Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Pflugers Arch ; 470(6): 937-947, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29500670

RESUMEN

The G protein-regulated inducer of neurite growth (GRIN) family has three isoforms (GRIN1-3), which bind to the Gαi/o subfamily of G protein that mediate signal processing via G protein-coupled receptors (GPCRs). Here, we show that GRIN3 is involved in regulation of dopamine-dependent behaviors and is essential for activation of the dopamine receptors (DAR)-ß-arrestin signaling cascade. Analysis of functional regions of GRIN3 showed that a di-cysteine motif (Cys751/752) is required for plasma membrane localization. GRIN3 was co-immunoprecipitated with GPCR kinases 2/6 and ß-arrestins 1/2. Among GRINs, only GRIN3, which is highly expressed in striatum, strongly interacted with ß-arrestin 2. We also generated GRIN3-knockout mice (GRIN3KO). GRIN3KO exhibited reduced locomotor activity and increased anxiety-like behavior in the elevated maze test, as well as a reduced locomoter response to dopamine stimulation. We also examined the phosphorylation of Akt at threonine 308 (phospho308-Akt), which is dephosphorylated via a ß-arrestin 2-mediated pathway. Dephosphorylation of phospho308-Akt via the D2R-ß-arrestin 2 signaling pathway was completely abolished in striatum of GRIN3KO. Our results suggest that GRIN3 has a role in recruitment and assembly of proteins involved in ß-arrestin-dependent, G protein-independent signaling.


Asunto(s)
Ansiedad/metabolismo , Cuerpo Estriado/metabolismo , Proteínas del Tejido Nervioso/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Dopaminérgicos/metabolismo , Transducción de Señal , beta-Arrestinas/metabolismo , Animales , Sitios de Unión , Células HEK293 , Humanos , Locomoción , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Unión Proteica
2.
FASEB J ; 31(3): 910-919, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27881485

RESUMEN

Cell migration to chemoattractants is critically important in both normal physiology and the pathogenesis of many diseases. In GPCR-mediated chemotaxis, GPCRs transduce the gradient of an extracellular chemotactic ligand into intracellular responses via the activation of heterotrimeric G proteins. However, ligand-induced G-protein activation has not been directly imaged as yet in mammalian chemotaxing cells. We developed a Förster resonance energy transfer (FRET) probe, R10-Gi, by linking the Gi-protein α subunit to the regulator of G-protein signaling domain. The R10-Gi probe was coupled with a chemoattractant leukotriene B4 (LTB4) receptor 1 (BLT1) that induced the receptor to display a high-affinity ligand binding activity (Kd = 0.91 nM) in HEK293 cells. The R10-Gi probe exhibited an increased FRET signal in accord with the LTB4-dependent activation of Gi Furthermore, neutrophil-like differentiated human leukemia cell line 60 that expressed the intrinsic BLT1 displayed temporal Gi-protein activation in an area localized to the leading edge during chemotaxis in a shallow gradient of LTB4 These findings afford an opportunity to clarify the mechanisms underlying the subcellular regulation of Gi-protein activity, as well as GPCR-mediated ligand sensing, during chemotaxis in mammalian cells.-Masuda, K., Kitakami, J., Kozasa, T., Kodama, T., Ihara, S., Hamakubo, T. Visualization of ligand-induced Gi-protein activation in chemotaxing cells.


Asunto(s)
Quimiotaxis , Transferencia Resonante de Energía de Fluorescencia/métodos , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HL-60 , Humanos , Leucotrieno B4/metabolismo , Microscopía Fluorescente/métodos , Unión Proteica , Transporte de Proteínas , Proteínas RGS/genética , Proteínas RGS/metabolismo , Receptores de Leucotrieno B4/metabolismo
3.
Blood ; 123(3): 442-50, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24081657

RESUMEN

von Willebrand factor (vWF) secretion by endothelial cells (ECs) is essential for hemostasis and thrombosis; however, the molecular mechanisms are poorly understood. Interestingly, we observed increased bleeding in EC-Gα13(-/-);Gα12(-/-) mice that could be normalized by infusion of human vWF. Blood from Gα12(-/-) mice exhibited significantly reduced vWF levels but normal vWF multimers and impaired laser-induced thrombus formation, indicating that Gα12 plays a prominent role in EC vWF secretion required for hemostasis and thrombosis. In isolated buffer-perfused mouse lungs, basal vWF levels were significantly reduced in Gα12(-/-), whereas thrombin-induced vWF secretion was defective in both EC-Gαq(-/-);Gα11(-/-) and Gα12(-/-) mice. Using siRNA in cultured human umbilical vein ECs and human pulmonary artery ECs, depletion of Gα12 and soluble N-ethylmaleimide-sensitive-fusion factor attachment protein α (α-SNAP), but not Gα13, inhibited both basal and thrombin-induced vWF secretion, whereas overexpression of activated Gα12 promoted vWF secretion. In Gαq, p115 RhoGEF, and RhoA-depleted human umbilical vein ECs, thrombin-induced vWF secretion was reduced by 40%, whereas basal secretion was unchanged. Finally, in vitro binding assays revealed that Gα12 N-terminal residues 10-15 mediated the binding of Gα12 to α-SNAP, and an engineered α-SNAP binding-domain minigene peptide blocked basal and evoked vWF secretion. Discovery of obligatory and complementary roles of Gα12 and Gαq/11 in basal vs evoked EC vWF secretion may provide promising new therapeutic strategies for treatment of thrombotic disease.


Asunto(s)
Células Endoteliales/citología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Factor de von Willebrand/metabolismo , Animales , Anticuerpos Monoclonales/química , Regulación de la Expresión Génica , Hemostasis , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Adhesividad Plaquetaria , Unión Proteica , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida/química , Trombosis
4.
Mol Pharmacol ; 86(3): 252-62, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24958816

RESUMEN

Gastrin-releasing peptide receptor (GRPR) is ectopically expressed in over 60% of colon cancers. GRPR expression has been correlated with increased colon cancer cell migration. However, the signaling pathway by which GRPR activation leads to increased cancer cell migration is not well understood. We set out to molecularly dissect the GRPR signaling pathways that control colon cancer cell migration through regulation of small GTPase RhoA. Our results show that GRP stimulation activates RhoA predominantly through G13 heterotrimeric G-protein signaling. We also demonstrate that postsynaptic density 95/disk-large/ZO-1 (PDZ)-RhoGEF (PRG), a member of regulator of G-protein signaling (RGS)-homology domain (RH) containing guanine nucleotide exchange factors (RH-RhoGEFs), is the predominant activator of RhoA downstream of GRPR. We found that PRG is required for GRP-stimulated colon cancer cell migration, through activation of RhoA-Rho-associated kinase (ROCK) signaling axis. In addition, PRG-RhoA-ROCK pathway also contributes to cyclo-oxygenase isoform 2 (Cox-2) expression. Increased Cox-2 expression is correlated with increased production of prostaglandin-E2 (PGE2), and Cox-2-PGE2 signaling contributes to total GRPR-mediated cancer cell migration. Our analysis reveals that PRG is overexpressed in colon cancer cell lines. Overall, our results have uncovered a key mechanism for GRPR-regulated colon cancer cell migration through the Gα13-PRG-RhoA-ROCK pathway.


Asunto(s)
Neoplasias del Colon/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Bombesina/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Células CACO-2 , Movimiento Celular , Neoplasias del Colon/patología , Ciclooxigenasa 2/biosíntesis , Dinoprostona/biosíntesis , Homólogo 4 de la Proteína Discs Large , Células HT29 , Humanos , Estructura Terciaria de Proteína , Transducción de Señal , Quinasas Asociadas a rho/metabolismo
5.
J Biol Chem ; 288(47): 33978-33984, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24114843

