Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 298(2): 101551, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34973340

RESUMEN

WHIM syndrome is a rare immunodeficiency disorder that is characterized by warts, hypogammaglobulinemia, infections, and myelokathexis. While several gain-of-function mutations that lead to C-terminal truncations, frame shifts and point mutations in the chemokine receptor CXCR4 have been identified in WHIM syndrome patients, the functional effect of these mutations are not fully understood. Here, we report on a new WHIM syndrome mutation that results in a frame shift within the codon for Ser339 (S339fs5) and compare the properties of S339fs5 with wild-type CXCR4 and a previously identified WHIM syndrome mutant, R334X. The S339fs5 and R334X mutants exhibited significantly increased signaling compared to wild-type CXCR4 including agonist-promoted calcium flux and extracellular-signal-regulated kinase activation. This increase is at least partially due to a significant decrease in agonist-promoted phosphorylation, ß-arrestin binding, and endocytosis of S339fs5 and R334X compared with wild-type CXCR4. Interestingly, there were also significant differences in receptor degradation, with S339fs5 having a very high basal level of degradation compared with that of R334X and wild-type CXCR4. In contrast to wild-type CXCR4, both R334X and S339fs5 were largely insensitive to CXCL12-promoted degradation. Moreover, while basal and agonist-promoted degradation of wild-type CXCR4 was effectively inhibited by the CXCR4 antagonist TE-14016, this had no effect on the degradation of the WHIM mutants. Taken together, these studies identify a new WHIM syndrome mutant, CXCR4-S339fs5, which promotes enhanced signaling, reduced phosphorylation, ß-arrestin binding and endocytosis, and a very high basal rate of degradation that is not protected by antagonist treatment.


Asunto(s)
Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4 , Verrugas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Humanos , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Verrugas/genética , Verrugas/metabolismo , beta-Arrestina 1/metabolismo , beta-Arrestinas/metabolismo
2.
Mol Pharmacol ; 91(6): 554-566, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28331048

RESUMEN

Phosphorylation of G protein-coupled receptors (GPCRs) is a key event for cell signaling and regulation of receptor function. Previously, using tandem mass spectrometry, we identified two phosphorylation sites at the distal C-terminal tail of the chemokine receptor CXCR4, but were unable to determine which specific residues were phosphorylated. Here, we demonstrate that serines (Ser) 346 and/or 347 (Ser-346/7) of CXCR4 are phosphorylated upon stimulation with the agonist CXCL12 as well as a CXCR4 pepducin, ATI-2341. ATI-2341, a Gαißγ heterotrimer-biased CXCR4 agonist, induced more robust phosphorylation of Ser-346/7 compared with CXCL12. Knockdown of G protein-coupled receptor kinase (GRK) 2, GRK3, or GRK6 reduced CXCL12-induced phosphorylation of Ser-346/7 with GRK3 knockdown having the strongest effect, while inhibition of the conventional protein kinase C (PKC) isoforms, particularly PKCα, reduced phosphorylation of Ser-346/7 induced by either CXCL12 or ATI-2341. The loss of GRK3- or PKC-mediated phosphorylation of Ser-346/7 impaired the recruitment of ß-arrestin to CXCR4. We also found that a pseudo-substrate peptide inhibitor for PKCζ effectively inhibited CXCR4 phosphorylation and signaling, most likely by functioning as a nonspecific CXCR4 antagonist. Together, these studies demonstrate the role Ser-346/7 plays in arrestin recruitment and initiation of receptor desensitization and provide insight into the dysregulation of CXCR4 observed in patients with various forms of WHIM syndrome.


Asunto(s)
Quinasa 3 del Receptor Acoplado a Proteína-G/metabolismo , Proteína Quinasa C/metabolismo , Receptores CXCR4/metabolismo , beta-Arrestinas/metabolismo , Células HEK293 , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Serina/metabolismo
3.
J Biol Chem ; 292(14): 5943-5956, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28213524

RESUMEN

G protein-coupled receptors (GPCRs) regulate many animal behaviors. GPCR signaling is mediated by agonist-promoted interactions of GPCRs with heterotrimeric G proteins, GPCR kinases (GRKs), and arrestins. To further elucidate the role of GRKs in regulating GPCR-mediated behaviors, we utilized the genetic model system Caenorhabditis elegans Our studies demonstrate that grk-2 loss-of-function strains are egg laying-defective and contain low levels of serotonin (5-HT) and high levels of the 5-HT metabolite 5-hydroxyindole acetic acid (5-HIAA). The egg laying defect could be rescued by the expression of wild type but not by catalytically inactive grk-2 or by the selective expression of grk-2 in hermaphrodite-specific neurons. The addition of 5-HT or inhibition of 5-HT metabolism also rescued the egg laying defect. Furthermore, we demonstrate that AMX-2 is the primary monoamine oxidase that metabolizes 5-HT in C. elegans, and we also found that grk-2 loss-of-function strains have abnormally high levels of AMX-2 compared with wild-type nematodes. Interestingly, GRK-2 was also found to interact with and promote the phosphorylation of AMX-2. Additional studies reveal that 5-HIAA functions to inhibit egg laying in a manner dependent on the 5-HT receptor SER-1 and the G protein GOA-1. These results demonstrate that GRK-2 modulates 5-HT metabolism by regulating AMX-2 function and that 5-HIAA may function in the SER-1 signaling pathway.


Asunto(s)
Proteínas de Caenorhabditis elegans/biosíntesis , Caenorhabditis elegans/metabolismo , Quinasas de Receptores Acoplados a Proteína-G/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Monoaminooxidasa/metabolismo , Serotonina/metabolismo , Transducción de Señal/fisiología , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Quinasas de Receptores Acoplados a Proteína-G/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Ácido Hidroxiindolacético/metabolismo , Monoaminooxidasa/genética , Receptores de Serotonina 5-HT2/genética , Receptores de Serotonina 5-HT2/metabolismo , Serotonina/genética
4.
Proc Natl Acad Sci U S A ; 110(52): E5088-97, 2013 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-24309376

RESUMEN

Short lipidated peptide sequences derived from various intracellular loop regions of G protein-coupled receptors (GPCRs) are named pepducins and act as allosteric modulators of a number of GPCRs. Recently, a pepducin selectively targeting the C-X-C chemokine receptor type 4 (CXCR4) was found to be an allosteric agonist, active in both cell-based assays and in vivo. However, the precise mechanism of action of this class of ligands remains poorly understood. In particular, given the diversity of signaling effectors that can be engaged by a given receptor, it is not clear whether pepducins can show biased signaling leading to functional selectivity. To explore the ligand-biased potential of pepducins, we assessed the effect of the CXCR4 selective pepducin, ATI-2341, on the ability of the receptor to engage the inhibitory G proteins (Gi1, Gi2 and Gi3), G13, and ß-arrestins. Using bioluminescence resonance energy transfer-based biosensors, we found that, in contrast to the natural CXCR4 ligand, stromal cell-derived factor-1α, which promotes the engagement of the three Gi subtypes, G13 and the two ß-arrestins, ATI-2341 leads to the engagement of the Gi subtypes but not G13 or the ß-arrestins. Calculation of the transduction ratio for each pathway revealed a strong negative bias of ATI-2341 toward G13 and ß-arrestins, revealing functional selectivity for the Gi pathways. The negative bias toward ß-arrestins results from the reduced ability of the pepducin to promote GPCR kinase-mediated phosphorylation of the receptor. In addition to revealing ligand-biased signaling of pepducins, these findings shed some light on the mechanism of action of a unique class of allosteric regulators.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/agonistas , Células Madre Hematopoyéticas/metabolismo , Lipopéptidos/metabolismo , Receptores CXCR4/metabolismo , Transferencia de Energía por Resonancia de Bioluminiscencia , Western Blotting , Citometría de Flujo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Células HEK293 , Humanos
5.
Mol Biol Cell ; 24(18): 2795-806, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23904266

RESUMEN

G protein-coupled receptor kinases (GRKs) play a central role in regulating receptor signaling, but recent studies suggest a broader role in modulating normal cellular functions. For example, GRK5 has been shown to localize to centrosomes and regulate microtubule nucleation and cell cycle progression. Here we demonstrate that GRK2 is also localized to centrosomes, although it has no role in centrosome duplication or microtubule nucleation. Of interest, knockdown of GRK2 inhibits epidermal growth factor receptor (EGFR)-mediated separation of duplicated centrosomes. This EGFR/GRK2-mediated process depends on the protein kinases mammalian STE20-like kinase 2 (Mst2) and Nek2A but does not involve polo-like kinase 1. In vitro analysis and dominant-negative approaches reveal that GRK2 directly phosphorylates and activates Mst2. Collectively these findings demonstrate that GRK2 is localized to centrosomes and plays a central role in mitogen-promoted centrosome separation most likely via its ability to phosphorylate Mst2.


Asunto(s)
Centrosoma/efectos de los fármacos , Centrosoma/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Secuencia de Aminoácidos , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Datos de Secuencia Molecular , Quinasas Relacionadas con NIMA , Fosfopéptidos/química , Fosfopéptidos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Serina-Treonina Quinasa 3
6.
Mol Pharmacol ; 82(6): 1150-61, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22962331

RESUMEN

The human hydroxycarboxylic acid receptor 2 (HCA2), also known as GPR109A and HM74a, was first identified as a niacin receptor and has recently received significant attention because of its potential to clinically modify plasma lipids in a favorable manner. Our recent studies have demonstrated that the niacin-induced internalization of HCA2 receptors is regulated by G protein-coupled receptor kinase (GRK) 2 and arrestin3 and that internalized receptors rapidly recycle back to the cell surface. The investigation presented here used a combination of amino acid deletion and site-directed mutagenesis to identify structural and functional domains within the HCA2 C terminus and explore their potential roles in receptor phosphorylation, desensitization, and internalization. We first constructed four mutants with deletions of 10 to 15 amino acids each that were distinct from truncated mutants. We successfully identified different domains responsible for receptor export, constitutive activity, desensitization, phosphorylation, and internalization. We also generated a comprehensive series of alanine substitution mutants, replacing conserved serine and threonine residues in the C terminus with alanine residues to pinpoint the key residues that are essential for GRK2-mediated phosphorylation and arrestin3 association. Moreover, we found that a sequence from residues 329 to 343 in the C-terminal tail of HCA2 plays a crucial role in keeping HCA2 in an inactive conformation. These data demonstrate the importance of distinct domains within the C terminus of HCA2 for receptor cell surface expression, desensitization, and internalization and phosphorylation and stabilization of an inactive receptor conformation.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/metabolismo , Secuencia de Aminoácidos , Aminoácidos/genética , Aminoácidos/metabolismo , Arrestinas/genética , Arrestinas/metabolismo , Línea Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Células HEK293 , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida/métodos , Mutación , Fosforilación , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptores Acoplados a Proteínas G/genética , Receptores Nicotínicos/genética
7.
Mol Cell ; 40(6): 877-92, 2010 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-21172654

RESUMEN

While the small GTPase Rac1 and its effectors are well-established mediators of mitogenic and motile signaling by tyrosine kinase receptors and have been implicated in breast tumorigenesis, little is known regarding the exchange factors (Rac-GEFs) that mediate ErbB receptor responses. Here, we identify the PIP(3)-Gßγ-dependent Rac-GEF P-Rex1 as an essential mediator of Rac1 activation, motility, cell growth, and tumorigenesis driven by ErbB receptors in breast cancer cells. Notably, activation of P-Rex1 in breast cancer cells requires the convergence of inputs from ErbB receptors and a Gßγ- and PI3Kγ-dependent pathway. Moreover, we identified the GPCR CXCR4 as a crucial mediator of P-Rex1/Rac1 activation in response to ErbB ligands. P-Rex1 is highly overexpressed in human breast cancers and their derived cell lines, particularly those with high ErbB2 and ER expression. In addition to the prognostic and therapeutic implications, our findings reveal an ErbB effector pathway that is crucial for breast cancer progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Oncogénicas v-erbB/metabolismo , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Células Tumorales Cultivadas
8.
J Biol Chem ; 285(29): 22605-18, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20460384

RESUMEN

Nicotinic acid (niacin) has been widely used as a favorable lipid-lowering drug for several decades, and the orphan G protein-coupled receptor GPR109A has been identified to be a receptor for niacin. Mechanistic investigations have shown that as a G(i)-coupled receptor, GPR109A inhibits adenylate cyclase activity upon niacin activation, thereby inhibiting free fatty acid liberation. However, the underlying molecular mechanisms that regulate signaling and internalization of GPR109A remain largely unknown. To further characterize GPR109A internalization, we made a construct to express GPR109A fused with enhanced green fluorescent protein (EGFP) at its carboxyl-terminal end. In stable GPR109A-EGFP-expressing HEK-293 cells, GPR109A-EGFP was mainly localized at the plasma membrane and was rapidly internalized in a dose- and time-dependent manner upon agonist stimulation. GPR109A internalization was completely blocked by hypertonic sucrose, indicating that GPR109A internalizes via the clathrin-coated pit pathway. Further investigation demonstrated that internalized GPR109A was recycled to the cell surface after the removal of agonist, and recycling of the internalized receptors was not blocked by treatment with acidotropic agents, NH(4)Cl and monensin. Pertussis toxin pretreatment not only inhibited forskolin-induced cAMP accumulation and intracellular Ca(2+) mobilization; it also significantly attenuated agonist-promoted GPR109A internalization. Moreover, RNA interference experiments showed that knockdown of GRK2 (G protein-coupled receptor kinase 2) and arrestin3 expression significantly impaired receptor internalization. Taken together, these results indicate that the agonist-induced internalization of GPR109A receptors is regulated by GRK2 and arrestin3 in a pertussis toxin-sensitive manner and that internalized receptor recycling is independent of endosomal acidification.


Asunto(s)
Arrestinas/metabolismo , Endocitosis , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Línea Celular , Clatrina/metabolismo , Invaginaciones Cubiertas de la Membrana Celular/efectos de los fármacos , Invaginaciones Cubiertas de la Membrana Celular/metabolismo , Dinaminas/metabolismo , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Modelos Biológicos , Toxina del Pertussis/farmacología , Transporte de Proteínas/efectos de los fármacos , Receptores Acoplados a Proteínas G/agonistas , Proteínas Recombinantes de Fusión/metabolismo , Familia-src Quinasas/metabolismo
9.
Biochem J ; 425(1): 169-78, 2009 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-19796012

RESUMEN

Tumor necrosis factor-α (TNFα) is a multifunctional cytokine involved in the pathophysiology of many chronic inflammatory diseases. TNFα activation of the nuclear factor κB (NFκB) signaling pathway particularly in macrophages has been implicated in many diseases. We demonstrate here that G-protein coupled receptor kinase-2 and 5 (GRK2 and 5) regulate TNFα-induced NFκB signaling in Raw264.7 macrophages. RNAi knockdown of GRK2 or 5 in macrophages significantly inhibits TNFα-induced IκBα phosphorylation and degradation, NFκB activation, and expression of the NFκB-regulated gene, macrophage inflammatory protein-1ß. Consistent with these results, over-expression of GRK2 or 5 enhances TNFα-induced NFκB activity. In addition,we show that GRK2 and 5 interact with IκBα via the N-terminal domain of IκBα and that IκBα isa substrate for GRK2 and 5 in vitro. Furthermore, we also find that GRK5 but not GRK2 phosphorylates IκBα at the same amino acid residues (Ser32/36) as that of IKKß. Interestingly,associated with these results, knockdown of IKKß in Raw264.7 macrophages did not affect TNFα-induced IκBα phosphorylation. Taken together, these results demonstrate that both GRK2 and 5 are important and novel mediators of a non-traditional IκBα-NFκB signaling pathway.


Asunto(s)
Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Western Blotting , Línea Celular , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Quinasas de Receptores Acoplados a Proteína-G/genética , Humanos , Inmunoprecipitación , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Inhibidor NF-kappaB alfa , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Interferencia de ARN
10.
FEBS Lett ; 583(9): 1463-8, 2009 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-19345219

RESUMEN

Neuropeptides of the adipokinetic hormone (AKH) family are among the best studied hormone peptides, but its signaling pathways remain to be elucidated. In this study, we molecularly characterized the signaling of Bombyx AKH receptor (AKHR) and its peptide ligands in HEK293 cells. In HEK293 cells stably expressing AKHR, AKH1 stimulation not only led to a ligand concentration dependent mobilization of intracellular Ca(2+) and cAMP accumulation, but also elicited transient activation of extracellular signal-regulated kinase 1/2 (ERK1/2) pathway. We observed that AKH receptor was rapidly internalized after AKH1 stimulation. We further demonstrated that AKH2 exhibited high activities in cAMP accumulation and ERK1/2 activation on AKHR comparable to AKH1, whereas AKH3 was much less effective.


Asunto(s)
Bombyx/metabolismo , Hormonas de Insectos/metabolismo , Oligopéptidos/metabolismo , Fragmentos de Péptidos/metabolismo , Ácido Pirrolidona Carboxílico/análogos & derivados , Receptores de Glucagón/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Calcio/metabolismo , Línea Celular , AMP Cíclico/metabolismo , Cartilla de ADN , Citometría de Flujo , Humanos , Hormonas de Insectos/química , Ligandos , Sistema de Señalización de MAP Quinasas , Microscopía Fluorescente , Datos de Secuencia Molecular , Oligopéptidos/química , Ácido Pirrolidona Carboxílico/química , Ácido Pirrolidona Carboxílico/metabolismo , Receptores de Glucagón/química
11.
Mol Pharmacol ; 74(2): 338-47, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18388243

RESUMEN

We have used RNA interference previously to demonstrate that G protein-coupled receptor kinase 2 (GRK2) regulates endogenously expressed H1 histamine receptor in human embryonic kidney 293 cells. In this report, we investigate the regulation of endogenously expressed M(3) muscarinic acetylcholine receptor (M(3) mAChR). We show that knockdown of GRK2, GRK3, or GRK6, but not GRK5, significantly increased carbachol-mediated calcium mobilization. Stable expression of wild-type GRK2 or a kinase-dead mutant (GRK2-K220R) reduced calcium mobilization after receptor activation, whereas GRK2 mutants defective in Galpha(q) binding (GRK2-D110A, GRK2-R106A, and GRK2-R106A/K220R) had no effect on calcium signaling, suggesting that GRK2 primarily regulates G(q) after M(3) mAChR activation. The knockdown of arrestin-2 or arrestin-3 also significantly increased carbachol-mediated calcium mobilization. Knockdown of GRK2 and the arrestins also significantly enhanced carbachol-mediated activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), whereas prolonged ERK1/2 activation was only observed with GRK2 or arrestin-3 knockdown. We also investigated the role of casein kinase-1alpha (CK1alpha) and found that knockdown of CK1alpha increased calcium mobilization but not ERK activation. In summary, our data suggest that multiple proteins dynamically regulate M(3) mAChR-mediated calcium signaling, whereas GRK2 and arrestin-3 play the primary role in regulating ERK activation.


Asunto(s)
Receptor Muscarínico M3/fisiología , Transducción de Señal/fisiología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Línea Celular , Humanos , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Receptor Muscarínico M3/agonistas , Receptor Muscarínico M3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
12.
Traffic ; 7(10): 1420-31, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16899088

RESUMEN

Upon activation, many G protein-coupled receptors (GPCRs) internalize by clathrin-mediated endocytosis and are subsequently sorted to undergo recycling or lysosomal degradation. Here we observe that sorting can take place much earlier than previously thought, by entry of different GPCRs into distinct populations of clathrin-coated pit (CCP). These distinct populations were revealed by analysis of two purinergic GPCRs, P2Y(1) and P2Y(12), which enter two populations of CCPs in a mutually exclusive manner. The mechanisms underlying early GPCR sorting involve differential kinase-dependent processes because internalization of P2Y(12) is mediated by GPCR kinases (GRKs) and arrestin, whereas P2Y(1) internalization is GRK- and arrestin-independent but requires protein kinase C. Importantly, the beta(2) adrenoceptor which also internalizes in a GRK-dependent manner also traffics exclusively to P2Y(12)-containing CCPs. Our data therefore reveal distinct populations of CCPs that sort GPCR cargo at the plasma membrane using different kinase-dependent mechanisms.


Asunto(s)
Membrana Celular/metabolismo , Clatrina/metabolismo , Invaginaciones Cubiertas de la Membrana Celular/metabolismo , Endocitosis/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Anticuerpos Monoclonales/metabolismo , Arrestina/metabolismo , Línea Celular , Membrana Celular/ultraestructura , Humanos , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y12
13.
Blood ; 105(9): 3552-60, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15665114

RESUMEN

Adenosine 5'-diphosphate (ADP) plays a central role in regulating platelet function by the activation of the G protein-coupled receptors P2Y(1) and P2Y(12). Although it is well established that aggregation responses of platelets to ADP desensitize, the underlying mechanisms involved remain unclear. In this study we demonstrate that P2Y(1)- and P2Y(12)-mediated platelet responses desensitize rapidly. Furthermore, we have established that these receptors desensitize by different kinase-dependent mechanisms. G protein-coupled receptor kinase (GRK) 2 and GRK6 are both endogenously expressed in platelets. Transient overexpression of dominant-negative mutants of these kinases or reductions in endogenous GRK expression by the use of specific siRNAs in 1321N1 cells showed that P2Y(12), but not P2Y(1), desensitization is mediated by GRKs. In contrast, desensitization of P2Y(1), but not P2Y(12), is largely dependent on protein kinase C activity. This study is the first to show that both P2Y(1) and P2Y(12) desensitize in human platelets, and it reveals ways in which their sensitivity to ADP may be differentially and independently altered.


Asunto(s)
Adenosina Difosfato/farmacología , Proteínas de la Membrana/fisiología , Activación Plaquetaria/efectos de los fármacos , Receptores Purinérgicos P2/fisiología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Relación Dosis-Respuesta a Droga , Quinasas de Receptores Acoplados a Proteína-G , Humanos , Proteína Quinasa C/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , ARN Interferente Pequeño/farmacología , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y12 , Transfección , Quinasas de Receptores Adrenérgicos beta
14.
J Biol Chem ; 280(3): 2197-204, 2005 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-15542600

RESUMEN

The H1 histamine receptor (H1HR) is a member of the G protein-coupled receptor superfamily and regulates numerous cellular functions through its activation of the G(q/11) subfamily of heterotrimeric G proteins. Although the H1HR has been shown to undergo desensitization in multiple cell types, the mechanisms underlying the regulation of H1HR signaling are poorly defined. To address this issue, we examined the effects of wild type and mutant G protein-coupled receptor kinases (GRKs) on the phosphorylation and signaling of human H1HR in HEK293 cells. Overexpression of GRK2 promoted H1HR phosphorylation in intact HEK293 cells and completely inhibited inositol phosphate production stimulated by H1HR, whereas GRK5 and GRK6 had lesser effects on H1HR phosphorylation and signaling. Interestingly, catalytically inactive GRK2 (GRK2-K220R) also significantly attenuated H1HR-mediated inositol phosphate production, as did an N-terminal fragment of GRK2 previously characterized as a regulator of G protein signaling (RGS) protein for Galpha(q/11). Disruption of this RGS function in holo-GRK2 by mutation (GRK2-D110A) partially reversed the quenching effect of GRK2, whereas deletion of both the kinase activity and RGS function (GRK2-D110A/K220R) effectively relieved the inhibition of inositol phosphate generation. To evaluate the role of endogenous GRKs on H1HR regulation, we used small interfering RNAs to selectively target GRK2 and GRK5, two of the primary GRKs expressed in HEK293 cells. A GRK2-specific small interfering RNA effectively reduced GRK2 expression and resulted in a significant increase in histamine-promoted calcium flux. In contrast, knockdown of GRK5 expression was without effect on H1HR signaling. These findings demonstrate that GRK2 is the principal kinase mediating H1 histamine receptor desensitization in HEK293 cells and suggest that rapid termination of H1HR signaling is mediated by both the kinase activity and RGS function of GRK2.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptores Histamínicos H1/metabolismo , Secuencia de Bases , Calcio/metabolismo , Catálisis , Línea Celular , Cartilla de ADN , Humanos , Fosfatos de Inositol/biosíntesis , Transporte Iónico , Fosforilación , Ensayo de Unión Radioligante , Transducción de Señal , Quinasas de Receptores Adrenérgicos beta
15.
J Biol Chem ; 278(51): 50908-14, 2003 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-14557268

RESUMEN

G protein-coupled receptor kinase 2 (GRK2) is a serine/threonine-specific protein kinase that mediates agonist-dependent phosphorylation of numerous G protein-coupled receptors. In an effort to identify proteins that regulate GRK2 function, we searched for interacting proteins by immunoprecipitation of endogenous GRK2 from HL60 cells. Subsequent analysis by gel electrophoresis and mass spectrometry revealed that GRK2 associates with heat shock protein 90 (Hsp90). GRK2 interaction with Hsp90 was confirmed by co-immunoprecipitation and was effectively disrupted by geldanamycin, an Hsp90-specific inhibitor. Interestingly, geldanamycin treatment of HL60 cells decreased the expression of endogenous GRK2 in a dose- and time-dependent manner, and metabolic labeling demonstrated that geldanamycin rapidly accelerated the degradation of newly synthesized GRK2. The use of various protease inhibitors suggested that GRK2 degradation induced by geldanamycin was predominantly through the proteasome pathway. To test whether Hsp90 plays a general role in regulating GRK maturation, additional GRKs were studied by transient expression in COS-1 cells and subsequent treatment with geldanamycin. These studies demonstrate that GRK3, GRK5, and GRK6 are also stabilized by interaction with Hsp90. Taken together, our work revealed that GRK interaction with heat shock proteins plays an important role in regulating GRK maturation.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Benzoquinonas , Cisteína Endopeptidasas/metabolismo , Estabilidad de Enzimas , Quinasa 3 del Receptor Acoplado a Proteína-G , Quinasa 5 del Receptor Acoplado a Proteína-G , Quinasas de Receptores Acoplados a Proteína-G , Células HL-60 , Humanos , Lactamas Macrocíclicas , Complejos Multienzimáticos/metabolismo , Pruebas de Precipitina , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Quinonas/farmacología , Quinasas de Receptores Adrenérgicos beta
16.
J Biol Chem ; 277(20): 17476-85, 2002 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-11880384

RESUMEN

Chemokine receptor CXCR4 plays an important role in the immune system and the cellular entry of human immunodeficiency virus type 1 (HIV-1). To probe the stereospecificity of the CXCR4-ligand interface, d-amino acid peptides derived from natural chemokines, viral macrophage inflammatory protein II (vMIP-II) and stromal cell-derived factor-1alpha (SDF-1alpha), were synthesized and found to compete with (125)I-SDF-1alpha and monoclonal antibody 12G5 binding to CXCR4 with potency and selectivity comparable with or higher than their l-peptide counterparts. This was surprising because of the profoundly different side chain topologies between d- and l-enantiomers, which circular dichroism spectroscopy showed adopt mirror image conformations. Further direct binding experiments using d-peptide labeled with fluorescein (designated as FAM-DV1) demonstrated that d- and l-peptides shared similar or at least overlapping binding site(s) on the CXCR4 receptor. Structure-activity analyses of related peptide analogs of mixed chiralities or containing alanine replacements revealed specific residues at the N-terminal half of the peptides as key binding determinants. Acting as CXCR4 antagonists and with much higher biological stability than l-counterparts, the d-peptides showed significant activity in inhibiting the replication of CXCR4-dependent HIV-1 strains. These results show the remarkable stereochemical flexibility of the CXCR4-peptide interface. Further studies to understand the mechanism of this unusual feature of the CXCR4 binding surface might aid the development of novel CXCR4-binding molecules like the d-peptides that have high affinity and stability.


Asunto(s)
Quimiocinas/metabolismo , VIH-1 , Receptores CXCR4/metabolismo , Alanina/metabolismo , Secuencia de Aminoácidos , Unión Competitiva , Línea Celular , Quimiocina CXCL12 , Quimiocina CXCL2 , Quimiocinas/química , Quimiocinas CXC/metabolismo , Dicroismo Circular , Humanos , Conformación Molecular , Datos de Secuencia Molecular , Monocinas/metabolismo , Receptores CXCR4/química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA