MCF7 (breast cancer) cells, the proportional cell surface expression (PCSE) of hGnRHRs is remarkably low (<1%), when compared with Xenopus laevis (X) GnRHRs ( approximately 40%). This distinction is retained at comparable whole cell expression levels, and the hGnRHR PCSE is increased by addition of the XGnRHR C-tail (h.XGnRHR) or by a membrane-permeant pharmacological chaperone (IN3). The IN3 effect is concentration- and time-dependent and IN3 also enhances the hGnRHR-mediated (but not h.XGnRHR- or mouse GnRHR-mediated) stimulation of [(3)H]inositol phosphate accumulation and the hGnRHR-mediated reduction in cell number. We also find that the PCSE for hGnRHRs and h.XGnRHRs is low and is greatly increased by IN3 in two hormone-dependent cancer lines, but is higher and less sensitive to IN3 in a gonadotrope line. Finally, we show that the effect of IN3 on hGnRHR PCSE is not mimicked or blocked by two peptide antagonists although they do increase the PCSE for h.XGnRHRs, revealing that an antagonist-occupied cell surface GnRHR conformation can differ from that of the unoccupied receptor. The low PCSE of hGnRHRs and this novel peptide antagonist effect may be important for understanding GnRHR function in extrapituitary sites.


Asunto(s)
Neoplasias de la Mama/metabolismo , Membrana Celular/metabolismo , Gonadotrofos/metabolismo , Antagonistas de Hormonas/farmacología , Neoplasias de la Próstata/metabolismo , Receptores LHRH/metabolismo , Animales , Neoplasias de la Mama/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular , Femenino , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/farmacología , Humanos , Indoles/farmacología , Masculino , Ratones , Oligopéptidos/farmacología , Neoplasias de la Próstata/patología , Piridinas/farmacología , Receptores LHRH/efectos de los fármacos , Receptores LHRH/genética , Transfección , Xenopus laevis
10.
J Biol Chem ; 281(5): 2701-10, 2006 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-16314413

RESUMEN

Activation of seven-transmembrane region receptors typically causes their phosphorylation with consequent arrestin binding and desensitization. Arrestins also act as scaffolds, mediating signaling to Raf and ERK and, for some receptors, inhibiting nuclear translocation of ERK. GnRH receptors (GnRHRs) act via Gq/11 to stimulate the phospholipase C/Ca2+/protein kinase C (PKC) cascade and the Raf/MEK/ERK cassette. Uniquely, type I mammalian GnRHRs lack the C-tails that are found in other seven-transmembrane region receptors (including nonmammalian GnRHRs) and are implicated in arrestin binding. Here we have compared ERK signaling by human GnRHRs (hGnRHRs) and Xenopus GnRHRs (XGnRHRs). In HeLa cells, XGnRHRs underwent rapid and arrestin-dependent internalization and caused arrestin/green fluorescent protein (GFP) translocation to the membrane and endosomes, whereas hGnRHRs did not. Internalized XGnRHRs were co-localized with arrestin-GFP, whereas hGnRHRs were not. Both receptors mediated transient ERK phosphorylation and nuclear translocation (revealed by immunohistochemistry or by imaging of co-transfected ERK2-GFP), and for both, ERK phosphorylation was reduced by PKC inhibition but not by inhibiting epidermal growth factor receptor autophosphorylation. In the presence of PKC inhibitor, Deltaarrestin-(319-418) blocked XGnRHR-mediated, but not hGnRHR-mediated, ERK phosphorylation. When receptor number was varied, hGnRHRs activated phospholipase C and ERK more efficiently than XGnRHRs but were less efficient at causing ERK2-GFP translocation. At high receptor number, XGnRHRs and hGnRHRs both caused ERK2-GFP translocation to the nucleus, but at low receptor number, XGnRHRs caused ERK2-GFP translocation, whereas hGnRHRs did not. Thus, experiments with XGnRHRs have revealed the first direct evidence of arrestin-mediated (probably G protein-independent) GnRHR signaling, whereas those with hGnRHRs imply that scaffolds other than arrestins can determine GnRHR effects on ERK compartmentalization.


Asunto(s)
Arrestina/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores LHRH/metabolismo , Animales , Compartimento Celular , Endocitosis , Activación Enzimática , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Fosforilación , Transporte de Proteínas , Transducción de Señal , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA