Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Toxicol Appl Pharmacol ; 382: 114750, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31518596

RESUMEN

Hepatic ischemia/reperfusion injury (H-IRI) is associated with irreversible liver damage. The current study aimed to investigate the protective effect of carvedilol against H-IRI in high-fructose high-fat diet (HFrHFD)-fed mice and the role of G protein-coupled receptor kinase 2 and 5 (GRK2 and GRK5). Mice were fed HFrHFD for 16 weeks; then mice were subjected to 30 min of ischemia followed by 1 h of reperfusion at the end of feeding period. Carvedilol (20 mg/kg, i.p.) was administered 30 min before ischemia. To explore the role of GRK2 and GRK5 in mediating carvedilol effects, paroxetine (GRK2 inhibitor, 10 mg/kg, i.p.) and amlexanox (GRK5 inhibitor, 25 mg/kg, i.p.) were administered 30 min before carvedilol administration. Liver function, histopathology and hepatic oxidative stress, as well as inflammatory and apoptotic markers were measured at the end of the experiment. In addition, adrenergic receptor downstream signals were measured in the liver. Results showed increased markers of liver injury (ALT and AST) in mice subjected to H-IRI. Moreover, liver injury was associated with slight collagen deposits as revealed by histopathology and elevated hepatic levels of oxidative stress, inflammatory and apoptotic markers. On the other hand, carvedilol protected mice against H-IRI and improved all associated pathological changes. Furthermore, pre-injection of either GRK2 or GRK5 inhibitor did not change carvedilol effects on serum ALT level and liver collagen deposits, while increased its antioxidant, anti-inflammatory and anti-apoptotic effects. In conclusion, carvedilol protects against H-IRI in HFrHFD-fed mice. GRK2 and GRK5 may not play a potential role in mediating this effect.


Asunto(s)
Carvedilol/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Fructosa/toxicidad , Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Daño por Reperfusión/prevención & control , Animales , Carvedilol/farmacología , Fructosa/administración & dosificación , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Quinasa 5 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Hepatopatías/metabolismo , Hepatopatías/prevención & control , Masculino , Ratones , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico , Daño por Reperfusión/inducido químicamente , Daño por Reperfusión/metabolismo , Vasodilatadores/farmacología , Vasodilatadores/uso terapéutico
2.
Cell Signal ; 51: 86-98, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30075183

RESUMEN

Generation of cAMP through Gs-coupled G protein-coupled receptor (GPCR) [e.g. ß2-adrenoceptor (ß2AR), adenosine A2B receptor (A2BR)] activation, induces arterial smooth muscle relaxation, counteracting the actions of vasoconstrictors. Gs-coupled GPCR signalling is regulated by G protein-coupled receptor kinases (GRK) and arrestin proteins, and dysregulation of Gs/GPCR signalling is thought play a role in the development of hypertension, which may be a consequence of enhanced GRK2 and/or arrestin expression. However, despite numerous studies indicating that ß2AR and A2BR can be substrates for GRK/arrestin proteins, currently little is known regarding GRK/arrestin regulation of these endogenous receptors in arterial smooth muscle. Here, endogenous GRK isoenzymes and arrestin proteins were selectively depleted using RNA-interference in rat arterial smooth muscle cells (RASM) and the consequences of this for ß2AR- and A2BR-mediated adenylyl cyclase (AC) signalling were determined by assessing cAMP accumulation. GRK2 or GRK5 depletion enhanced and prolonged ß2AR/AC signalling, while combined deletion of GRK2/5 has an additive effect. Conversely, activation of AC by A2BR was regulated by GRK5, but not GRK2. ß2AR desensitization was attenuated following combined GRK2/GRK5 knockdown, but not by depletion of individual GRKs, arrestins, or by inhibiting PKA. Arrestin3 (but not arrestin2) depletion enhanced A2BR-AC signalling and attenuated A2BR desensitization, while ß2AR-AC signalling was regulated by both arrestin isoforms. This study provides a first demonstration of how different complements of GRK and arrestin proteins contribute to the regulation of signalling and desensitization of these important receptors mediating vasodilator responses in arterial smooth muscle.


Asunto(s)
Aorta/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Quinasas de Receptores Acoplados a Proteína-G/fisiología , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptor de Adenosina A2B/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Arrestina beta 2/fisiología , Adenilil Ciclasas/metabolismo , Animales , Aorta/citología , Arrestinas/genética , Arrestinas/fisiología , Células Cultivadas , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Músculo Liso/citología , Miocitos del Músculo Liso/citología , Ratas , Ratas Wistar , Transducción de Señal , Arrestina beta 2/genética
3.
Cereb Cortex ; 28(2): 421-432, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334166

RESUMEN

Impairments in social behaviors are features of a number of psychiatric diseases associated with subtle alterations in the medial prefrontal cortex (mPFC) circuitry. G protein-coupled receptor kinase (GRK) 5 is widely expressing in the cortex, however, its role in regulation of the mPFC activity and the development of social behaviors and psychiatric disorders is unclear. Here, we found that GRK5 dificiency in mice caused social behavior impairments. Further morphological, electrophysiological, and biochemical analyses showed abnormal postsynaptic ultrastructure, impaired excitatory synaptic transmission, the increased association of raptor with mTOR, and overactivated mTORC1-S6K signaling in the mPFC of Grk5-/- mice. Conditional knockdown of GRK5 in the mPFC caused impairments in social interaction and social novelty recognition behaviors; whereas selectively overexpressing GRK5 in the mPFC of Grk5-/- mice rescued the social novelty recognition phenotype. Inhibition of the overactivated mTORC1-S6K signaling pathway by rapamycin or mGluR5 antagonist ameliorated the deficiency of the excitatory synaptic transmission in the mPFC and the social recognition of Grk5-/- mice. These results indicate that GRK5 is critical for maintaining normal mTORC1 signaling and connectivity in mPFC, and normal social behavior.


Asunto(s)
Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Corteza Prefrontal/metabolismo , Transducción de Señal/fisiología , Conducta Social , Animales , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Quinasa 5 del Receptor Acoplado a Proteína-G/deficiencia , Inmunosupresores/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Corteza Prefrontal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología
4.
J Cardiovasc Pharmacol ; 70(3): 129-141, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28328744

RESUMEN

G protein-coupled receptor kinases (GRKs) are classically known for their role in regulating the activity of the largest known class of membrane receptors, which influence diverse biological processes in every cell type in the human body. As researchers have tried to uncover how this family of kinases, containing only 7 members, achieves selective and coordinated control of receptors, they have uncovered a growing number of noncanonical activities for these kinases. These activities include phosphorylation of nonreceptor targets and kinase-independent molecular interactions. In particular, GRK2, GRK3, and GRK5 are the predominant members expressed in the heart. Their canonical and noncanonical actions within cardiac and other tissues have significant implications for cardiovascular function in healthy animals and for the development and progression of disease. This review summarizes what is currently known regarding the activity of these kinases, and particularly the role of GRK2 and GRK5 in the molecular alterations that occur during heart failure. This review further highlights areas of GRK regulation that remain poorly understood and how they may represent novel targets for therapeutic development.


Asunto(s)
Enfermedades Cardiovasculares/enzimología , Quinasas de Receptores Acoplados a Proteína-G/fisiología , Transducción de Señal/fisiología , Animales , Enfermedades Cardiovasculares/patología , Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Humanos
5.
Circ Res ; 117(12): 1001-12, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26515328

RESUMEN

RATIONALE: G protein-coupled receptor kinases (GRKs) are dynamic regulators of cellular signaling. GRK5 is highly expressed within myocardium and is upregulated in heart failure. Although GRK5 is a critical regulator of cardiac G protein-coupled receptor signaling, recent data has uncovered noncanonical activity of GRK5 within nuclei that plays a key role in pathological hypertrophy. Targeted cardiac elevation of GRK5 in mice leads to exaggerated hypertrophy and early heart failure after transverse aortic constriction (TAC) because of GRK5 nuclear accumulation. OBJECTIVE: In this study, we investigated the role of GRK5 in physiological, swimming-induced hypertrophy (SIH). METHODS AND RESULTS: Cardiac-specific GRK5 transgenic mice and nontransgenic littermate control mice were subjected to a 21-day high-intensity swim protocol (or no swim sham controls). SIH and specific molecular and genetic indices of physiological hypertrophy were assessed, including nuclear localization of GRK5, and compared with TAC. Unlike after TAC, swim-trained transgenic GRK5 and nontransgenic littermate control mice exhibited similar increases in cardiac growth. Mechanistically, SIH did not lead to GRK5 nuclear accumulation, which was confirmed in vitro as insulin-like growth factor-1, a known mediator of physiological hypertrophy, was unable to induce GRK5 nuclear translocation in myocytes. We found specific patterns of altered gene expression between TAC and SIH with GRK5 overexpression. Further, SIH in post-TAC transgenic GRK5 mice was able to preserve cardiac function. CONCLUSIONS: These data suggest that although nuclear-localized GRK5 is a pathological mediator after stress, this noncanonical nuclear activity of GRK5 is not induced during physiological hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomegalia/patología , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Cardiomegalia/genética , Células Cultivadas , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología , Ratas
6.
J Biol Chem ; 289(38): 26119-26130, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25104355

RESUMEN

G protein-coupled receptor kinases 2 (GRK2) and 5 (GRK5) are fundamental regulators of cardiac performance in adults but are less well characterized for their function in the hearts of embryos. GRK2 and -5 belong to different subfamilies and function as competitors in the control of certain receptors and signaling pathways. In this study, we used zebrafish to investigate whether the fish homologs of GRK2 and -5, Grk2/3 and Grk5, also have unique, complementary, or competitive roles during heart development. We found that they differentially regulate the heart rate of early embryos and equally facilitate heart function in older embryos and that both are required to develop proper cardiac morphology. A loss of Grk2/3 results in dilated atria and hypoplastic ventricles, and the hearts of embryos depleted in Grk5 present with a generalized atrophy. This Grk5 morphant phenotype was associated with an overall decrease of early cardiac progenitors as well as a reduction in the area occupied by myocardial progenitor cells. In the case of Grk2/3, the progenitor decrease was confined to a subset of precursor cells with a committed ventricular fate. We attempted to rescue the GRK loss-of-function heart phenotypes by downstream activation of Hedgehog signaling. The Grk2/3 loss-of-function embryos were rescued by this approach, but Grk5 embryos failed to respond. In summary, we found that GRK2 and GRK5 control cardiac function as well as morphogenesis during development although with different morphological outcomes.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Corazón/embriología , Proteínas de Pez Cebra/fisiología , Animales , Proliferación Celular , Células Madre Embrionarias/fisiología , Técnicas de Silenciamiento del Gen , Corazón/crecimiento & desarrollo , Frecuencia Cardíaca , Proteínas Hedgehog/metabolismo , Morfolinos/genética , Contracción Miocárdica , Miocardio/citología , Miocardio/enzimología , Neovascularización Fisiológica , Tamaño de los Órganos , Organogénesis , Transducción de Señal , Pez Cebra
7.
Biochem Biophys Res Commun ; 429(1-2): 99-104, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23111327

RESUMEN

The G-protein-coupled receptor kinase 5 (GRK5) is an important member of the threonine/serine kinase family that phosphorylates and regulates the G-protein-coupled receptor (GPCR) signaling pathway. GRK5 is highly expressed in adipose tissue and may act as an adipogenetic factor under high-fat load [1]. Insulin resistance is associated with the pathogenesis of metabolic disorders such as type 2 diabetes and obesity; however, the potential role of GRK5 in insulin resistance is unknown. We characterized the biochemical and molecular alterations related to metabolic complications observed in GRK5(-/-) mice. These mice, which are partially resistant to obesity induced by a high-fat diet, had impaired glucose tolerance and insulin sensitivity, as well as disruption of AKT signaling transduction compared with their wild-type littermates. Further study showed that the decreased insulin sensitivity was not attributable to alterations in inflammatory status such as the NF-κB signaling pathway or inflammatory gene expression. Instead, hepatic steatosis and changes of mRNA in genes involved in hepatic glucose and lipid homeostasis were found. Overall, our data identified GRK5 as a positive regulator of insulin sensitivity. Our results showed that this protein is a potential therapeutic target in the treatment of insulin resistance and related disorders.


Asunto(s)
Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Resistencia a la Insulina , Insulina/farmacología , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Hígado Graso/genética , Hígado Graso/metabolismo , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Expresión Génica , Glucosa/metabolismo , Homeostasis , Inflamación/metabolismo , Inflamación/patología , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
8.
J Urol ; 187(1): 322-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22099983

RESUMEN

PURPOSE: The limited success of cancer therapeutics is largely attributable to the ability of cancer to become resistant to conventional cytotoxic chemotherapy. Thus, further identification of signaling molecules and pathways that influence tumorigenesis is needed to increase the overall therapeutic options. GRKs, originally recognized for their conserved role in GPCR signal control, have now emerged as regulators of additional biological molecules and functions. MATERIALS AND METHODS: We used Western blot analysis to determine GRK expression in prostate cancer and RNA interference to establish the role of GRK5 in prostate cancer growth and progression through the cell cycle. RESULTS: GRK5 was expressed highly in the aggressive prostate cancer PC3 cell line and its silencing by RNA interference attenuated in vitro cell proliferation. PC3 cells that stably expressed lentiviral small hairpin RNA and targeted GRK5 evidence reduced xenograft tumor growth in mice. This was reversed by rescuing expression with wild-type but not with kinase inactive K215R GRK5, implying the need of GRK5 kinase activity for tumor growth. To investigate possible cellular mechanism(s) for GRK5 in cell growth regulation we tested whether kinase activity would impact cell cycle progression. Like forced over expression of kinase-inactive K215R GRK5, GRK5 knockdown led to G2/M arrest in the cell cycle. Also, evidence revealed that the loss of GRK5 activity resulted in decreased cyclin D1 expression, Rb protein phosphorylation and E2F target gene expression involved in cell cycle control. CONCLUSIONS: Results provide direct evidence that GRK5 has an immediate role in the regulation of prostate tumor growth.


Asunto(s)
Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Humanos , Masculino , Ratones
9.
J Cell Biol ; 194(6): 905-20, 2011 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-21930777

RESUMEN

Neuronal morphogenesis requires extensive membrane remodeling and cytoskeleton dynamics. In this paper, we show that GRK5, a G protein-coupled receptor kinase, is critically involved in neurite outgrowth, dendrite branching, and spine morphogenesis through promotion of filopodial protrusion. Interestingly, GRK5 is not acting as a kinase but rather provides a key link between the plasma membrane and the actin cytoskeleton. GRK5 promoted filamentous actin (F-actin) bundling at the membranes of dynamic neuronal structures by interacting with both F-actin and phosphatidylinositol-4,5-bisphosphate. Moreover, separate domains of GRK5 mediated the coupling of actin cytoskeleton dynamics and membrane remodeling and were required for its effects on neuronal morphogenesis. Accordingly, GRK5 knockout mice exhibited immature spine morphology and deficient learning and memory. Our findings identify GRK5 as a critical mediator of dendritic development and suggest that coordinated actin cytoskeleton and membrane remodeling mediated by bifunctional actin-bundling and membrane-targeting molecules, such as GRK5, is crucial for proper neuronal morphogenesis and the establishment of functional neuronal circuitry.


Asunto(s)
Actinas/metabolismo , Membrana Celular/metabolismo , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Neuronas/citología , Animales , Bovinos , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Microscopía Fluorescente , Morfogénesis , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
10.
Mol Pharmacol ; 77(3): 405-15, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20038610

RESUMEN

Phosphorylation of G protein-coupled receptors (GPCRs) by GPCR kinases (GRKs) is a major mechanism of desensitization of these receptors. GPCR activation of GRKs involves an allosteric site on GRKs distinct from the catalytic site. Although recent studies have suggested an important role of the N- and C-termini and domains surrounding the kinase active site in allosteric activation, the nature of that site and the relative roles of the RH domain in particular remain unknown. Based on evolutionary trace analysis of both the RH and kinase domains of the GRK family, we identified an important cluster encompassing helices 3, 9, and 10 in the RH domain in addition to sites in the kinase domain. To define its function, a panel of GRK5 and -6 mutants was generated and screened by intact-cell assay of constitutive GRK phosphorylation of the beta(2)-adrenergic receptor (beta 2AR), in vitro GRK phosphorylation of light-activated rhodopsin, and basal catalytic activity measured by tubulin phosphorylation and autophosphorylation. A number of double mutations within helices 3, 9, and 10 reduced phosphorylation of the beta2AR and rhodopsin by 50 to 90% relative to wild-type GRK, as well as autophosphorylation and tubulin phosphorylation. Based on these results, helix 9 peptide mimetics were designed, and several were found to inhibit rhodopsin phosphorylation by GRK5 with an IC(50) of approximately 30 microM. In summary, our studies have uncovered previously unrecognized functionally important sites in the regulator of G-protein signaling homology domain of GRK5 and -6 and identified a peptide inhibitor with potential for specific blockade of GRK-mediated phosphorylation of receptors.


Asunto(s)
Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Quinasas de Receptores Acoplados a Proteína-G/fisiología , Proteínas RGS/fisiología , Receptores Adrenérgicos beta 2/metabolismo , Rodopsina/metabolismo , Secuencia de Aminoácidos , Línea Celular , Humanos , Datos de Secuencia Molecular , Fosforilación/fisiología , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Transducción de Señal/fisiología
11.
Brain Res Bull ; 78(4-5): 145-51, 2009 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-18955119

RESUMEN

Recent studies suggested that G protein-coupled receptor kinase 5 (GRK5) deficiency plays a significant role in early Alzheimer's disease (AD) pathogenesis, and that the GRK5 knockout (GRK5KO) mouse displays an early Alzheimer-like cognitive deficit associated with increased hippocampal axonal defects and synaptic degenerative changes. Gender is known to play a role in AD, with females showing more extensive pathologic changes in brain compared to males. Although GRK5 deficiency is linked to AD, it is unknown whether the pathologic changes solely driven by the GRK5 deficiency are gender-dependent. To determine this, extent of the pathologic changes in aged GRK5KO mice was compared between genders. We find that female GRK5KO mice had a 2.5-fold increase in hippocampal swollen axonal clusters compared to male GRK5KO mice. Moreover, hippocampal levels of several synaptic proteins, including synaptophysin, were significantly lower in the female than the male. In addition, although increased Luteinizing hormone (LH) activity is believed to play a significant role in the gender phenomenon in AD, we found that desensitization of LH receptor is not affected by the GRK5 deficiency. Therefore, the worsened pathologic changes in the female mice cannot be attributed to an impaired LH receptor desensitization. Taken together, this study demonstrates a synergistic interaction between GRK5 deficiency and gender in promoting early AD-like pathologic changes in the female GRK5KO mice, although the underlying molecular mechanisms remain to be elucidated.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Quinasa 5 del Receptor Acoplado a Proteína-G/deficiencia , Hipocampo/metabolismo , Sinapsis/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Axones/metabolismo , Axones/patología , Western Blotting , Modelos Animales de Enfermedad , Femenino , Fluoroinmunoensayo/métodos , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Hipocampo/patología , Indoles/metabolismo , Cinesinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Receptores de HL/metabolismo , Factores Sexuales , Sinapsis/patología , Sinaptofisina/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo
12.
Mol Pharmacol ; 75(3): 626-36, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19092051

RESUMEN

Signaling by the platelet-derived growth factor receptor-beta (PDGFRbeta) is diminished when the PDGFRbeta is phosphorylated on seryl residues by G protein-coupled receptor kinase-5 (GRK5), but mechanisms for GRK5 activation by the PDGFRbeta remain obscure. We therefore tested whether the PDGFRbeta is able to tyrosine-phosphorylate and thereby activate GRK5. Purified GRK5 was tyrosine-phosphorylated by the wild-type PDGFRbeta to a stoichiometry of 0.8 mol phosphate/mol GRK5, an extent approximately 5 times greater than observed with a Y857F PDGFRbeta mutant that fails to phosphorylate exogenous substrates but autophosphorylates and activates Src normally. The degree of PDGFRbeta-mediated phosphorylation of GRK5 correlated with GRK5 activity, as assessed by seryl phosphorylation of the PDGFRbeta in purified protein preparations, in intact cells expressing a tyrosine-to-phenylalanine GRK5 mutant, and in GRK5 peptide phosphorylation assays. However, tyrosyl phosphorylation of GRK5 was not necessary for GRK5-mediated phosphorylation of the beta(2)-adrenergic receptor, even though beta(2)-adrenergic receptor activation promoted tyrosyl phosphorylation of GRK5 in smooth muscle cells. Phosphorylation of the PDGFRbeta by GRK5 in smooth muscle cells or in purified protein preparations reduced PDGFRbeta-mediated peptide phosphorylation. In contrast, phosphorylation of GRK5 by the PDGFRbeta enhanced the V(max) of GRK5-mediated peptide phosphorylation, by 3.4-fold, without altering the GRK5 K(M) for peptide. We conclude that GRK5 tyrosyl phosphorylation is required for the activation of GRK5 by the PDGFRbeta, but not by the beta(2)-adrenergic receptor, and that by activating GRK5, the PDGFRbeta triggers its own desensitization.


Asunto(s)
Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Secuencia de Aminoácidos , Animales , Catálisis , Bovinos , Línea Celular , Quinasa 5 del Receptor Acoplado a Proteína-G/química , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Fosforilación/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/química , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Spodoptera , Especificidad por Sustrato/fisiología , Tirosina/metabolismo
13.
Proc Natl Acad Sci U S A ; 105(34): 12457-62, 2008 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-18711143

RESUMEN

G protein-coupled receptor (GPCR) kinases (GRKs) are critical regulators of cellular signaling and function. In cardiomyocytes, GRK2 and GRK5 are two GRKs important for myocardial regulation, and both have been shown to be up-regulated in the dysfunctional heart. We report that increased levels and activity of GRK5 in failing myocardium may have unique significance due to its nuclear localization, a property not shared by GRK2. We find that transgenic mice with elevated cardiac GRK5 levels have exaggerated hypertrophy and early heart failure compared with control mice after pressure overload. This pathology is not present in cardiac GRK2-overexpressing mice or in mice with overexpression of a mutant GRK5 that is excluded from the nucleus. Nuclear accumulation of GRK5 is enhanced in myocytes after aortic banding in vivo and in vitro in myocytes after increased G alpha q activity, the trigger for pressure-overload hypertrophy. GRK5 enhances activation of MEF2 in concert with Gq signals, demonstrating that nuclear localized GRK5 regulates gene transcription via a pathway critically linked to myocardial hypertrophy. Mechanistically, we show that this is due to GRK5 acting, in a non-GPCR manner, as a class II histone deacetylase (HDAC) kinase because it can associate with and phosphorylate the myocyte enhancer factor-2 repressor, HDAC5. Moreover, significant HDAC activity can be found with GRK5 in the heart. Our data show that GRK5 is a nuclear HDAC kinase that plays a key role in maladaptive cardiac hypertrophy apparently independent of any action directly on GPCRs.


Asunto(s)
Núcleo Celular/enzimología , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Miocitos Cardíacos/enzimología , Animales , Quinasa 5 del Receptor Acoplado a Proteína-G/análisis , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/etiología , Histona Desacetilasas/metabolismo , Hipertrofia/enzimología , Hipertrofia/etiología , Factores de Transcripción MEF2 , Ratones , Ratones Transgénicos , Miocitos Cardíacos/ultraestructura , Factores Reguladores Miogénicos/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA