Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Am J Pathol ; 193(11): 1817-1832, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37423551

RESUMO

Annexin A1 (AnxA1) is the primary mediator of the anti-inflammatory actions of glucocorticoids. AnxA1 functions as a pro-resolving mediator in cultured rat conjunctival goblet cells to ensure tissue homeostasis through stimulation of intracellular [Ca2+] ([Ca2+]i) and mucin secretion. AnxA1 has several N-terminal peptides with anti-inflammatory properties of their own, including Ac2-26, Ac2-12, and Ac9-25. The increase in [Ca2+]i caused by AnxA1 and its N-terminal peptides in goblet cells was measured to determine the formyl peptide receptors used by the compounds and the action of the peptides on histamine stimulation. Changes in [Ca2+]i were determined by using a fluorescent Ca2+ indicator. AnxA1 and its peptides each activated formyl peptide receptors in goblet cells. AnxA1 and Ac2-26 at 10-12 mol/L and Ac2-12 at 10-9 mol/L inhibited the histamine-stimulated increase in [Ca2+]i, as did resolvin D1 and lipoxin A4 at 10-12 mol/L, whereas Ac9-25 did not. AnxA1 and Ac2-26 counter-regulated the H1 receptor through the p42/p44 mitogen-activated protein kinase/extracellular regulated kinase 1/2, ß-adrenergic receptor kinase, and protein kinase C pathways, whereas Ac2-12 counter-regulated only through ß-adrenergic receptor kinase. In conclusion, current data show that the N-terminal peptides Ac2-26 and Ac2-12, but not Ac9-25, share multiple functions with the full-length AnxA1 in goblet cells, including inhibition of histamine-stimulated increase in [Ca2+]i and counter-regulation of the H1 receptor. These actions suggest a potential pharmaceutical application of the AnxA1 N-terminal peptides Ac2-26 and Ac2-12 in homeostasis and ocular inflammatory diseases.


Assuntos
Anexina A1 , Ratos , Animais , Anexina A1/farmacologia , Anexina A1/química , Anexina A1/metabolismo , Células Caliciformes/metabolismo , Receptores de Formil Peptídeo/metabolismo , Histamina/farmacologia , Peptídeos/farmacologia , Anti-Inflamatórios/farmacologia , Quinases de Receptores Adrenérgicos beta/metabolismo
2.
Neuron ; 109(14): 2256-2274.e9, 2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34139149

RESUMO

Astrocytes respond to neurotransmitters and neuromodulators using G-protein-coupled receptors (GPCRs) to mediate physiological responses. Despite their importance, there has been no method to genetically, specifically, and effectively attenuate astrocyte Gq GPCR pathways to explore consequences of this prevalent signaling mechanism in vivo. We report a 122-residue inhibitory peptide from ß-adrenergic receptor kinase 1 (ißARK; and inactive D110A control) to attenuate astrocyte Gq GPCR signaling. ißARK significantly attenuated Gq GPCR Ca2+ signaling in brain slices and, in vivo, altered behavioral responses, spared other GPCR responses, and did not alter astrocyte spontaneous Ca2+ signals, morphology, electrophysiological properties, or gene expression in the striatum. Furthermore, brain-wide attenuation of astrocyte Gq GPCR signaling with ißARK using PHP.eB adeno-associated viruses (AAVs), when combined with c-Fos mapping, suggested nuclei-specific contributions to behavioral adaptation and spatial memory. ißARK extends the toolkit needed to explore functions of astrocyte Gq GPCR signaling within neural circuits in vivo.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Quinases de Receptores Adrenérgicos beta/metabolismo , Animais , Cálcio/metabolismo , Camundongos , Neurônios/metabolismo
3.
Mol Pharmacol ; 98(4): 497-507, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32362586

RESUMO

Based on studies using mutations of the µ-opioid receptor (MOR), phosphorylation of multiple sites on the C-terminus has been recognized as a critical step underlying acute desensitization and the development of cellular tolerance. The aim of this study is to explore which kinases mediate desensitization of MOR in brain slices from drug-naïve and morphine-treated animals. Whole-cell recordings from locus coeruleus neurons were made, and the agonist-induced increase in potassium conductance was measured. In slices from naïve animals, pharmacological inhibition of G-protein receptor kinase (GRK2/3) with compound 101 blocked acute desensitization. Following chronic treatment with morphine, compound 101 was less effective at blocking acute desensitization. Compound 101 blocked receptor internalization in tissue from both naïve and morphine-treated animals, suggesting that GRK2/3 remained active. Kinase inhibitors aimed at blocking protein kinase C and c-Jun N-terminal kinase had no effect on desensitization in tissue taken from naïve animals. However, in slices taken from morphine-treated animals, the combination of these blockers along with compound 101 was required to block acute desensitization. Acute desensitization of the potassium conductance induced by the somatostatin receptor was also blocked by compound 101 in slices from naïve but not morphine-treated animals. As was observed with MOR, it was necessary to use the combination of kinase inhibitors to block desensitization of the somatostatin receptor in slices from morphine-treated animals. The results show that chronic treatment with morphine results in a surprising and heterologous adaptation in kinase-dependent desensitization. SIGNIFICANCE STATEMENT: The results show that chronic treatment with morphine induced heterologous adaptations in kinase regulation of G protein coupled receptor (GPCR) desensitization. Although the canonical mechanism for acute desensitization through phosphorylation by G protein-coupled receptor kinase is supported in tissue taken from naïve animals, following chronic treatment with morphine, the acute kinase-dependent desensitization of GPCRs is disrupted such that additional kinases, including protein kinase C and c-Jun N-terminal kinase, contribute to desensitization.


Assuntos
Locus Cerúleo/metabolismo , Morfina/administração & dosagem , Receptores Opioides mu/metabolismo , Quinases de Receptores Adrenérgicos beta/metabolismo , Animais , Tolerância a Medicamentos , Feminino , Locus Cerúleo/efeitos dos fármacos , Masculino , Morfina/farmacologia , Técnicas de Patch-Clamp , Fosforilação , Potássio/metabolismo , Ratos
4.
Mol Biol Rep ; 47(6): 4631-4650, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32303958

RESUMO

Neoplastically transformed astrocytes express functionally active cell surface ß adrenergic receptors (ßARs). Treatment of glioma models in vitro and in vivo with ß adrenergic agonists variably amplifies or attenuates cellular proliferation. In the majority of in vivo models, ß adrenergic agonists generally reduce cellular proliferation. However, treatment with ß adrenergic agonists consistently reduces tumor cell invasive potential, angiogenesis, and metastasis. ß adrenergic agonists induced decreases of invasive potential are chiefly mediated through reductions in the expression of matrix metalloproteinases types 2 and 9. Treatment with ß adrenergic agonists also clearly reduce tumoral neoangiogenesis, which may represent a putatively useful mechanism to adjuvantly amplify the effects of bevacizumab. Bevacizumab is a monoclonal antibody targeting the vascular endothelial growth factor receptor. We may accordingly designate ßagonists to represent an enhancer of bevacizumab. The antiangiogenic effects of ß adrenergic agonists may thus effectively render an otherwise borderline effective therapy to generate significant enhancement in clinical outcomes. ß adrenergic agonists upregulate expression of the major histocompatibility class II DR alpha gene, effectively potentiating the immunogenicity of tumor cells to tumor surveillance mechanisms. Authors have also demonstrated crossmodal modulation of signaling events downstream from the ß adrenergic cell surface receptor and microtubular polymerization and depolymerization. Complex effects and desensitization mechanisms of the ß adrenergic signaling may putatively represent promising therapeutic targets. Constant stimulation of the ß adrenergic receptor induces its phosphorylation by ß adrenergic receptor kinase (ßARK), rendering it a suitable substrate for alternate binding by ß arrestins 1 or 2. The binding of a ß arrestin to ßARK phosphorylated ßAR promotes receptor mediated internalization and downregulation of cell surface receptor and contemporaneously generates a cell surface scaffold at the ßAR. The scaffold mediated activation of extracellular regulated kinase 1/2, compared with protein kinase A mediated activation, preferentially favors cytosolic retention of ERK1/2 and blunting of nuclear translocation and ensuant pro-transcriptional activity. Thus, ßAR desensitization and consequent scaffold assembly effectively retains the cytosolic homeostatic functions of ERK1/2 while inhibiting its pro-proliferative effects. We suggest these mechanisms specifically will prove quite promising in developing primary and adjuvant therapies mitigating glioma growth, angiogenesis, invasive potential, and angiogenesis. We suggest generating compounds and targeted mutations of the ß adrenergic receptor favoring ß arrestin binding and scaffold facilitated activation of ERK1/2 may hold potential promise and therapeutic benefit in adjuvantly treating most or all cancers. We hope our discussion will generate fruitful research endeavors seeking to exploit these mechanisms.


Assuntos
Glioma/metabolismo , Receptores Adrenérgicos beta/metabolismo , beta-Arrestinas/metabolismo , Agonistas Adrenérgicos beta/metabolismo , Animais , Carcinoma/genética , Carcinoma/metabolismo , Proliferação de Células , Glioma/genética , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Transporte Proteico , Receptores Adrenérgicos beta 2/genética , Transdução de Sinais/efeitos dos fármacos , Neoplasias da Coluna Vertebral/genética , Neoplasias da Coluna Vertebral/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Quinases de Receptores Adrenérgicos beta/metabolismo , beta-Arrestinas/fisiologia
5.
Nat Commun ; 6: 8480, 2015 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-26412387

RESUMO

Optogenetics has provided a revolutionary approach to dissecting biological phenomena. However, the generation and use of optically active GPCRs in these contexts is limited and it is unclear how well an opsin-chimera GPCR might mimic endogenous receptor activity. Here we show that a chimeric rhodopsin/ß2 adrenergic receptor (opto-ß2AR) is similar in dynamics to endogenous ß2AR in terms of: cAMP generation, MAP kinase activation and receptor internalization. In addition, we develop and characterize a novel toolset of optically active, functionally selective GPCRs that can bias intracellular signalling cascades towards either G-protein or arrestin-mediated cAMP and MAP kinase pathways. Finally, we show how photoactivation of opto-ß2AR in vivo modulates neuronal activity and induces anxiety-like behavioural states in both fiber-tethered and wireless, freely moving animals when expressed in brain regions known to contain ß2ARs. These new GPCR approaches enhance the utility of optogenetics and allow for discrete spatiotemporal control of GPCR signalling in vitro and in vivo.


Assuntos
Optogenética , Quinases de Receptores Adrenérgicos beta/metabolismo , Animais , Ansiedade/enzimologia , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo
6.
J Cell Mol Med ; 18(11): 2135-46, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25216213

RESUMO

The cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKA) is an elementary molecule involved in both acute and chronic modulation of cardiac function. Substantial research in recent years has highlighted the importance of A-kinase anchoring proteins (AKAP) therein as they act as the backbones of major macromolecular signalling complexes of the ß-adrenergic/cAMP/PKA pathway. This review discusses the role of AKAP-associated protein complexes in acute and chronic cardiac modulation by dissecting their role in altering the activity of different ion channels, which underlie cardiac action potential (AP) generation. In addition, we review the involvement of different AKAP complexes in mechanisms of cardiac remodelling and arrhythmias.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Eletrofisiologia , Coração/fisiopatologia , Miocárdio/enzimologia , Potenciais de Ação , AMP Cíclico/metabolismo , Humanos , Canais Iônicos/metabolismo , Miocárdio/metabolismo , Transdução de Sinais , Quinases de Receptores Adrenérgicos beta/metabolismo
7.
Resuscitation ; 83(5): 640-4, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22115934

RESUMO

OBJECTIVE: Post-resuscitation myocardial dysfunction is a major cause of fatality in patients receiving successful cardiopulmonary resuscitation. The mechanism of post-resuscitation myocardial dysfunction is largely unknown, although is generally considered related to ischaemia occurring during cardiac arrest and resuscitation and/or reperfusion injury after restoration of circulation. A key mechanism responsible for reduced contractile reserves in chronic heart failure is impaired ß-adrenergic receptor signalling. Thus, we hypothesised that ß-adrenergic receptor signalling is markedly abnormal in the post-resuscitation period following cardiopulmonary resuscitation. METHODS: Male landrace domestic pigs were randomised into a sham group (anaesthetised and instrumented, no ventricular fibrillation) or cardiopulmonary resuscitation (CPR) group (ventricular fibrillation) (n=8 per group). Haemodynamic and echocardiographic data were recorded. ß-Adrenergic receptor signalling was assessed at 6h after the operation by measuring myocardial adenylate cyclase activity, ß-adrenergic receptor density and ß-adrenergic receptor kinase expression. RESULTS: Left ventricular function in the CPR group was significantly decreased at 6 h after restoration of spontaneous circulation. Basal and isoproterenol-stimulated adenylate cyclase activity was blunted in the CPR group compared with the sham group. Total ß-AR density was significantly decreased in CPR group compared with the sham group. Myocardial ß-adrenergic receptor kinase expression was 2.03-fold greater in the CPR group than in the sham group. CONCLUSIONS: ß-Adrenergic receptor signalling is markedly impaired in the post-resuscitation period, which may be a mechanism of post-resuscitation myocardial dysfunction.


Assuntos
Quinases de Receptores Acoplados a Proteína G/metabolismo , Parada Cardíaca/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Fibrilação Ventricular/metabolismo , Quinases de Receptores Adrenérgicos beta/metabolismo , Adenilil Ciclases/metabolismo , Animais , Reanimação Cardiopulmonar , Ecocardiografia , Parada Cardíaca/fisiopatologia , Hemodinâmica , Immunoblotting , Masculino , Infarto do Miocárdio/fisiopatologia , Placebos , Ensaio Radioligante , Ressuscitação , Transdução de Sinais , Suínos , Função Ventricular Esquerda
8.
J Mol Cell Cardiol ; 51(4): 462-7, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21256851

RESUMO

Heart failure (HF) is devastating disease with poor prognosis. Elevated sympathetic nervous system activity and outflow, leading to pathologic attenuation and desensitization of ß-adrenergic receptors (ß-ARs) signaling and responsiveness, are salient characteristic of HF progression. These pathologic effects on ß-AR signaling and HF progression occur in part due to Gßγ-mediated signaling, including recruitment of receptor desensitizing kinases such as G-protein coupled receptor (GPCR) kinase 2 (GRK2) and phosphoinositide 3-kinase (PI3K), which subsequently phosphorylate agonist occupied GPCRs. Additionally, chronic GPCR signaling signals chronically dissociated Gßγ subunits to interact with multiple effector molecules that activate various signaling cascades involved in HF pathophysiology. Importantly, targeting Gßγ signaling with large peptide inhibitors has proven a promising therapeutic paradigm in the treatment of HF. We recently described an approach to identify small molecule Gßγ inhibitors that selectively block particular Gßγ functions by specifically targeting a Gßγ protein-protein interaction "hot spot." Here we describe their effects on Gßγ downstream signaling pathways, including their role in HF pathophysiology. We suggest a promising therapeutic role for small molecule inhibition of pathologic Gßγ signaling in the treatment of HF. This article is part of a special issue entitled "Key Signaling Molecules in Hypertrophy and Heart Failure."


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Terapia de Alvo Molecular , Antagonistas Adrenérgicos beta/uso terapêutico , Animais , Fármacos Cardiovasculares/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Subunidades beta da Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidades gama da Proteína de Ligação ao GTP/antagonistas & inibidores , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases de Receptores Adrenérgicos beta/antagonistas & inibidores , Quinases de Receptores Adrenérgicos beta/metabolismo
9.
J Cell Mol Med ; 15(2): 258-69, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20015194

RESUMO

Inducible nitric oxide synthase (iNOS) is a major source of nitric oxide during inflammation whose activity is thought to be controlled primarily at the expression level. The B1 kinin receptor (B1R) post-translationally activates iNOS beyond its basal activity via extracellular signal regulated kinase (ERK)-mediated phosphorylation of Ser(745) . Here we identified the signalling pathway causing iNOS activation in cytokine-treated endothelial cells or HEK293 cells transfected with iNOS and B1R. To allow kinetic measurements of nitric oxide release, we used a sensitive porphyrinic microsensor (response time = 10 msec.; 1 nM detection limit). B1Rs signalled through Gαi coupling as ERK and iNOS activation were inhibited by pertussis toxin. Furthermore, transfection of constitutively active mutant Gαi Q204L but not Gαq Q209L resulted in high basal iNOS-derived nitric oxide. G-ßγ subunits were also necessary as transfection with the ß-adrenergic receptor kinase C-terminus inhibited the response. B1R-dependent iNOS activation was also inhibited by Src family kinase inhibitor PP2 and trans-fection with dominant negative Src. Other ERK-MAP kinase members were involved as the response was inhibited by dominant negative H-Ras, Raf kinase inhibitor, ERK activation inhibitor and MEK inhibitor PD98059. In contrast, PI3 kinase inhibitor LY94002, calcium chelator 1,2-bis-(o-Aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid, tetraacetoxymethyl ester (BAPTA-AM), protein kinase C inhibitor calphostin C and protein kinase C activator PMA had no effect. Angiotensin converting enzyme inhibitor enalaprilat also directly activated B1Rs to generate high output nitric oxide via the same pathway. These studies reveal a new mechanism for generating receptor-regulated high output nitric oxide in inflamed endothelium that may play an important role in the development of vascular inflammation.


Assuntos
Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/biossíntese , Receptor B1 da Bradicinina/metabolismo , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Linhagem Celular , Citocinas/farmacologia , Enalaprilato/farmacologia , Células Endoteliais/metabolismo , Ativação Enzimática , Flavonoides/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Pirimidinas/farmacologia , Receptor B1 da Bradicinina/agonistas , Receptor B1 da Bradicinina/genética , Transfecção , Quinases de Receptores Adrenérgicos beta/metabolismo , Quinases da Família src/antagonistas & inibidores
10.
J Cell Physiol ; 226(1): 181-6, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20677219

RESUMO

This study was undertaken to determine alterations in the ß-adrenoceptor (ß-AR) signaling system in male and female rats at 4 weeks after the induction of arteriovenous (AV) fistula or shunt. AV shunt produced a greater degree of cardiac hypertrophy and larger increase in cardiac output in male than in female animals. Increases in plasma levels of norepinephrine and epinephrine (EPI) due to AV shunt were also higher in male than females. While no difference in the ß(1)-AR affinity was seen in males and females, AV shunt induced increase in ß(1)-AR density in female rats was higher than that in males. Furthermore, no changes in basal adenylyl cyclase (AC) V/VI mRNA levels were seen; however, the increase in EPI-stimulated AC activities was greater in AV shunt females than in males. AV shunt decreased myocardial ß(1)-AR mRNA level in male rats and increased ß(2)-AR mRNA level in female hearts; an increase in G(i)-protein mRNA was detected only in male hearts. Although GRK2 gene expression was increased in both sexes, an increase in GRK3 mRNA was seen only in AV shunt female rats. ß-arrestin1 mRNA was elevated in females whereas ß-arrestin 2 gene expression was increased in both male and female AV shunt rats. While these data demonstrate gender associated differences in various components of the ß-AR system in cardiac hypertrophy due to AV shunt, only higher levels of plasma catecholamines may account for the greater increase in cardiac output and higher degree of cardiac hypertrophy in males.


Assuntos
Fístula Arteriovenosa/complicações , Fístula Arteriovenosa/metabolismo , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Receptores Adrenérgicos beta/metabolismo , Caracteres Sexuais , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Animais , Arrestinas/genética , Arrestinas/metabolismo , Débito Cardíaco , Cardiomegalia/fisiopatologia , Catecolaminas/sangue , Ecocardiografia , Feminino , Regulação da Expressão Gênica/fisiologia , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta/genética , Remodelação Ventricular/fisiologia , Quinases de Receptores Adrenérgicos beta/genética , Quinases de Receptores Adrenérgicos beta/metabolismo , beta-Arrestina 2 , beta-Arrestinas
11.
J Biol Chem ; 285(53): 41290-9, 2010 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-21044958

RESUMO

G protein-activated inwardly rectifying potassium (GIRK or Kir3) channels are directly gated by the ßγ subunits of G proteins and contribute to inhibitory neurotransmitter signaling pathways. Paradoxically, volatile anesthetics such as halothane inhibit these channels. We find that neuronal Kir3 currents are highly sensitive to inhibition by halothane. Given that Kir3 currents result from increased Gßγ available to the channels, we asked whether reducing available Gßγ to the channel would adversely affect halothane inhibition. Remarkably, scavenging Gßγ using the C-terminal domain of ß-adrenergic receptor kinase (cßARK) resulted in channel activation by halothane. Consistent with this effect, channel mutants that impair Gßγ activation were also activated by halothane. A single residue, phenylalanine 192, occupies the putative Gßγ gate of neuronal Kir3.2 channels. Mutation of Phe-192 at the gate to other residues rendered the channel non-responsive, either activated or inhibited by halothane. These data indicated that halothane predominantly interferes with Gßγ-mediated Kir3 currents, such as those functioning during inhibitory synaptic activity. Our report identifies the molecular correlate for anesthetic inhibition of Kir3 channels and highlights the significance of these effects in modulating neurotransmitter-mediated inhibitory signaling.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/química , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Anestésicos , Animais , Sítios de Ligação , Linhagem Celular , Halotano/química , Hipocampo/metabolismo , Humanos , Neurotransmissores/química , Oócitos/metabolismo , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , Xenopus , Quinases de Receptores Adrenérgicos beta/metabolismo
12.
Sci Signal ; 3(121): ra37, 2010 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-20460648

RESUMO

Evidence from Drosophila and cultured cell studies supports a role for heterotrimeric guanosine triphosphate-binding proteins (G proteins) in Wnt signaling. Wnt inhibits the degradation of the transcriptional regulator beta-catenin. We screened the alpha and betagamma subunits of major families of G proteins in a Xenopus egg extract system that reconstitutes beta-catenin degradation. We found that Galpha(o), Galpha(q), Galpha(i2), and Gbetagamma inhibited beta-catenin degradation. Gbeta(1)gamma(2) promoted the phosphorylation and activation of the Wnt co-receptor low-density lipoprotein receptor-related protein 6 (LRP6) by recruiting glycogen synthase kinase 3 (GSK3) to the membrane and enhancing its kinase activity. In both a reporter gene assay and an in vivo assay, c-betaARK (C-terminal domain of beta-adrenergic receptor kinase), an inhibitor of Gbetagamma, blocked LRP6 activity. Several components of the Wnt-beta-catenin pathway formed a complex: Gbeta(1)gamma(2), LRP6, GSK3, axin, and dishevelled. We propose that free Gbetagamma and Galpha subunits, released from activated G proteins, act cooperatively to inhibit beta-catenin degradation and activate beta-catenin-mediated transcription.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Relacionadas a Receptor de LDL/biossíntese , Transcrição Gênica/fisiologia , beta Catenina/metabolismo , Animais , Drosophila melanogaster , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Quinase 3 da Glicogênio Sintase/genética , Células HeLa , Humanos , Proteínas Relacionadas a Receptor de LDL/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Transdução de Sinais/fisiologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Xenopus laevis , beta Catenina/genética , Quinases de Receptores Adrenérgicos beta/genética , Quinases de Receptores Adrenérgicos beta/metabolismo
13.
Mol Endocrinol ; 24(2): 436-46, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20051480

RESUMO

Pasireotide (SOM230) is currently under clinical evaluation as a successor compound to octreotide for the treatment of acromegaly, Cushing's disease, and carcinoid tumors. Whereas octreotide acts primarily via the sst(2A) somatostatin receptor, pasireotide was designed to exhibit octreotide-like sst(2A) activity combined with enhanced binding to other somatostatin receptor subtypes. In the present study, we used phophosite-specific antibodies to examine agonist-induced phosphorylation of the rat sst(2A) receptor. We show that somatostatin and octreotide stimulate the complete phosphorylation of a cluster of four threonine residues within the cytoplasmic (353)TTETQRT(359) motif in a variety of cultured cell lines in vitro as well as in intact animals in vivo. This phosphorylation was mediated by G protein-coupled receptor kinases (GRK) 2 and 3 and followed by rapid cointernalization of the receptor and ss-arrestin into the same endocytic vesicles. In contrast, pasireotide failed to promote substantial phosphorylation and internalization of the rat sst(2A) receptor. In the presence of octreotide or SS-14, SOM230 showed partial agonist behavior, inhibiting phosphorylation, and internalization of sst(2A). Upon overexpression of GRK2 or GRK3, pasireotide stimulated selective phosphorylation of Thr356 and Thr359 but not of Thr353 or Thr354 within the (353)TTETQRT(359) motif. Pasireotide-mediated phosphorylation led to the formation of relatively unstable beta-arrestin-sst(2A) complexes that dissociated at or near the plasma membrane. Thus, octreotide and pasireotide are equally active in inducing classical G protein-dependent signaling via the sst(2A) somatostatin receptor. Yet, we find that they promote strikingly different patterns of sst(2A) receptor phosphorylation and, hence, stimulate functionally distinct pools of beta-arrestin.


Assuntos
Octreotida/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Somatostatina/agonistas , Animais , Anticorpos Fosfo-Específicos/isolamento & purificação , Arrestinas/genética , Arrestinas/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Endocitose/efeitos dos fármacos , Humanos , Ligantes , Masculino , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Fosforilação/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Hipófise/patologia , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional/genética , Ratos , Ratos Wistar , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inibidores , Receptores de Somatostatina/genética , Somatostatina/antagonistas & inibidores , Somatostatina/farmacologia , Treonina/genética , Treonina/metabolismo , Quinases de Receptores Adrenérgicos beta/antagonistas & inibidores , Quinases de Receptores Adrenérgicos beta/genética , Quinases de Receptores Adrenérgicos beta/metabolismo , beta-Arrestinas
14.
Cell Calcium ; 47(1): 55-64, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20056275

RESUMO

We have previously reported that P2Y(2) purinoceptors and muscarinic M(3) receptors trigger Ca(2+) responses in HT-29 cells that differ in their timecourse, the Ca(2+) response to P2Y(2) receptor activation being marked by a more rapid decline of intracellular Ca(2+) concentration ([Ca(2+)](i)) after the peak response and that this rapid decline of [Ca(2+)](i) was slowed in cells expressing heterologous beta-adrenergic receptor kinase (betaARK). In the present study, we demonstrate that, during P2Y(2) receptor activation, betaARK expression increases the rate of Gd(3+)-sensitive Mn(2+) influx, a measure of the rate of store-operated Ca(2+) entry from the extracellular space, during P2Y(2) activation and that this effect of betaARK is mimicked by exogenous alpha-subunits of G(q), G(11) and G(i2). The effect of betaARK on the rate of Mn(2+) influx is thus attributable to its ability to scavenge G protein betagamma-subunits released during activation of P2Y(2) receptor. We further find that the effect of betaARK on the rate of Mn(2+) influx during P2Y(2) receptor activation can be overcome by arachidonic acid. In addition, the UTP-induced Mn(2+) influx rate was significantly increased by inhibitors of phospholipase A(2) (PLA(2)) and an siRNA directed against PLA(2)beta, but not by an siRNA directed against PLA(2)alpha or by inhibitors of arachidonic acid metabolism. These findings provide evidence for the existence of a P2Y(2) receptor-activated signalling system that acts in parallel with depletion of intracellular Ca(2+) stores to inhibit Ca(2+) influx across the cell membrane. This signalling process is mediated via Gbetagamma and involves PLA(2)beta and arachidonic acid.


Assuntos
Células Epiteliais/metabolismo , Retroalimentação Fisiológica , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Receptores Purinérgicos P2/metabolismo , Trifosfato de Adenosina/metabolismo , Ácido Araquidônico/farmacologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Engenharia Genética , Fosfolipases A2 do Grupo IV/genética , Fosfolipases A2 do Grupo IV/metabolismo , Células HT29 , Humanos , Manganês/metabolismo , RNA Interferente Pequeno/genética , Receptor Muscarínico M3/metabolismo , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y2 , Quinases de Receptores Adrenérgicos beta/genética , Quinases de Receptores Adrenérgicos beta/metabolismo
15.
Auton Autacoid Pharmacol ; 28(1): 29-39, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18257749

RESUMO

1 The role of the protein tyrosine kinase, c-Src, on the function and phosphorylation of alpha1B-adrenoceptors (alpha1B-AR) and their association with G-protein-coupled receptor kinase (GRK) isozymes was studied. 2 Inhibitors of this kinase (PP2 and Src Inhibitor II) decreased ( approximately 50-75%) noradrenaline- (NA) and phorbol myristate acetate-mediated receptor phosphorylation. Expression of a dominant-negative mutant of c-Src similarly reduced receptor phosphorylation induced by the natural agonists, active phorbol esters and endothelin-1 (ET-1). 3 c-Src, GRK2, GRK3 and GRK5 coimmunoprecipitate with alpha1B-ARs in the basal state. In cells treated with NA or phorbol myristate acetate the amount of coimmunoprecipitated GRK2 and GRK3 increased ( approximately 2- to 3-fold), while treatment with ET-1 only augmented the amount of coimmunoprecipitated GRK2 ( approximately 2-fold). The Src inhibitor, PP2, markedly attenuated all these increases. 4 Cell pretreatment with PP2 amplified the increase in intracellular-free calcium observed with NA, in the basal state and after the stimulation (desensitization) induced by ET-1. 5 The data suggest a role of c-Src in alpha1B-AR desensitization/phosphorylation and in the interaction of these ARs with GRKs.


Assuntos
Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Quinases de Receptores Adrenérgicos beta/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1 , Animais , Células COS , Cálcio/metabolismo , Chlorocebus aethiops , Cricetinae , Relação Dose-Resposta a Droga , Endotelina-1/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Norepinefrina/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas pp60(c-src)/antagonistas & inibidores , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Pirimidinas/farmacologia , Ratos , Receptores Adrenérgicos alfa 1/genética , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo , Transfecção
16.
Physiol Genomics ; 32(2): 182-9, 2008 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-17971502

RESUMO

Numerous genetically engineered animal models of heart failure (HF) exhibit multiple characteristics of human HF, including aberrant beta-adrenergic signaling. Several of these HF models can be rescued by cardiac-targeted expression of the Gbetagamma inhibitory carboxy-terminus of the beta-adrenergic receptor kinase (betaARKct). We recently reported microarray analysis of gene expression in multiple animal models of HF and their betaARKct rescue, where we identified gene expression patterns distinct and predictive of HF and rescue. We have further investigated the muscle LIM protein knockout model of HF (MLP-/-), which closely parallels human dilated cardiomyopathy disease progression and aberrant beta-adrenergic signaling, and their betaARKct rescue. A group of known and novel genes was identified and validated by quantitative real-time PCR whose expression levels predicted phenotype in both the larger HF group and in the MLP-/- subset. One of these novel genes is herein identified as Nogo, a protein widely studied in the nervous system, where it plays a role in regeneration. Nogo expression is altered in HF and normalized with rescue, in an isoform-specific manner, using left ventricular tissue harvested from both animal and human subjects. To investigate cell type-specific expression of Nogo in the heart, immunofluorescence and confocal microscopy were utilized. Nogo expression appears to be most clearly associated with cardiac fibroblasts. To our knowledge, this is the first report to demonstrate the relationship between Nogo expression and HF, including cell-type specificity, in both mouse and human HF and phenotypic rescue.


Assuntos
Insuficiência Cardíaca/patologia , Proteínas Musculares/genética , Proteínas da Mielina/genética , Miocárdio/patologia , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Deleção de Genes , Perfilação da Expressão Gênica , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Humanos , Imuno-Histoquímica , Proteínas com Domínio LIM , Masculino , Camundongos , Camundongos Knockout , Proteínas Musculares/metabolismo , Proteínas da Mielina/metabolismo , Miocárdio/metabolismo , Proteínas Nogo , Análise de Sequência com Séries de Oligonucleotídeos , Quinases de Receptores Adrenérgicos beta/genética , Quinases de Receptores Adrenérgicos beta/metabolismo
17.
J Card Fail ; 13(5): 401-14, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17602988

RESUMO

Heart failure (HF) is a leading cause of morbidity and mortality in Western countries and projections reveal that HF incidence in the coming years will rise significantly because of an aging population. Pharmacologic therapy has considerably improved HF treatment during the last 2 decades, but fails to rescue failing myocardium and to increase global cardiac function. Therefore, novel therapeutic approaches to target the underlying molecular defects of ventricular dysfunction and to increase the outcome of patients in HF are needed. Failing myocardium generally exhibits distinct changes in beta-adrenergic receptor (betaAR) signaling and intracellular Ca2+-handling providing opportunities for research. Recent advances in transgenic and gene therapy techniques have presented novel therapeutic strategies to alter myocardial function and to target both betaAR signaling and Ca2+-cycling. In this review, we will discuss functional alterations of the betaAR system and Ca2+-handling in HF as well as corresponding therapeutic strategies. We will then focus on recent in vivo gene therapy strategies using the targeted inhibition of the betaAR kinase (betaARK1 or GRK2) and the restoration of S100A1 protein expression to support the injured heart and to reverse or prevent HF.


Assuntos
Cálcio/fisiologia , Receptor Quinase 1 Acoplada a Proteína G/fisiologia , Terapia Genética , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Receptores Adrenérgicos beta/fisiologia , Animais , Cálcio/metabolismo , Cardiomiopatia Hipertrófica/fisiopatologia , Cardiomiopatia Hipertrófica/terapia , Modelos Animais de Doenças , Receptor Quinase 1 Acoplada a Proteína G/sangue , Quinase 2 de Receptor Acoplado a Proteína G , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Insuficiência Cardíaca/metabolismo , Humanos , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Proteínas S100/metabolismo , Quinases de Receptores Adrenérgicos beta/metabolismo , Quinases de Receptores Adrenérgicos beta/fisiologia
18.
Mol Pharmacol ; 72(3): 582-91, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17573483

RESUMO

The closely related G protein-coupled receptor kinases GRK2 and GRK3 are both expressed in cardiac myocytes. Although GRK2 has been extensively investigated in terms of regulation of cardiac beta-adrenergic receptors, the substrate specificities of the two GRK isoforms at G protein-coupled receptors (GPCR) are poorly understood. In this study, the substrate specificities of GRK2 and GRK3 at GPCRs that control cardiac myocyte function were determined in fully differentiated adult cardiac myocytes. Concentration-effect relationships of GRK2, GRK3, and their respective competitive inhibitors, GRK2ct and GRK3ct, at endogenous endothelin, alpha(1)-adrenergic, and beta(1)-adrenergic receptor-generated responses in cardiac myocytes were achieved by adenovirus gene transduction. GRK3 and GRK3ct were highly potent and efficient at the endothelin receptors (IC(50) for GRK3, 5 +/- 0.7 pmol/mg of protein; EC(50) for GRK3ct, 2 +/- 0.2 pmol/mg of protein). The alpha(1)-adrenergic receptor was also a preferred substrate of GRK3 (IC(50),7 +/- 0.4 pmol/mg of protein). GRK2 lacked efficacy at both endothelin and alpha(1)-adrenergic receptors despite massive overexpression. On the contrary, both GRK2ct and GRK3ct enhanced beta(1)-adrenergic receptor-induced cAMP production with comparable potencies. However, the potency of GRK3ct at beta(1)-adrenergic receptors was at least 20-fold lower than that at endothelin receptors. In conclusion, this study demonstrates distinct substrate specificities of GRK2 and GRK3 at different GPCRs in fully differentiated adult cardiac myocytes. As inferred from the above findings, GRK2 may play its primary role in regulation of cardiac contractility and chronotropy by controlling beta(1)-adrenergic receptors, whereas GRK3 may play important roles in regulation of cardiac growth and hypertrophy by selectively controlling endothelin and alpha(1)-adrenergic receptors.


Assuntos
Regulação Enzimológica da Expressão Gênica , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Quinases de Receptores Adrenérgicos beta/metabolismo , Adenoviridae/genética , Animais , Células Cultivadas , Quinase 2 de Receptor Acoplado a Proteína G , Quinase 3 de Receptor Acoplado a Proteína G , Genes Reporter , Concentração Inibidora 50 , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Miocárdio/citologia , Miocárdio/enzimologia , Ratos , Ratos Wistar , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta 1/genética , Receptores de Endotelina/metabolismo , Especificidade por Substrato , Transdução Genética , Quinases de Receptores Adrenérgicos beta/análise , Quinases de Receptores Adrenérgicos beta/genética
19.
Am J Physiol Renal Physiol ; 293(3): F877-84, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17567939

RESUMO

The renal dopamine system plays an important role in sodium homeostasis and a defect in dopamine D1 receptor (D1R) function is present in hypertension, diabetes, and aging. Our previous studies in hyperinsulinemic animals and in renal cell cultures treated with insulin showed decrease in D1R number and defective coupling to G proteins; however, the exact mechanisms remained unknown. Therefore, we investigated insulin-mediated D1R desensitization and underlying molecular mechanism in opossum kidney (OK) cells. Chronic exposure (24 h) of OK cells to 10 nM insulin caused significant decrease in D1R number and agonist affinity. The D1R was hyperserine phosphorylated, uncoupled from G proteins and SKF38393, a D1R agonist, failed to stimulate G proteins and inhibit Na-K-ATPase activity. Insulin increased protein kinase C (PKC) activity and caused G protein-coupled receptor kinase 2 (GRK2) translocation to the membranes. Tyrosine kinase inhibitor genistein and phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin blocked insulin-mediated PKC activation and GRK2 membranous translocation. In addition to genistein and wortmannin, GRK2 membranous tranlocation was also blocked by PKC inhibitor chelerythrine chloride and GRK2-specific siRNA. Genistein, wortmannin, chelerythrine chloride, and GRK2 siRNA abrogated D1R serine phosphorylation and normalized D1R expression and affinity in insulin-treated cells. Furthermore, these inhibitors and siRNA restored D1R G protein coupling and ability of SKF38393 to inhibit Na-K-ATPase activity. In conclusion, insulin-induced D1R desensitization involves PI3K, PKC, and GRK2. Insulin activates PI3K-PKC-GRK2 cascade, causing D1R serine phosphorylation, which leads to D1R downregulation and uncoupling from G proteins, and results in the failure of SKF38393 to stimulate G proteins and inhibit Na-K-ATPase activity.


Assuntos
Insulina/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/metabolismo , Receptores de Dopamina D1/metabolismo , Quinases de Receptores Adrenérgicos beta/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica , Rim/citologia , Gambás , Fosforilação , Transporte Proteico
20.
FEBS Lett ; 581(17): 3204-10, 2007 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-17583697

RESUMO

G-protein coupled receptor kinase 3 (GRK3) mediates desensitization of alpha(2)-adrenergic (alpha(2)-AR) and CRF(1) receptors. CRF(1) receptors, alpha(2)-AR and GRK3, are localized to the primary source of noradrenergic inputs to higher brain centers critical in both the response to stress and the development of depression, namely, locus coeruleus (LC). This study utilizing CATH.a cells (derived from the LC), demonstrates for the first time, that the stress hormone, CRF selectively up-regulates GRK3 expression via an ERK1/2-mediated mechanism accompanied by the activation of Sp-1 and Ap-2 transcription factors. This observation has important implications for the regulation of stress signaling in the brain.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Quinases de Receptores Adrenérgicos beta/genética , Animais , Células Cultivadas , Hormônio Liberador da Corticotropina/farmacologia , Quinase 3 de Receptor Acoplado a Proteína G , Regulação da Expressão Gênica/efeitos dos fármacos , Locus Cerúleo/citologia , Locus Cerúleo/metabolismo , RNA Mensageiro/metabolismo , Ratos , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição AP-2/metabolismo , Quinases de Receptores Adrenérgicos beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...