RESUMEN

We previously characterized a Gα12-specific signaling pathway that stimulates the transcription of the E3 ligase RFFL via the protein kinase ARAF and ERK. This pathway leads to persistent PKC activation and is important for sustaining fibroblast migration. However, questions remain regarding how Gα12 specifically activates ARAF, which transcription factor is involved in Gα12-mediated RFFL expression, and whether RFFL is important for cell migration stimulated by other signaling mechanisms that can activate ERK. In this study, we show that replacement of the Gα12 residue Arg-264 with Gln, which is the corresponding Gα13 residue, abrogates the ability of Gα12 to interact with or activate ARAF. We also show that Gα12 can no longer interact with and activate an ARAF mutant with its C-terminal sequence downstream of the kinase domain being replaced with the corresponding CRAF sequence. These results explain why Gα12, but not Gα13, specifically activates ARAF but not CRAF. Together with our finding that recombinant Gα12 is sufficient for stimulating the kinase activity of ARAF, this study reveals an ARAF activation mechanism that is different from that of CRAF. In addition, we show that this Gα12-ARAF-ERK pathway stimulates RFFL transcription through the transcription factor c-Myc. We further demonstrate that EGF, which signals through CRAF, and an activated BRAF mutant also activate PKC and stimulate cell migration through up-regulating RFFL expression. Thus, RFFL-mediated PKC activation has a broad significance in cell migration regulation.


Asunto(s)
Movimiento Celular/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Ubiquitina-Proteína Ligasas/biosíntesis , Quinasas raf/metabolismo , Sustitución de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis , Activación Enzimática/fisiología , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Células HEK293 , Humanos , Ratones , Mutación Missense , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ubiquitina-Proteína Ligasas/genética , Quinasas raf/genética
6.
J Biol Chem ; 286(23): 20625-36, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21507947

RESUMEN

RH-RhoGEFs are a family of guanine nucleotide exchange factors that contain a regulator of G protein signaling homology (RH) domain. The heterotrimeric G protein Gα(13) stimulates the guanine nucleotide exchange factor (GEF) activity of RH-RhoGEFs, leading to activation of RhoA. The mechanism by which Gα(13) stimulates the GEF activity of RH-RhoGEFs, such as p115RhoGEF, has not yet been fully elucidated. Here, specific residues in Gα(13) that mediate activation of p115RhoGEF are identified. Mutation of these residues significantly impairs binding of Gα(13) to p115RhoGEF as well as stimulation of GEF activity. These data suggest that the exchange activity of p115RhoGEF is stimulated allosterically by Gα(13) and not through its interaction with a secondary binding site. A crystal structure of Gα(13) bound to the RH domain of p115RhoGEF is also presented, which differs from a previously crystallized complex with a Gα(13)-Gα(i1) chimera. Taken together, these data provide new insight into the mechanism by which p115RhoGEF is activated by Gα(13).


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/química , Factores de Intercambio de Guanina Nucleótido/química , Complejos Multienzimáticos/química , Regulación Alostérica/fisiología , Animales , Cristalografía por Rayos X , Activación Enzimática/fisiología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Ratones , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Mutación , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Factores de Intercambio de Guanina Nucleótido Rho , Relación Estructura-Actividad
7.
Nat Prod Res ; 36(6): 1552-1557, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33550857

RESUMEN

Manuka honey (MkH), derived from New Zealand manuka tree (Leptospermum scoparium), is considered a therapeutic agent owing to its antibacterial, antioxidant, antifungal, antiviral, anti-inflammatory, and wound healing activities. In this study, the inhibitory effect of five honey types, including MkH, on HIV-1 RT activity was evaluated, using an RT assay colorimetric kit, according to the manufacturer's instructions with slight modifications. MkH exerted the strongest inhibitory effect in a dose-dependent manner, with a half maximal inhibitory concentration (IC50) of approximately 14.8 mg/mL. Moreover, among the MkH constituents, methylglyoxal (MGO) and 2-methoxybenzoic acid (2-MBA) were determined to possess anti-HIV-1 RT activity. MGO and 2-MBA in MkH were identified by High Performance Liquid Chromatography (HPLC) and Liquid Chromatograph - Mass Spectrometry (LC-MS/MS). The findings suggest that the inhibitory effect of MkH on the HIV-1 RT activity is mediated by multiple constituents with different physical and chemical properties.


Asunto(s)
VIH-1 , Miel , Cromatografía Liquida , Miel/análisis , Humanos , ADN Polimerasa Dirigida por ARN , Espectrometría de Masas en Tándem
8.
Nat Cell Biol ; 5(12): 1095-103, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14634662

RESUMEN

G alpha 13 stimulates the guanine nucleotide exchange factors (GEFs) for Rho, such as p115Rho-GEF. Activated Rho induces numerous cellular responses, including actin polymerization, serum response element (SRE)-dependent gene transcription and transformation. p115Rho-GEF contains a Regulator of G protein Signalling domain (RGS box) that confers GTPase activating protein (GAP) activity towards G alpha 12 and G alpha 13 (ref. 3). In contrast, classical RGS proteins (such as RGS16 and RGS4) exhibit RGS domain-dependent GAP activity on G alpha i and G alpha q, but not G alpha 12 or G alpha 13 (ref 4). Here, we show that RGS16 inhibits G alpha 13-mediated, RhoA-dependent reversal of stellation and SRE activation. The RGS16 amino terminus binds G alpha 13 directly, resulting in translocation of G alpha 13 to detergent-resistant membranes (DRMs) and reduced p115Rho-GEF binding. RGS4 does not bind G alpha 13 or attenuate G alpha 13-dependent responses, and neither RGS16 nor RGS4 affects G alpha 12-mediated signalling. These results elucidate a new mechanism whereby a classical RGS protein regulates G alpha 13-mediated signal transduction independently of the RGS box.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Proteínas/metabolismo , Proteínas RGS/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/metabolismo , Línea Celular Tumoral , Retroalimentación Fisiológica/genética , Regulación de la Expresión Génica/genética , Genes Reguladores/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Factores de Intercambio de Guanina Nucleótido Rho
9.
J Cell Biol ; 169(6): 885-96, 2005 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-15955845

RESUMEN

Extracellular stimuli that activate cell surface receptors modulate glucocorticoid actions via as yet unclear mechanisms. Here, we report that the guanine nucleotide-binding protein (G protein)-coupled receptor-activated WD-repeat Gbeta interacts with the glucocorticoid receptor (GR), comigrates with it into the nucleus and suppresses GR-induced transactivation of the glucocorticoid-responsive genes. Association of Ggamma with Gbeta is necessary for this action of Gbeta. Both endogenous and enhanced green fluorescent protein (EGFP)-fused Gbeta2 and Ggamma2 proteins were detected in the nucleus at baseline, whereas a fraction of EGFP-Gbeta2 and DsRed2-GR comigrated to the nucleus or the plasma membrane, depending on the exposure of cells to dexamethasone or somatostatin, respectively. Gbeta2 was associated with GR/glucocorticoid response elements (GREs) in vivo and suppressed activation function-2-directed transcriptional activity of the GR. We conclude that the Gbetagamma complex interacts with the GR and suppresses its transcriptional activity by associating with the transcriptional complex formed on GR-responsive promoters.


Asunto(s)
Núcleo Celular/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas Represoras/metabolismo , Activación Transcripcional/genética , Transporte Activo de Núcleo Celular/genética , Animales , Células COS , Membrana Celular/genética , Membrana Celular/metabolismo , Núcleo Celular/genética , Chlorocebus aethiops , Dexametasona/farmacología , Subunidades gamma de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Proteínas de Unión al GTP , Glucocorticoides/metabolismo , Células HCT116 , Células HeLa , Humanos , Sustancias Macromoleculares , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Regiones Promotoras Genéticas/genética , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Receptores de Cinasa C Activada , Elementos de Respuesta/efectos de los fármacos , Elementos de Respuesta/genética , Somatostatina/farmacología
10.
J Neurosci ; 28(52): 14176-88, 2008 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-19109500

RESUMEN

G-protein (Gbetagamma)-mediated voltage-dependent inhibition of N- and P/Q-type Ca(2+) channels contributes to presynaptic inhibition and short-term synaptic plasticity. The voltage dependence derives from the dissociation of Gbetagamma from the inhibited channels, but the underlying molecular and biophysical mechanisms remain largely unclear. In this study we investigated the role in this process of Ca(2+) channel beta subunit (Ca(v)beta) and a rigid alpha-helical structure between the alpha-interacting domain (AID), the primary Ca(v)beta docking site on the channel alpha(1) subunit, and the pore-lining IS6 segment. Gbetagamma inhibition of P/Q-type channels was reconstituted in giant inside-out membrane patches from Xenopus oocytes. Large populations of channels devoid of Ca(v)beta were produced by washing out a mutant Ca(v)beta with a reduced affinity for the AID. These beta-less channels were still inhibited by Gbetagamma, but without any voltage dependence, indicating that Ca(v)beta is indispensable for voltage-dependent Gbetagamma inhibition. A truncated Ca(v)beta containing only the AID-binding guanylate kinase (GK) domain could fully confer voltage dependence to Gbetagamma inhibition. Gbetagamma did not alter inactivation properties, and channels recovered from Gbetagamma inhibition exhibited the same activation property as un-inhibited channels, indicating that Gbetagamma does not dislodge Ca(v)beta from the inhibited channel. Furthermore, voltage-dependent Gbetagamma inhibition was abolished when the rigid alpha-helix between the AID and IS6 was disrupted by insertion of multiple glycines, which also eliminated Ca(v)beta regulation of channel gating, revealing a pivotal role of this rigid alpha-helix in both processes. These results suggest that depolarization-triggered movement of IS6, coupled to the subsequent conformational change of the Gbetagamma-binding pocket through a rigid alpha-helix induced partly by the Ca(v)beta GK domain, causes the dissociation of Gbetagamma and is fundamental to voltage-dependent Gbetagamma inhibition.


Asunto(s)
Canales de Calcio Tipo N/fisiología , Proteínas de Unión al GTP/fisiología , Activación del Canal Iónico/fisiología , Secuencia de Aminoácidos , Animales , Biofisica , Células Cultivadas , Estimulación Eléctrica/métodos , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/farmacología , Guanilato-Quinasas/metabolismo , Insectos , Activación del Canal Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Modelos Moleculares , Mutación/genética , Oocitos , Técnicas de Placa-Clamp/métodos , Unión Proteica , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Subunidades de Proteína/farmacología , Factores de Tiempo , Xenopus laevis
11.
Neurosignals ; 17(1): 55-70, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19212140

RESUMEN

Accumulating data indicate that G12 subfamily (Galpha12/13)-mediated signaling pathways play pivotal roles in a variety of physiological processes, while aberrant regulation of this pathway has been identified in various human diseases. It has been demonstrated that Galpha12/13-mediated signals form networks with other signaling proteins at various levels, from cell surface receptors to transcription factors, to regulate cellular responses. Galpha12/13 have slow rates of nucleotide exchange and GTP hydrolysis, and specifically target RhoGEFs containing an amino-terminal RGS homology domain (RH-RhoGEFs), which uniquely function both as a GAP and an effector for Galpha12/13. In this review, we will focus on the mechanisms regulating the Galpha12/13 signaling system, particularly the Galpha12/13-RH-RhoGEF-Rho pathway, which can regulate a wide variety of cellular functions from migration to transformation.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Transducción de Señal , Animales , Enfermedades Cardiovasculares/metabolismo , Reguladores de Proteínas de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Sistema Inmunológico/fisiología , Neoplasias/metabolismo , Spodoptera , Proteínas de Unión al GTP rho/metabolismo
12.
Mol Cell Biol ; 26(13): 5190-200, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16782902

RESUMEN

The phagocyte NADPH oxidase consists of multiple protein subunits that interact with each other to form a functional superoxide-generating complex. Although the essential components for superoxide production have been well characterized, other proteins potentially involved in the regulation of NADPH oxidase activation remain to be identified. We report here that the Galphai subunit of heterotrimeric G proteins is a novel binding partner for p67phox in transfected HEK293T cells and peripheral blood polymorphonuclear leukocytes. p67phox preferably interacted with inactive Galphai. Expression of p67phox caused a dose-dependent decrease in intracellular cyclic AMP concentration, suggesting altered function of Galphai. We identified a fragment of p67phox, consisting of the PB1 domain and the C-terminal SH3 domain, to be critical for the interaction with Galphai. Because these domains are involved in the interaction with p47phox and p40phox, the relationship between the respective binding events was investigated. Wild-type Galphai, but not its QL mutant, could promote the interaction between p67phox and p47phox. However, the interaction between p67phox and p40phox was not affected by either Galphai form. These results provide the first evidence for an interaction between p67phox and an alpha subunit of heterotrimeric G proteins, suggesting a potential role for Galphai in the regulation or activation of NADPH oxidase.


Asunto(s)
Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , NADH Deshidrogenasa/metabolismo , Fosfoproteínas/metabolismo , Animales , Sitios de Unión , Células Cultivadas , AMP Cíclico/antagonistas & inhibidores , AMP Cíclico/metabolismo , Activación Enzimática , Humanos , NADPH Oxidasas/metabolismo , Fagocitos/enzimología , Fosfoproteínas/genética , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína/genética , Ratas , Eliminación de Secuencia , Dominios Homologos src/genética
13.
Cell Signal ; 19(8): 1681-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17449226

RESUMEN

Galpha12 and Galpha13 transduce signals from G protein-coupled receptors to RhoA through RhoGEFs containing an RGS homology (RH) domain, such as p115 RhoGEF or leukemia-associated RhoGEF (LARG). The RH domain of p115 RhoGEF or LARG binds with high affinity to active forms of Galpha12 and Galpha13 and confers specific GTPase-activating protein (GAP) activity, with faster GAP responses detected in Galpha13 than in Galpha12. At the same time, Galpha13, but not Galpha12, directly stimulates the RhoGEF activity of p115 RhoGEF or nonphosphorylated LARG in reconstitution assays. In order to better understand the molecular mechanism by which Galpha13 regulates RhoGEF activity through interaction with RH-RhoGEFs, we sought to identify the region(s) of Galpha13 involved in either the GAP response or RhoGEF activation. For this purpose, we generated chimeras between Galpha12 and Galpha13 subunits and characterized their biochemical activities. In both cell-based and reconstitution assays of RhoA activation, we found that replacing the carboxyl-terminal region of Galpha12 (residues 267-379) with that of Galpha13 (residues 264-377) conferred gain-of-function to the resulting chimeric subunit, Galpha12C13. The inverse chimera, Galpha13C12, exhibited basal RhoA activation which was similar to Galpha12. In contrast to GEF assays, GAP assays showed that Galpha12C13 or Galpha13C12 chimeras responded to the GAP activity of p115 RhoGEF or LARG in a manner similar to Galpha12 or Galpha13, respectively. We conclude from these results that the carboxyl-terminal region of Galpha13 (residues 264-377) is essential for its RhoGEF stimulating activity, whereas the amino-terminal alpha helical and switch regions of Galpha12 and Galpha13 are responsible for their differential GAP responses to the RH domain.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/química , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Proteínas RGS/química , Proteínas de Unión al GTP rho/química , Proteínas de Unión al GTP rho/metabolismo , Animales , Baculoviridae/genética , Células HeLa , Humanos , Modelos Biológicos , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Spodoptera/citología , Spodoptera/metabolismo , Spodoptera/virología , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA/metabolismo
14.
FEBS Lett ; 580(16): 3879-82, 2006 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-16797547

RESUMEN

HEK293 cells were transfected with cDNAs for Gbeta1(W332A) [a mutant Gbeta1], Ggamma2, and inward rectifier K+ channels (Kir3.1/Kir3.2). Application of Gbeta1gamma2 protein to these cells activated the K+ channels only slightly. When mu-opioid receptors and Kir3.1/Kir3.2 were transfected, application of a mu-opioid agonist induced a Kir3 current. However, co-expression of Gbeta1(W332A) suppressed this current. Most likely, Gbeta1(W332A) inhibited the action of the endogenous Gbeta. Such a dominant negative effect of Gbeta1(W332A) was also observed in neuronal Kir3 channels in locus coeruleus. The mutant, Gbeta1(W332A) protein, although inactive, retains its ability to bind Kir3 and prevents the wild type Gbeta from activating the channel.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Genes Dominantes/genética , Mutación/genética , Animales , Bovinos , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Humanos , Locus Coeruleus/fisiología , Ratones , Ratas , Ratas Long-Evans , Receptores Opioides mu/metabolismo , Factores de Tiempo
15.
Circ Res ; 91(10): 961-9, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12433842

RESUMEN

In neonatal cardiomyocytes, activation of the G(q)-coupled alpha(1)-adrenergic receptor (alpha(1)AR) induces hypertrophy by activating mitogen-activated protein kinases, including c-Jun NH(2)-terminal kinase (JNK). Here, we show that JNK activation is essential for alpha(1)AR-induced hypertrophy, in that alpha(1)AR-induced hypertrophic responses, such as reorganization of the actin cytoskeleton and increased protein synthesis, could be blocked by expressing the JNK-binding domain of JNK-interacting protein-1, a specific inhibitor of JNK. We also identified the classes and subunits of G proteins that mediate alpha(1)AR-induced JNK activation and hypertrophic responses by generating several recombinant adenoviruses that express polypeptides capable of inhibiting the function of specific G-protein subunits. alpha(1)AR-induced JNK activation was inhibited by the expression of carboxyl terminal regions of Galpha(q), Galpha(12), and Galpha(13). JNK activation was also inhibited by the Galpha(q/11)- or Galpha(12/13)-specific regulator of G-protein signaling (RGS) domains and by C3 toxin but was not affected by treatment with pertussis toxin or by expression of the carboxyl terminal region of G protein-coupled receptor kinase 2, a polypeptide that sequesters Gbetagamma. alpha(1)AR-induced hypertrophic responses were inhibited by Galpha(q/11)- and Galpha(12/13)-specific RGS domains, C3 toxin, and the carboxyl terminal region of G protein-coupled receptor kinase 2 but not by pertussis toxin. Activation of Rho was inhibited by carboxyl terminal regions of Galpha(12) and Galpha(13) but not by Galpha(q). Our findings suggest that alpha(1)AR-induced hypertrophic responses are mediated in part by a Galpha(12/13)-Rho-JNK pathway, in part by a G(q/11)-JNK pathway that is Rho independent, and in part by a Gbetagamma pathway that is JNK independent.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Cardiomegalia/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Miocardio/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , ADP Ribosa Transferasas/farmacología , Adenoviridae/genética , Animales , Animales Recién Nacidos , Toxinas Botulínicas/farmacología , Cardiomegalia/etiología , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Proteínas Portadoras/farmacología , Células Cultivadas , Proteínas de Unión al ADN/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocardio/citología , Oxidantes/farmacología , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Toxina del Pertussis/farmacología , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transfección , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
16.
Trends Pharmacol Sci ; 25(2): 61-3, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15106629

RESUMEN

G-protein-mediated signaling is the most widely used signaling mechanism in cells and its regulation is crucial for various physiological functions. G-protein-coupled receptor (GPCR) kinases (GRKs) are involved in the desensitization of GPCR signals. Recently, the X-ray crystal structure of GRK2 complexed with G beta gamma was demonstrated and revealed the intimate association of three important signaling modules with G beta gamma to regulate GRK2 activity.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Transducción de Señal/fisiología , Reguladores de Proteínas de Unión al GTP/fisiología , Relación Estructura-Actividad , Quinasas de Receptores Adrenérgicos beta
17.
FASEB J ; 17(3): 440-2, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12514120

RESUMEN

RGS proteins finely tune heterotrimeric G-protein signaling. Implying the need for such fine-tuning in the developing vascular system, in situ hybridization revealed a striking and extensive expression pattern of Rgs5 in the arterial walls of E12.5-E17.5 mouse embryos. The distribution and location of the Rgs5-positive cells typified that of pericytes and strikingly overlapped the known expression pattern of platelet-derived growth factor receptor (PDGFR)-beta. Both E14.5 PDGFR-beta- and platelet-derived growth factor (PDGF)-B-deficient mice exhibited markedly reduced levels of Rgs5 in their vascular plexa and small arteries. This likely reflects the loss of pericytes in the mutant mice. RGS5 acts as a potent GTPase activating protein for Gi(alpha) and Gq(alpha) and it attenuated angiotensin II-, endothelin-1-, sphingosine-1-phosphate-, and PDGF-induced ERK-2 phosphorylation. Together these results indicate that RGS5 exerts control over PDGFR-beta and GPCR-mediated signaling pathways active during fetal vascular maturation.


Asunto(s)
Arterias/citología , Proteínas Inmediatas-Precoces/metabolismo , Lisofosfolípidos , Pericitos/metabolismo , Proteínas RGS/metabolismo , Receptores de Superficie Celular , Receptores Acoplados a Proteínas G , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Esfingosina/análogos & derivados , Células 3T3 , Angiotensina II/antagonistas & inhibidores , Animales , Arterias/embriología , Arterias/metabolismo , Endotelina-1/antagonistas & inhibidores , Hibridación in Situ , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Modelos Biológicos , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Proteínas RGS/genética , Proteínas RGS/fisiología , ARN Mensajero/biosíntesis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores Lisofosfolípidos , Transducción de Señal , Esfingosina/antagonistas & inhibidores , Transcripción Genética
18.
Methods Enzymol ; 390: 475-83, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15488195

RESUMEN

We have identified the novel Galphaz-binding protein, which is referred to as the G-protein-regulated inducer of neurite outgrowth (GRIN1) using the far-western method. GRIN1 is expressed specifically in brain and binds preferentially to the activated form of alpha subunits of Gz, Gi, and Go. Coexpression of GRIN1 and the activated form of Galphao induce neurite outgrowth in Neuro2a cells. We have further identified two human GRIN1 homologs, GRIN2 and GRIN3, in the database. This article shows that GRIN2 can also bind to the GTP-bound form of Galphao. These findings suggest that the GRIN1 family may function as a downstream effector for Galphao to regulate neurite growth.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Humanos , Proteínas de la Membrana , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Unión Proteica , Proteína Quinasa C/aislamiento & purificación , Proteína Quinasa C/metabolismo , Proteína Quinasa C-alfa , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de N-Metil-D-Aspartato , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia
19.
Methods Enzymol ; 390: 285-94, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15488184

RESUMEN

Three mammalian Rho guanine nucleotide exchange factors (RhoGEFs), leukemia-associated RhoGEF (LARG), p115RhoGEF, and PDZ-RhoGEF, contain regulator of G-protein signaling (RGS) domains within their amino-terminal regions. These RhoGEFs link signals from heterotrimeric G12/13 protein-coupled receptors to Rho GTPase activation, leading to various cellular responses, such as actin reorganization and gene expression. The activity of these RhoGEFs is regulated by Galpha12/13 through their RGS domains. Because RhoGEFs stimulate guanine nucleotide exchange by Rho GTPases, RhoGEF activation can be measured by monitoring GTP binding to or GDP dissociation from Rho GTPases. This article describes methods used to perform reconstitution assays to measure the activity of RhoGEFs regulated by Galpha12/13.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas RGS/metabolismo , Animales , Línea Celular , Subunidades alfa de la Proteína de Unión al GTP G12-G13/aislamiento & purificación , Factores de Intercambio de Guanina Nucleótido/aislamiento & purificación , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Difosfato/metabolismo , Fosforilación , Factores de Intercambio de Guanina Nucleótido Rho , Proteína de Unión al GTP rhoA/aislamiento & purificación , Proteína de Unión al GTP rhoA/metabolismo
20.
Methods Enzymol ; 389: 244-65, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15313570

RESUMEN

Given recent efforts to determine the sequence information on thousands of genes in the human genome, the current challenge is to identify the functions of these genes, including those encoding the regulator of G-protein signaling protein gene superfamily, and to establish their roles in particular signaling pathways in a native system. Increasingly, reverse genetic approaches are being used to address these questions. This article compares two powerful approaches [ribozyme and "short interfering" RNA (siRNA) techniques] under identical conditions for the first report on the suppression of endogenous RGS domain-containing RhoGEFs. The siRNA technique was found to be much more potent than ribozyme targeting at the same mRNA site of RGS-RhoGEFs. Also, the three siRNAs targeting LARG, PDZ-RhoGEF, and p115-RhoGEF are able to discriminate the closely related sequences within this RGS-RhoGEF gene family.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas RGS/antagonistas & inhibidores , Proteínas RGS/química , ARN Catalítico/metabolismo , ARN Interferente Pequeño/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , Factores de Intercambio de Guanina Nucleótido/química , Humanos , Luciferasas/genética , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Catalítico/biosíntesis , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA