Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 756
Filtrar
1.
Am J Pathol ; 193(11): 1817-1832, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37423551

RESUMEN

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.


Asunto(s)
Anexina A1 , Ratas , Animales , Anexina A1/farmacología , Anexina A1/química , Anexina A1/metabolismo , Células Caliciformes/metabolismo , Receptores de Formil Péptido/metabolismo , Histamina/farmacología , Péptidos/farmacología , Antiinflamatorios/farmacología , Quinasas de Receptores Adrenérgicos beta/metabolismo
2.
Mol Pharmacol ; 98(4): 497-507, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32362586

RESUMEN

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.


Asunto(s)
Locus Coeruleus/metabolismo , Morfina/administración & dosificación , Receptores Opioides mu/metabolismo , Quinasas de Receptores Adrenérgicos beta/metabolismo , Animales , Tolerancia a Medicamentos , Femenino , Locus Coeruleus/efectos de los fármacos , Masculino , Morfina/farmacología , Técnicas de Placa-Clamp , Fosforilación , Potasio/metabolismo , Ratas
3.
Mol Biol Rep ; 47(6): 4631-4650, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32303958

RESUMEN

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.


Asunto(s)
Glioma/metabolismo , Receptores Adrenérgicos beta/metabolismo , beta-Arrestinas/metabolismo , Agonistas Adrenérgicos beta/metabolismo , Animales , Carcinoma/genética , Carcinoma/metabolismo , Proliferación Celular , Glioma/genética , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Transporte de Proteínas , Receptores Adrenérgicos beta 2/genética , Transducción de Señal/efectos de los fármacos , Neoplasias de la Columna Vertebral/genética , Neoplasias de la Columna Vertebral/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Quinasas de Receptores Adrenérgicos beta/metabolismo , beta-Arrestinas/fisiología
4.
J Pharmacol Exp Ther ; 356(2): 503-13, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26604244

RESUMEN

After hindlimb ischemia (HI), increased catecholamine levels within the ischemic muscle can cause dysregulation of ß2-adrenergic receptor (ß2AR) signaling, leading to reduced revascularization. Indeed, in vivo ß2AR overexpression via gene therapy enhances angiogenesis in a rat model of HI. G protein-coupled receptor kinase 2 (GRK2) is a key regulator of ßAR signaling, and ß adrenergic receptor kinase C-terminal peptide (ßARKct), a peptide inhibitor of GRK2, has been shown to prevent ßAR down-regulation and to protect cardiac myocytes and stem cells from ischemic injury through restoration of ß2AR protective signaling (i.e., protein kinase B/endothelial nitric oxide synthase). Herein, we tested the potential therapeutic effects of adenoviral-mediated ßARKct gene transfer in an experimental model of HI and its effects on ßAR signaling and on endothelial cell (EC) function in vitro. Accordingly, in this study, we surgically induced HI in rats by femoral artery resection (FAR). Fifteen days of ischemia resulted in significant ßAR down-regulation that was paralleled by an approximately 2-fold increase in GRK2 levels in the ischemic muscle. Importantly, in vivo gene transfer of the ßARKct in the hindlimb of rats at the time of FAR resulted in a marked improvement of hindlimb perfusion, with increased capillary and ßAR density in the ischemic muscle, compared with control groups. The effect of ßARKct expression was also assessed in vitro in cultured ECs. Interestingly, ECs expressing the ßARKct fenoterol, a ß2AR-agonist, induced enhanced ß2AR proangiogenic signaling and increased EC function. Our results suggest that ßARKct gene therapy and subsequent GRK2 inhibition promotes angiogenesis in a model of HI by preventing ischemia-induced ß2AR down-regulation.


Asunto(s)
Terapia Genética/tendencias , Miembro Posterior/irrigación sanguínea , Isquemia/genética , Neovascularización Patológica/genética , Receptores Adrenérgicos beta 2/genética , Quinasas de Receptores Adrenérgicos beta/genética , Animales , Bovinos , Células Cultivadas , Isquemia/terapia , Masculino , Neovascularización Patológica/terapia , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 2/metabolismo , Quinasas de Receptores Adrenérgicos beta/administración & dosificación
5.
J Cell Mol Med ; 18(11): 2135-46, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25216213

RESUMEN

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.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Electrofisiología , Corazón/fisiopatología , Miocardio/enzimología , Potenciales de Acción , AMP Cíclico/metabolismo , Humanos , Canales Iónicos/metabolismo , Miocardio/metabolismo , Transducción de Señal , Quinasas de Receptores Adrenérgicos beta/metabolismo
6.
Nat Med ; 13(3): 315-23, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17322894

RESUMEN

Cardiac overstimulation by the sympathetic nervous system (SNS) is a salient characteristic of heart failure, reflected by elevated circulating levels of catecholamines. The success of beta-adrenergic receptor (betaAR) antagonists in heart failure argues for SNS hyperactivity being pathogenic; however, sympatholytic agents targeting alpha2AR-mediated catecholamine inhibition have been unsuccessful. By investigating adrenal adrenergic receptor signaling in heart failure models, we found molecular mechanisms to explain the failure of sympatholytic agents and discovered a new strategy to lower SNS activity. During heart failure, there is substantial alpha2AR dysregulation in the adrenal gland, triggered by increased expression and activity of G protein-coupled receptor kinase 2 (GRK2). Adrenal gland-specific GRK2 inhibition reversed alpha2AR dysregulation in heart failure, resulting in lowered plasma catecholamine levels, improved cardiac betaAR signaling and function, and increased sympatholytic efficacy of a alpha2AR agonist. This is the first demonstration, to our knowledge, of a molecular mechanism for SNS hyperactivity in heart failure, and our study identifies adrenal GRK2 activity as a new sympatholytic target.


Asunto(s)
Glándulas Suprarrenales/enzimología , Hiperfunción de las Glándulas Suprarrenales/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Insuficiencia Cardíaca/enzimología , Regulación hacia Arriba/fisiología , Quinasas de Receptores Adrenérgicos beta/biosíntesis , Glándulas Suprarrenales/patología , Glándulas Suprarrenales/fisiopatología , Hiperfunción de las Glándulas Suprarrenales/fisiopatología , Animales , Células Cultivadas , Quinasa 2 del Receptor Acoplado a Proteína-G , Insuficiencia Cardíaca/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Quinasas de Receptores Adrenérgicos beta/antagonistas & inhibidores , Quinasas de Receptores Adrenérgicos beta/fisiología
7.
Nat Cell Biol ; 7(8): 785-96, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16094730

RESUMEN

Phosphoinositide 3-kinase (PI(3)K) is a unique enzyme characterized by both lipid and protein kinase activities. Here, we demonstrate a requirement for the protein kinase activity of PI(3)K in agonist-dependent beta-adrenergic receptor (betaAR) internalization. Using PI(3)K mutants with either protein or lipid phosphorylation activity, we identify the cytoskeletal protein non-muscle tropomyosin as a substrate of PI(3)K, which is phosphorylated in a wortmannin-sensitive manner on residue Ser 61. A constitutively dephosphorylated (S61A) tropomyosin mutant blocks agonist-dependent betaAR internalization, whereas a tropomyosin mutant that mimics constitutive phosphorylation (S61D) complements the PI(3)K mutant, with only lipid phosphorylation activity reversing the defective betaAR internalization. Notably, knocking down endogenous tropomyosin expression using siRNAs that target different regions if tropomyosin resulted in complete inhibition of betaAR endocytosis, showing that non-muscle tropomyosin is essential for agonist-mediated receptor internalization. These studies demonstrate a previously unknown role for the protein phosphorylation activity of PI(3)K in betaAR internalization and identify non-muscle tropomyosin as a cellular substrate for protein kinase activity of PI(3)K.


Asunto(s)
Endocitosis/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas/metabolismo , Receptores Adrenérgicos beta/metabolismo , Actinas/metabolismo , Complejo 2 de Proteína Adaptadora/metabolismo , Androstadienos/farmacología , Arrestinas/metabolismo , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Histonas/metabolismo , Humanos , Modelos Biológicos , Mutación/fisiología , Fosfatidilinositol 3-Quinasas/genética , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/genética , ARN Interferente Pequeño/genética , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Serina/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transfección , Transferrina/metabolismo , Tropomiosina/genética , Tropomiosina/metabolismo , Wortmanina , Quinasas de Receptores Adrenérgicos beta , beta-Arrestinas
8.
J Cell Biol ; 177(1): 127-37, 2007 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-17403928

RESUMEN

We demonstrate a role for protein kinase casein kinase 2 (CK2) in the phosphorylation and regulation of the M3-muscarinic receptor in transfected cells and cerebellar granule neurons. On agonist occupation, specific subsets of receptor phosphoacceptor sites (which include the SASSDEED motif in the third intracellular loop) are phosphorylated by CK2. Receptor phosphorylation mediated by CK2 specifically regulates receptor coupling to the Jun-kinase pathway. Importantly, other phosphorylation-dependent receptor processes are regulated by kinases distinct from CK2. We conclude that G protein-coupled receptors (GPCRs) can be phosphorylated in an agonist-dependent fashion by protein kinases from a diverse range of kinase families, not just the GPCR kinases, and that receptor phosphorylation by a defined kinase determines a specific signalling outcome. Furthermore, we demonstrate that the M3-muscarinic receptor can be differentially phosphorylated in different cell types, indicating that phosphorylation is a flexible regulatory process where the sites that are phosphorylated, and hence the signalling outcome, are dependent on the cell type in which the receptor is expressed.


Asunto(s)
Quinasa de la Caseína II/fisiología , Receptor Muscarínico M3/metabolismo , Secuencias de Aminoácidos , Animales , Células CHO , Quinasa de la Caseína II/antagonistas & inhibidores , Células Cultivadas , Secuencia de Consenso , Cricetinae , Cricetulus , Humanos , Ratones , Datos de Secuencia Molecular , Neuronas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Receptor Muscarínico M3/química , Transducción de Señal , Quinasas de Receptores Adrenérgicos beta/metabolismo
9.
Nat Med ; 11(9): 952-8, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16142243

RESUMEN

Nitric oxide (NO) production by endothelial cell nitric oxide synthase (eNOS) in sinusoidal endothelial cells is reduced in the injured liver and leads to intrahepatic portal hypertension. We sought to understand the mechanism underlying defective eNOS function. Phosphorylation of the serine-threonine kinase Akt, which activates eNOS, was substantially reduced in sinusoidal endothelial cells from injured livers. Overexpression of Akt in vivo restored phosphorylation of Akt and production of NO and reduced portal pressure in portal hypertensive rats. We found that Akt physically interacts with G-protein-coupled receptor kinase-2 (GRK2), and that this interaction inhibits Akt activity. Furthermore, GRK2 expression increased in sinusoidal endothelial cells from portal hypertensive rats and knockdown of GRK2 restored Akt phosphorylation and NO production, and normalized portal pressure. Finally, after liver injury, GRK2-deficient mice developed less severe portal hypertension than control mice. Thus, an important mechanism underlying impaired activity of eNOS in injured sinusoidal endothelial cells is defective phosphorylation of Akt caused by overexpression of GRK2 after injury.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Células Endoteliales/enzimología , Hipertensión Portal/metabolismo , Óxido Nítrico Sintasa/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Células Cultivadas , Quinasa 2 del Receptor Acoplado a Proteína-G , Isoenzimas , Masculino , Óxido Nítrico , Óxido Nítrico Sintasa de Tipo III , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Sprague-Dawley , Quinasas de Receptores Adrenérgicos beta
10.
J Mol Cell Cardiol ; 51(4): 462-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21256851

RESUMEN

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."


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Terapia Molecular Dirigida , Antagonistas Adrenérgicos beta/uso terapéutico , Animales , Fármacos Cardiovasculares/uso terapéutico , Evaluación Preclínica de Medicamentos , Subunidades beta de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades gamma de la Proteína de Unión al GTP/antagonistas & inhibidores , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal/efectos de los fármacos , Quinasas de Receptores Adrenérgicos beta/antagonistas & inhibidores , Quinasas de Receptores Adrenérgicos beta/metabolismo
11.
J Cell Mol Med ; 15(2): 258-69, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20015194

RESUMEN

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.


Asunto(s)
Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/biosíntesis , Receptor de Bradiquinina B1/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Línea Celular , Citocinas/farmacología , Enalaprilato/farmacología , Células Endoteliales/metabolismo , Activación Enzimática , Flavonoides/farmacología , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Pirimidinas/farmacología , Receptor de Bradiquinina B1/agonistas , Receptor de Bradiquinina B1/genética , Transfección , Quinasas de Receptores Adrenérgicos beta/metabolismo , Familia-src Quinasas/antagonistas & inhibidores
12.
J Biol Chem ; 285(53): 41290-9, 2010 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-21044958

RESUMEN

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.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/química , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Anestésicos , Animales , Sitios de Unión , Línea Celular , Halotano/química , Hipocampo/metabolismo , Humanos , Neurotransmisores/química , Oocitos/metabolismo , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína , Xenopus , Quinasas de Receptores Adrenérgicos beta/metabolismo
13.
J Cell Physiol ; 226(1): 181-6, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20677219

RESUMEN

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.


Asunto(s)
Fístula Arteriovenosa/complicaciones , Fístula Arteriovenosa/metabolismo , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Receptores Adrenérgicos beta/metabolismo , Caracteres Sexuales , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Animales , Arrestinas/genética , Arrestinas/metabolismo , Gasto Cardíaco , Cardiomegalia/fisiopatología , Catecolaminas/sangre , Ecocardiografía , Femenino , Regulación de la Expresión Génica/fisiología , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta/genética , Remodelación Ventricular/fisiología , Quinasas de Receptores Adrenérgicos beta/genética , Quinasas de Receptores Adrenérgicos beta/metabolismo , Arrestina beta 2 , beta-Arrestinas
14.
BMC Cancer ; 11: 256, 2011 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-21679469

RESUMEN

BACKGROUND: Recently we reported that activation of Epac1, an exchange protein activated by cAMP, increases melanoma cell migration via Ca 2+ release from the endoplasmic reticulum (ER). G-protein ßγ subunits (Gßγ) are known to act as an independent signaling molecule upon activation of G-protein coupled receptor. However, the role of Gßγ in cell migration and Ca 2+ signaling in melanoma has not been well studied. Here we report that there is crosstalk of Ca 2+ signaling between Gßγ and Epac in melanoma, which plays a role in regulation of cell migration. METHODS: SK-Mel-2 cells, a human metastatic melanoma cell line, were mainly used in this study. Intracellular Ca 2+ was measured with Fluo-4AM fluorescent dyes. Cell migration was examined using the Boyden chambers. RESULTS: The effect of Gßγ on Epac-induced cell migration was first examined. Epac-induced cell migration was inhibited by mSIRK, a Gßγ -activating peptide, but not its inactive analog, L9A, in SK-Mel-2 cells. Guanosine 5', α-ß-methylene triphosphate (Gp(CH2)pp), a constitutively active GTP analogue that activates Gßγ, also inhibited Epac-induced cell migration. In addition, co-overexpression of ß1 and γ2, which is the major combination of Gßγ, inhibited Epac1-induced cell migration. By contrast, when the C-terminus of ß adrenergic receptor kinase (ßARK-CT), an endogenous inhibitor for Gßγ, was overexpressed, mSIRK's inhibitory effect on Epac-induced cell migration was negated, suggesting the specificity of mSIRK for Gßγ. We next examined the effect of mSIRK on Epac-induced Ca 2+ response. When cells were pretreated with mSIRK, but not with L9A, 8-(4-Methoxyphenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate (8-pMeOPT), an Epac-specific agonist, failed to increase Ca 2+ signal. Co-overexpression of ß1 and γ2 subunits inhibited 8-pMeOPT-induced Ca 2+ elevation. Inhibition of Gßγ with ßARK-CT or guanosine 5'-O-(2-thiodiphosphate) (GDPßS), a GDP analogue that inactivates Gßγ, restored 8-pMeOPT-induced Ca 2+ elevation even in the presence of mSIRK. These data suggested that Gßγ inhibits Epac-induced Ca 2+ elevation. Subsequently, the mechanism by which Gßγ inhibits Epac-induced Ca 2+ elevation was explored. mSIRK activates Ca 2+ influx from the extracellular space. In addition, W-5, an inhibitor of calmodulin, abolished mSIRK's inhibitory effects on Epac-induced Ca 2+ elevation, and cell migration. These data suggest that, the mSIRK-induced Ca 2+ from the extracellular space inhibits the Epac-induced Ca 2+ release from the ER, resulting suppression of cell migration. CONCLUSION: We found the cross talk of Ca 2+ signaling between Gßγ and Epac, which plays a major role in melanoma cell migration.


Asunto(s)
Señalización del Calcio/fisiología , Subunidades beta de la Proteína de Unión al GTP/fisiología , Proteínas de Unión al GTP/fisiología , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Melanoma/patología , Proteínas de Neoplasias/fisiología , Secuencia de Aminoácidos , Bloqueadores de los Canales de Calcio/farmacología , Calmodulina/fisiología , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Subunidades beta de la Proteína de Unión al GTP/genética , Proteínas de Unión al GTP/genética , Factores de Intercambio de Guanina Nucleótido/fisiología , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/farmacología , Humanos , Melanoma/secundario , Datos de Secuencia Molecular , Proteínas de Neoplasias/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Proteínas Recombinantes de Fusión/fisiología , Proteínas Recombinantes/farmacología , Tionucleótidos/farmacología , Quinasas de Receptores Adrenérgicos beta/antagonistas & inhibidores
15.
Nat Med ; 9(3): 315-21, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12592402

RESUMEN

Polymorphonuclear leukocytes (PMNs) are critical effector cells of the innate immune system that protect the host by migrating to inflammatory sites and killing pathogenic microbes. We addressed the role of chemokine receptor desensitization induced by G-protein-coupled receptor kinases (GRKs) in the feedback control of PMN migration. We show that the chemokine macrophage inflammatory protein-2 (MIP-2) induces GRK2 and GRK5 expression in PMNs through phosphoinositide-3-kinase (PI3K)-gamma signaling. We also show that lipopolysaccharide (LPS)-activated signaling through the Toll-like receptor (TLR)-4 pathway transcriptionally downregulates the expression of GRK2 and GRK5 in response to MIP-2. The reduced expression of GRKs lowers chemokine receptor desensitization and markedly augments the PMN migratory response. These data indicate that TLR4 modulation of PMN surface chemokine receptor expression subsequent to the downregulation of GRK2 and GRK5 expression is a critical determinant of PMN migration.


Asunto(s)
Movimiento Celular/fisiología , Proteínas de Drosophila , Glicoproteínas de Membrana/metabolismo , Neutrófilos/fisiología , Receptores de Superficie Celular/metabolismo , Receptores de Quimiocina/metabolismo , Transducción de Señal/fisiología , Animales , Quimiocina CXCL2 , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo/fisiología , Activación Enzimática , Inhibidores Enzimáticos/metabolismo , Quinasa 5 del Receptor Acoplado a Proteína-G , Humanos , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Monocinas/metabolismo , Neutrófilos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Toll-Like 4 , Receptores Toll-Like , Quinasas de Receptores Adrenérgicos beta
16.
Neuron ; 109(14): 2256-2274.e9, 2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34139149

RESUMEN

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.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Quinasas de Receptores Adrenérgicos beta/metabolismo , Animales , Calcio/metabolismo , Ratones , Neuronas/metabolismo
17.
J Exp Med ; 187(5): 801-6, 1998 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-9480990

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV)/human herpesvirus 8, which is consistently present in tissues of patients with Kaposi's sarcoma and primary effusion lymphomas, contains a gene that encodes a G protein-coupled receptor (KSHV-GPCR). We recently showed that KSHV-GPCR exhibits constitutive signaling via activation of phosphoinositide-specific phospholipase C and stimulates cell proliferation and transformation. In this study, we determined whether normal cellular mechanisms could inhibit constitutive signaling by KSHV-GPCR and thereby KSHV-GPCR-stimulated proliferation. We show that coexpression of GPCR-specific kinases (GRKs) and activation of protein kinase C inhibit constitutive signaling by KSHV-GPCR in COS-1 monkey kidney cells and in mouse NIH 3T3 cells. Moreover, GRK-5 but not GRK-2 inhibits KSHV-GPCR-stimulated proliferation of rodent fibroblasts. These data provide evidence that cell regulatory pathways of receptor desensitization may be therapeutic targets in human diseases involving constitutively active receptors.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Herpesvirus Humano 8/patogenicidad , Proteína Quinasa C/fisiología , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Quimiocina/fisiología , Sarcoma de Kaposi/virología , Proteínas Virales/fisiología , Células 3T3 , Animales , Células COS , División Celular , Células Cultivadas , Quinasa 5 del Receptor Acoplado a Proteína-G , Herpesvirus Humano 8/genética , Fosfatos de Inositol/metabolismo , Ratones , Transducción de Señal , Transfección , Quinasas de Receptores Adrenérgicos beta
18.
Nature ; 426(6966): 574-9, 2003 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-14654844

RESUMEN

Feedback inhibition is a fundamental principle in signal transduction allowing rapid adaptation to different stimuli. In mammalian cells, the major feedback inhibitor for G-protein-coupled receptors (GPCR) is G-protein-coupled receptor kinase 2 (GRK-2), which phosphorylates activated receptors, uncouples them from G proteins and initiates their internalization. The functions of GRK-2 are indispensable and need to be tightly controlled. Dysregulation promotes disorders such as hypertension or heart failure. In our search for a control mechanism for this vital kinase, here we show that the Raf kinase inhibitor protein (RKIP) is a physiological inhibitor of GRK-2. After stimulation of GPCR, RKIP dissociates from its known target, Raf-1 (refs 6-8), to associate with GRK-2 and block its activity. This switch is triggered by protein kinase C (PKC)-dependent phosphorylation of the RKIP on serine 153. The data delineate a new principle in signal transduction: by activating PKC, the incoming receptor signal is enhanced both by removing an inhibitor from Raf-1 and by blocking receptor internalization. A physiological role for this mechanism is shown in cardiomyocytes in which the downregulation of RKIP restrains beta-adrenergic signalling and contractile activity.


Asunto(s)
Proteína de Unión a Andrógenos , Proteínas Portadoras/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Inhibidores Enzimáticos/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Animales , Encéfalo/metabolismo , Proteínas Portadoras/genética , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G , Quinasa 3 del Receptor Acoplado a Proteína-G , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Proteínas de Unión a Fosfatidiletanolamina , Proteínas de Transferencia de Fosfolípidos , Fosforilación , Pruebas de Precipitina , Prostateína , Unión Proteica , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Interferencia de ARN , Ratas , Secretoglobinas , Transducción de Señal , Especificidad por Sustrato , Uteroglobina , Quinasas de Receptores Adrenérgicos beta
19.
Curr Biol ; 16(20): 2042-7, 2006 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-17055984

RESUMEN

p38 Mitogen-activated protein kinases (MAPK) are a family of Ser/Thr kinases that regulate important cellular processes such as stress responses, differentiation, and cell-cycle control . Activation of MAPK is achieved through a linear signaling cascade in which upstream kinases (MAPKKs) dually phosphorylate MAPKs at a conserved 3-amino-acid motif (Thr-X-Tyr) . G-protein-coupled receptor kinases (GRKs) are known to selectively phosphorylate G-protein-coupled receptors (GPCRs) and thus trigger desensitization . We report that GRK2 is a novel inactivating kinase of p38MAPK. p38 associates with GRK2 endogenously and is phosphorylated by GRK2 at Thr-123, a residue located at its docking groove. Mimicking phosphorylation at this site impairs the binding and activation of p38 by MKK6 and diminishes the capacity of p38 to bind and phosphorylate its substrates. Accordingly, p38 activation is decreased or increased when cellular GRK2 levels are enhanced or reduced, respectively. Changes in GRK2 levels and activity can modify p38-dependent processes such as differentiation of preadipocytic cells and LPS-induced cytokine release, enhanced in macrophages from GRK2(+/-) mice. Phosphorylation of p38 at a region key for its interaction with different partners uncovers a new mechanism for the regulation of this important family of kinases.


Asunto(s)
Regulación hacia Abajo , Quinasas de Receptores Adrenérgicos beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Western Blotting , Línea Celular , Cartilla de ADN , Electroforesis en Gel Bidimensional , Activación Enzimática/fisiología , Quinasa 2 del Receptor Acoplado a Proteína-G , Humanos , Inmunoprecipitación , Macrófagos/metabolismo , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Fosforilación , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
20.
J Clin Invest ; 116(6): 1547-60, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16741575

RESUMEN

For over a century, there has been intense debate as to the reason why some cardiac stresses are pathological and others are physiological. One long-standing theory is that physiological overloads such as exercise are intermittent, while pathological overloads such as hypertension are chronic. In this study, we hypothesized that the nature of the stress on the heart, rather than its duration, is the key determinant of the maladaptive phenotype. To test this, we applied intermittent pressure overload on the hearts of mice and tested the roles of duration and nature of the stress on the development of cardiac failure. Despite a mild hypertrophic response, preserved systolic function, and a favorable fetal gene expression profile, hearts exposed to intermittent pressure overload displayed pathological features. Importantly, intermittent pressure overload caused diastolic dysfunction, altered beta-adrenergic receptor (betaAR) function, and vascular rarefaction before the development of cardiac hypertrophy, which were largely normalized by preventing the recruitment of PI3K by betaAR kinase 1 to ligand-activated receptors. Thus stress-induced activation of pathogenic signaling pathways, not the duration of stress or the hypertrophic growth per se, is the molecular trigger of cardiac dysfunction.


Asunto(s)
Presión Sanguínea/fisiología , Vasos Sanguíneos , Cardiomegalia , Corazón/fisiología , Miocardio/patología , Estrés Fisiológico , Antagonistas Adrenérgicos beta/metabolismo , Animales , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiología , Vasos Sanguíneos/fisiopatología , Gasto Cardíaco Bajo , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Células Cultivadas , Ecocardiografía , Femenino , Regulación de la Expresión Génica , Hemodinámica , Humanos , Hipertrofia Ventricular Izquierda , Metoprolol/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/citología , Miocardio/metabolismo , Fenotipo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal/fisiología , Quinasas de Receptores Adrenérgicos beta/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda