Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncol Rep ; 51(2)2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38099418

RESUMO

C­X­C motif chemokine 12 (CXCL12) promotes metastasis of several tumors by affecting cell migration and invasion via its receptors, C­X­C chemokine receptor type (CXCR)4 and CXCR7. Current therapeutic approaches focus on the selective inactivation of either CXCR4 or CXCR7 in patients with cancer. Alternative strategies may emerge from the analysis of downstream events that mediate the migratory effects of CXCL12 in cancer cells. While CXCR4 activates cell signaling through both G proteins and arrestins, CXCR7 is believed to preferentially signal through arrestins. The present study analyzed the CXCL12­dependent chemotaxis of A549, C33A, DLD­1, MDA­MB­231 and PC­3 cells, in which either the activity of G proteins, EGFR or Src kinase was inhibited pharmacologically or the expression of arrestins was inhibited by RNA interference. The results demonstrated that CXCL12­induced migration of A549, C33A, DLD­1, MDA­MB­231 and PC­3 cells was attenuated by the Gαi/o­inhibitor pertussis toxin (PTX), but was unaffected by small interfering RNA­mediated gene silencing of ß­arrestin1/2. In particular, the sensitivity of DLD­1 migration to PTX was unexpected, as it is solely dependent on the non­classical chemokine receptor, CXCR7. Furthermore, chemotactic responses to CXCL12 were additionally prevented by inhibiting EGFR activity via AG1478 and Src kinase activity via Src inhibitor­1. In conclusion, the results of the present study suggest that G protein­ and Src­dependent transactivation of EGFR is a common mechanism through which CXCL12­bound CXCR4 and/or CXCR7 control cancer cell migration and metastasis. These findings highlight EGFR as a potential therapeutic target that interferes with CXCL12­induced cancer expansion.


Assuntos
Neoplasias , Receptores CXCR , Humanos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Ativação Transcricional , Receptores CXCR/genética , Receptores CXCR/metabolismo , Transdução de Sinais , Proteínas de Ligação ao GTP , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Movimento Celular , Arrestinas/genética , Arrestinas/metabolismo , Arrestinas/farmacologia , Quinases da Família src/genética , Quinases da Família src/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo
2.
Eur J Pharmacol ; 956: 175952, 2023 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-37541367

RESUMO

BACKGROUND: Beta arrestins had been known as intracellular adaptors that uncouple and inactivate the G protein-coupled receptors that they interact with. Their roles as signal initiators for some receptors have recently been recognized. SCOPE OF REVIEW: In this review, we focused on their role in mediating metabolic modulation primarily in relation to insulin signaling. Commenced by the upstream receptor, they seem to act like intracellular hubs that divert the metabolic profile of the cell. The amount of metabolic substrates in circulation and their usage/deposition by tissues are controlled by the contribution of all systems in the organism. This control is enabled by the release of hormones such as insulin, glucagon and glucagon-like peptide-1. Intriguingly, some ligands -either agonists or antagonists-of different classes of receptors have preferential properties mediated by ß arrestins. This is not surprizing considering that substrate supply and usage should parallel physiological function such as hormone release or muscle contraction. MAJOR CONCLUSIONS: Available data indicate that ß arrestins conduct the regulatory role in insulin secretion and action. They may be good candidates to target when the upstream signal demands the function that may compromise the cell. An example is carvedilol that is protective by preventing the stimulatory effects of excessive catecholamines, stimulates mitochondrial function and has preferential clinical outcomes in metabolic disorders.


Assuntos
Arrestinas , Transdução de Sinais , beta-Arrestinas/metabolismo , Secreção de Insulina , Arrestinas/metabolismo , Arrestinas/farmacologia , Insulina/metabolismo
3.
Methods ; 92: 64-71, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26320830

RESUMO

The realization that G-protein coupled receptors (GPCR) engage several cell signaling mechanisms simultaneously has led to a multiplication of research aimed at developing biased ligands exerting a selective action on subsets of responses downstream of a given receptor. Several tools have been developed to identify such ligands using recombinant cell systems. However the validation of biased ligand activity in animal models remains a serious challenge. Here we present a general strategy that can be used to validate biased ligand activity in vivo and supports it as a strategy for further drug development. In doing so, we placed special attention on strategies allowing to discriminate between G-protein and beta-arrestin mediated mechanisms. We also underscore differences between in vitro and in vivo systems and suggest avenues for tool development to streamline the translation of biased ligands development to pre-clinical animal models.


Assuntos
Arrestinas/metabolismo , Modelos Animais , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Animais , Arrestinas/farmacologia , Haloperidol/metabolismo , Haloperidol/farmacologia , Humanos , Ligantes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica/fisiologia , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , beta-Arrestinas
4.
Methods ; 92: 87-93, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26297537

RESUMO

Many GPCRs are able to activate multiple distinct signaling pathways, and these may include biochemical cascades activated via either heterotrimeric G proteins or by ß-arrestins. The relative potencies and/or efficacies among a series of agonists that act on a common receptor can vary depending upon which signaling pathway is being activated. This phenomenon is known as biased signaling or functional selectivity, and is presumed to reflect underlying differences in ligand binding affinities for alternate conformational states of the receptor. The first part of this review discusses how various cellular GPCR interacting proteins (GIPs) can influence receptor conformation and thereby affect ligand-receptor interactions and contribute to signaling bias. Upon activation, receptors trigger biochemical cascades that lead to altered cellular function, and measuring points along the cascade (e.g., second messenger production) conveys information about receptor activity. As a signal continues along its way, the observed concentration dependence of a GPCR ligand may change due to amplification and saturation of biochemical steps. The second part of this review considers additional cellular factors that affect signal processing, focusing mainly on structural elements and deamplification mechanisms, and discusses the relevance of these to measurements of potency and functional selectivity.


Assuntos
Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Animais , Arrestinas/metabolismo , Arrestinas/farmacologia , Humanos , Ligação Proteica/fisiologia , Transdução de Sinais/efeitos dos fármacos , beta-Arrestinas
5.
Mol Pharmacol ; 88(4): 816-24, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25934731

RESUMO

Sustained activation of G protein-coupled receptors can lead to a rapid decline in signaling through acute receptor desensitization. In the case of the µ-opioid receptor (MOPr), this desensitization may play a role in the development of analgesic tolerance. It is understood that phosphorylation of MOPr promotes association with ß-arrestin proteins, which then facilitates desensitization and receptor internalization. Agonists that induce acute desensitization have been shown to induce a noncanonical high-affinity agonist binding state in MOPr, conferring a persistent memory of prior receptor activation. In the current study, live-cell confocal imaging was used to investigate the role of receptor phosphorylation in agonist binding to MOPr. A phosphorylation cluster in the C-terminal tail of MOPr was identified as a mediator of agonist-induced affinity changes in MOPr. This site is unique from the primary phosphorylation cluster responsible for ß-arrestin binding and internalization. Electrophysiologic measurements of receptor function suggest that both phosphorylation clusters may play a parallel role during acute receptor desensitization. Desensitization was unaffected by alanine mutation of either phosphorylation cluster, but was largely eliminated when both clusters were mutated. Overall, this work suggests that there are multiple effects of MOPr phosphorylation that appear to regulate MOPr function: one affecting ß-arrestin binding and a second affecting agonist binding.


Assuntos
Analgésicos Opioides/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Analgésicos Opioides/química , Analgésicos Opioides/farmacologia , Animais , Arrestinas/química , Arrestinas/metabolismo , Arrestinas/farmacologia , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , beta-Arrestinas
6.
Curr Protoc Pharmacol ; 68: 2.12.1-2.12.26, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25737158

RESUMO

Only one out of four mammalian arrestin subtypes, arrestin-3, facilitates the activation of JNK family kinases. Here we describe two different protocols used for elucidating the mechanisms involved. One is based on reconstitution of signaling modules from purified proteins: arrestin-3, MKK4, MKK7, JNK1, JNK2, and JNK3. The main advantage of this method is that it unambiguously establishes which effects are direct because only intended purified proteins are present in these assays. The key drawback is that the upstream-most kinases of these cascades, ASK1 or other MAPKKKs, are not available in purified form, limiting reconstitution to incomplete two-kinase modules. The other approach is used for analyzing the effects of arrestin-3 on JNK activation in intact cells. In this case, signaling modules include ASK1 and/or other MAPKKKs. However, as every cell expresses thousands of different proteins their possible effects on the readout cannot be excluded. Nonetheless, the combination of in vitro reconstitution from purified proteins and cell-based assays makes it possible to elucidate the mechanisms of arrestin-3-dependent activation of JNK family kinases.


Assuntos
Arrestinas/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 7/isolamento & purificação , MAP Quinase Quinase Quinase 4/isolamento & purificação , Células Cultivadas , Ativação Enzimática , Humanos , Técnicas In Vitro , Fosforilação , Ligação Proteica , Transfecção
7.
Regul Pept ; 185: 29-33, 2013 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-23816471

RESUMO

Colonic dysmotility occurs in diabetes and the patients exhibit diarrhea or constipation. The pathogenetic mechanisms underlying colonic dysmotility in diabetic patients remain poorly understood. The effects of ß-arrestin2 on colonic contraction in diabetic rats were investigated for the first time. Male SD rats were treated with a single intraperitoneally injected dose of streptozotocin, and those displaying sustained high blood glucose were selected as diabetes mellitus models. Longitudinal muscle strips of the distal colon were prepared to monitor contraction of the colon in vitro. Expression of ß-arrestin2 was investigated by Western blot analysis. Anti-ß-arrestin2 antibody had no direct effect on the contraction of distal colonic strips in both normal and diabetic rats. Carbachol-induced contractions of distal colonic strips were higher in diabetic rats than in normal rats. Anti-ß-arrestin2 antibody partly blocked carbachol-induced increases of distal colonic strips in diabetic rats. The expression level of ß-arrestin2 protein in the colon was higher in diabetic rats than in normal rats. These results suggest that ß-arrestin2 is involved in the increase of distal colonic contraction in diabetic rats.


Assuntos
Arrestinas/fisiologia , Colo/fisiopatologia , Diabetes Mellitus Experimental/fisiopatologia , Músculo Liso/fisiopatologia , Animais , Arrestinas/farmacologia , Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Colo/efeitos dos fármacos , Colo/metabolismo , Diabetes Mellitus Experimental/metabolismo , Técnicas In Vitro , Masculino , Contração Muscular , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Ratos , Ratos Sprague-Dawley , beta-Arrestinas
8.
Mol Cell Endocrinol ; 364(1-2): 71-82, 2012 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-22954680

RESUMO

Activating mutations in the follicle-stimulating hormone (FSH) receptor (FSHR) gene are rarely detected due to the absence of a clearly defined phenotype, particularly in men. We here report the biochemical features of a novel mutation in the first extracellular loop of the FSHR. The mutation (N431I) was detected in an asymptomatic man exhibiting normal spermatogenesis, suppressed serum FSH, and normal or elevated levels of biochemical markers of FSH action. Employing different experimental strategies on HEK-293 cells transiently expressing the N431I FSHR mutant, we found that the mutation led to decreased cell surface plasma membrane expression of the receptor protein, but conferred a low level of constitutive activity associated with markedly altered agonist-stimulated desensitization and internalization. These latter features may contribute and/or amplify the persistent activation of the receptor in both absence and presence of agonist and provide new insights into opportunities for adjuvant therapies based on disruption of these processes.


Assuntos
Arrestinas/farmacologia , Receptores do FSH/genética , Espermatogênese/efeitos dos fármacos , Testículo/metabolismo , Sequência de Bases , AMP Cíclico/metabolismo , Hormônio Foliculoestimulante/metabolismo , Hormônio Foliculoestimulante/farmacologia , Genes Reporter , Células HEK293 , Humanos , Luciferases , Masculino , Dados de Sequência Molecular , Mutação , Fenótipo , Receptores do FSH/metabolismo , Espermatogênese/genética , Testículo/efeitos dos fármacos , Testículo/patologia , Transfecção , Adulto Jovem
9.
Cell Physiol Biochem ; 30(3): 642-52, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22854413

RESUMO

Angiotensin II (AngII) is a key peptide in cardiovascular homeostasis and is a ligand for the Angiotensin II type 1 and 2 seven transmembrane receptors (AT(1)R and AT(2)R). The AT(1) receptor is a seven-transmembrane (7TM) G protein-coupled receptor (GPCR) mediating the majority of the physiological functions of AngII. The AT(1)R mediates its effects through both G protein-dependent and independent signaling, which can be separated by functionally selective agonists. In the present study we investigate the effect of AngII and the ß-arrestin biased agonist [SII]AngII on ischemia-reperfusion injury in rat hearts. Isolated hearts mounted in a Langendorff perfused rat heart preparations showed that preconditioning with [SII]AngII reduced the infarct size induced by global ischemia from 46±8.4% to 22±3.4%. In contrast, neither preconditioning with AngII nor postconditioning with AngII or [SII]AngII had a protective effect. Together these results demonstrate a cardioprotective effect of simultaneous blockade of G protein signaling and activation of G protein independent signaling through AT(1) receptors.


Assuntos
Receptor Tipo 1 de Angiotensina/metabolismo , Traumatismo por Reperfusão/metabolismo , Angiotensina II/farmacologia , Animais , Arrestinas/farmacologia , Cardiotônicos/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Técnicas In Vitro , Masculino , Pressão , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/fisiopatologia , beta-Arrestinas
10.
Br J Anaesth ; 107(5): 774-81, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21926413

RESUMO

BACKGROUND: Tolerance to the analgesic effect of opioids complicates the management of persistent pain states. We tested whether the intrathecal infusion of small interfering RNA (siRNA) against ß-arrestin 2 would reduce tolerance to chronic morphine use and the severity of precipitated morphine withdrawal. METHODS: Intrathecal ß-arrestin 2 (2 µg siRNA per 10 µl per rat) was injected once daily for 3 days. Rats then received a continuous intrathecal infusion of morphine (2 nmol h⁻¹) or saline for 7 days. Daily tail-flick (TF) and intrathecal morphine challenge tests were performed to assess the effect of intrathecal ß-arrestin 2 siRNA on antinociception and tolerance to morphine. Naloxone withdrawal (2 mg kg⁻¹) was performed to assess morphine dependence. RESULTS: In the daily TF test, the antinociception of intrathecal morphine was increased and maintained in rats receiving ß-arrestin 2 siRNA compared with the control group (morphine alone). In the probe response test, rats receiving morphine infusion with ß-arrestin 2 siRNA treatment showed a significant left shift in their dose-response curve, as measured by per cent maximal possible effect (MPE), such that the AD50 was significantly decreased by a factor of 5.6 when compared with that of morphine-infused rats. In the naloxone-induced withdrawal tests, rats receiving ß-arrestin 2 siRNA injection with morphine infusion showed a significant reduction in four of the six signs of withdrawal. CONCLUSIONS: We show here that intrathecal ß-arrestin 2 siRNA in rats enhances analgesia and attenuates naloxone-induced withdrawal symptoms. This may warrant further investigation in the context of long-term use of intrathecal opioids for controlling chronic pain.


Assuntos
Analgésicos Opioides/farmacologia , Arrestinas/farmacologia , Morfina/farmacologia , RNA Interferente Pequeno/farmacologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Tolerância a Medicamentos , Injeções Espinhais , Masculino , Dependência de Morfina/fisiopatologia , Naloxona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Dor/tratamento farmacológico , Medição da Dor , Ratos , Ratos Sprague-Dawley , Cloreto de Sódio/administração & dosagem , Síndrome de Abstinência a Substâncias/fisiopatologia , beta-Arrestina 2 , beta-Arrestinas
11.
Cell Signal ; 23(11): 1767-76, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21740964

RESUMO

The cardiovascular hormone angiotensin II (AngII) exerts its actions via two G protein-coupled receptor (GPCR) subtypes, AT(1) and AT(2), which often display antagonistic functions. Methodological constraints have so far precluded detailed analyses of the ligand-dependency, cellular localization, and functional relevance of AngII receptor interactions in live cells. In this study, we utilize a protein-fragment complementation assay (PCA) and GPCR-Heteromer Identification Technology (GPCR-HIT) to provide the first detailed investigation of the ligand-dependency and cellular localization of AngII receptor interactions in human embryonic kidney 293 cells. Fluorescent-tagged receptor constructs for PCA and GPCR-HIT displayed normal affinity and selectivity for AngII (AT(1): IC(50)=1.0-1.6nM; AT(2): IC(50)=2.0-3.0nM). Well-characterized angiotensin receptor interactions were used as positive and negative controls to demonstrate the sensitivity and specificity of these fluorescence-based assays. We report that AT(1)-AT(2) receptor heteromers form constitutively, are localized to the plasma membrane and perinuclear compartments, and do not internalize following AngII stimulation despite arrestin being recruited specifically to the heteromer. Our findings using novel fluorescence-based technologies reveal a previously unrecognized mechanism of angiotensin receptor cross-talk involving cross-inhibition of AT(1) receptor internalization through heteromerization with the AT(2) receptor subtype.


Assuntos
Angiotensina II/metabolismo , Arrestinas/metabolismo , Bioensaio , Fragmentos de Peptídeos/análise , Receptor Cross-Talk/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Sistema Renina-Angiotensina/fisiologia , Angiotensina II/farmacologia , Arrestinas/farmacologia , Membrana Celular/metabolismo , Movimento Celular , Fluorescência , Células HEK293 , Humanos , Ligantes , Microscopia Confocal , Fragmentos de Peptídeos/química , Fosforilação , Plasmídeos , Polimerização , Ligação Proteica , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/genética , Sensibilidade e Especificidade , Transfecção
13.
Artigo em Inglês | MEDLINE | ID: mdl-21476967

RESUMO

Parathyroid hormone (PTH) is a principle regulator of bone and calcium metabolism and PTH analogs hold great promise as a therapy for metabolic bone diseases such as osteoporosis. PTH acts principally through the type IPTH/PTH-related peptide receptor (PTH1R), a G protein coupled receptor (GPCR). GPCRs are a family of seven transmembrane cell surface receptors that share conserved structural, functional, and regulatory properties. Recent studies demonstrate that the complex metabolic effects induced by PTH1R stimulation are not entirely a consequence of conventional GPCR signaling. ß-arrestins, in addition to their GPCR desensitizing actions, also serve as multifunctional scaffolding proteins linking the PTH1R to signaling molecules independent of the classic G protein coupled second messenger-dependent pathways. In vitro, D-Trp(12),Tyr(34)-bPTH(7-34) (PTH-ßarr), a ß-arrestin selective biased agonist for the PTH1R, antagonizes receptor-G protein coupling but activates arrestin-dependent signaling. In vivo, intermittent administration of, PTH-ßarr to mice, induces anabolic bone formation, completely independent of classic G protein-coupled signaling mechanisms. While both PTH-ßarr and the conventional agonist PTH(1-34) stimulate anabolic bone formation in mice, unlike PTH(1-34), which activates G protein coupling, PTH-ßarr does not induce hypercalcemia or increase markers of bone resorption. This newly recognized ability of ß-arrestins to serve as signal transducers for the PTH1R represents an innovative paradigm of receptor signaling which can be targeted to induce a subset of physiologic responses in bone. Exploitation of ß-arrestin biased agonism may offer therapeutic benefit for the treatment of metabolic bone diseases such as osteoporosis.


Assuntos
Arrestinas/farmacologia , Reabsorção Óssea/metabolismo , Osteogênese/efeitos dos fármacos , Receptor Tipo 1 de Hormônio Paratireóideo/agonistas , Animais , Arrestinas/metabolismo , Reabsorção Óssea/etiologia , Agonismo Parcial de Drogas , Humanos , Camundongos , Modelos Biológicos , Receptor Tipo 1 de Hormônio Paratireóideo/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , beta-Arrestinas
14.
J Biol Chem ; 286(8): 6707-19, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21156802

RESUMO

Given the importance of G-protein-coupled receptors as pharmacological targets in medicine, efforts directed at understanding the molecular mechanism by which pharmacological compounds regulate their presence at the cell surface is of paramount importance. In this context, using confocal microscopy and bioluminescence resonance energy transfer, we have investigated internalization and intracellular trafficking of the cholecystokinin-2 receptor (CCK2R) in response to both natural and synthetic ligands with different pharmacological features. We found that CCK and gastrin, which are full agonists on CCK2R-induced inositol phosphate production, rapidly and abundantly stimulate internalization. Internalized CCK2R did not rapidly recycle to plasma membrane but instead was directed to late endosomes/lysosomes. CCK2R endocytosis involves clathrin-coated pits and dynamin and high affinity and prolonged binding of ß-arrestin1 or -2. Partial agonists and antagonists on CCK2R-induced inositol phosphate formation and ERK1/2 phosphorylation did not stimulate CCK2R internalization or ß-arrestin recruitment to the CCK2R but blocked full agonist-induced internalization and ß-arrestin recruitment. The extreme C-terminal region of the CCK2R (and more precisely phosphorylatable residues Ser(437)-Xaa(438)-Thr(439)-Thr(440)-Xaa(441)-Ser(442)-Thr(443)) were critical for ß-arrestin recruitment. However, this region and ß-arrestins were dispensable for CCK2R internalization. In conclusion, this study allowed us to classify the human CCK2R as a member of class B G-protein-coupled receptors with regard to its endocytosis features and identified biased agonists of the CCK2R. These new important insights will allow us to investigate the role of internalized CCK2R·ß-arrestin complexes in cancers expressing this receptor and to develop new diagnosis and therapeutic strategies targeting this receptor.


Assuntos
Arrestinas/farmacologia , Colecistocinina/farmacologia , Gastrinas/farmacologia , Receptor de Colecistocinina B/agonistas , Receptor de Colecistocinina B/metabolismo , Arrestinas/genética , Arrestinas/metabolismo , Colecistocinina/genética , Colecistocinina/metabolismo , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Endossomos/genética , Endossomos/metabolismo , Gastrinas/genética , Gastrinas/metabolismo , Células HEK293 , Humanos , Lisossomos/genética , Lisossomos/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Receptor de Colecistocinina B/genética , beta-Arrestinas
15.
EMBO J ; 29(19): 3222-35, 2010 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-20802461

RESUMO

ß-Arrestins have been implicated in the regulation of multiple signalling pathways. However, their role in organism development is not well understood. In this study, we report a new in vivo function of the Drosophila ß-arrestin Kurtz (Krz) in the regulation of two distinct developmental signalling modules: MAPK ERK and NF-κB, which transmit signals from the activated receptor tyrosine kinases (RTKs) and the Toll receptor, respectively. Analysis of the expression of effectors and target genes of Toll and the RTK Torso in krz maternal mutants reveals that Krz limits the activity of both pathways in the early embryo. Protein interaction studies suggest a previously uncharacterized mechanism for ERK inhibition: Krz can directly bind and sequester an inactive form of ERK, thus preventing its activation by the upstream kinase, MEK. A simultaneous dysregulation of different signalling systems in krz mutants results in an abnormal patterning of the embryo and severe developmental defects. Our findings uncover a new in vivo function of ß-arrestins and present a new mechanism of ERK inhibition by the Drosophila ß-arrestin Krz.


Assuntos
Arrestinas/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Inibidores Enzimáticos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Transdução de Sinais/fisiologia , Receptores Toll-Like/metabolismo , Animais , Arrestinas/farmacologia , Western Blotting , Células Cultivadas , Drosophila/metabolismo , Proteínas de Drosophila/farmacologia , Inibidores Enzimáticos/farmacologia , Técnicas de Inativação de Genes , Imunoprecipitação , Hibridização in Situ Fluorescente , Mutação/genética , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
Endocrinology ; 150(5): 2419-28, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19116340

RESUMO

We report here the desensitization and internalization of the relaxin receptor (RXFP1) after agonist activation in both primary human decidual cells and HEK293 cells stably transfected with RXFP1. The importance of beta-arrestin 2 in these processes has also been demonstrated. Thus, in HEK-RXFP1 cells the desensitization of RXFP1 was significantly increased when beta-arrestin 2 was overexpressed. After relaxin activation, beta-arrestin 2 was translocated to the cell membrane and RXFP1 underwent rapid internalization. We have previously shown that RXFP1 forms dimers/oligomers during its biosynthesis and trafficking to the plasma membrane, we now show that internalization of RXFP1 occurs through this dimerization/oligomerization. In nonagonist stimulated cells, it is known that the majority of the RXFP1 is located intracellularly and was confirmed in the cells used here. Constitutive internalization of RXFP1 could account for this and indeed, slow but robust constitutive internalization, which was increased after agonist stimulation was demonstrated. A carboxyl-terminal deleted RXFP1 variant had a similar level of constitutive agonist-independent internalization as the wild-type RXFP1 but lost sensitivity to agonist stimulation. This demonstrated the importance of the carboxyl terminus in agonist-stimulated receptor internalization. These data suggest that the autocrine/paracrine actions of relaxin in the decidua are under additional controls at the level of expression of its receptor on the surface of its target cells.


Assuntos
Linhagem Celular/metabolismo , Decídua/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Arrestinas/farmacologia , Comunicação Autócrina/genética , Comunicação Autócrina/fisiologia , Técnicas de Cultura de Células , Linhagem Celular/efeitos dos fármacos , Células Cultivadas , Decídua/efeitos dos fármacos , Dimerização , Feminino , Expressão Gênica/fisiologia , Humanos , Modelos Biológicos , Comunicação Parácrina/genética , Comunicação Parácrina/fisiologia , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/efeitos dos fármacos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Receptores de Peptídeos/agonistas , Receptores de Peptídeos/química , Relaxina/farmacologia , Transfecção
17.
Nature ; 457(7233): 1146-9, 2009 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-19122674

RESUMO

Insulin resistance, a hallmark of type 2 diabetes, is a defect of insulin in stimulating insulin receptor signalling, which has become one of the most serious public health threats. Upon stimulation by insulin, insulin receptor recruits and phosphorylates insulin receptor substrate proteins, leading to activation of the phosphatidylinositol-3-OH kinase (PI(3)K)-Akt pathway. Activated Akt phosphorylates downstream kinases and transcription factors, thus mediating most of the metabolic actions of insulin. Beta-arrestins mediate biological functions of G-protein-coupled receptors by linking activated receptors with distinct sets of accessory and effecter proteins, thereby determining the specificity, efficiency and capacity of signals. Here we show that in diabetic mouse models, beta-arrestin-2 is severely downregulated. Knockdown of beta-arrestin-2 exacerbates insulin resistance, whereas administration of beta-arrestin-2 restores insulin sensitivity in mice. Further investigation reveals that insulin stimulates the formation of a new beta-arrestin-2 signal complex, in which beta-arrestin-2 scaffolds Akt and Src to insulin receptor. Loss or dysfunction of beta-arrestin-2 results in deficiency of this signal complex and disturbance of insulin signalling in vivo, thereby contributing to the development of insulin resistance and progression of type 2 diabetes. Our findings provide new insight into the molecular pathogenesis of insulin resistance, and implicate new preventive and therapeutic strategies against insulin resistance and type 2 diabetes.


Assuntos
Arrestinas/deficiência , Resistência à Insulina/fisiologia , Animais , Arrestinas/genética , Arrestinas/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Técnicas de Silenciamento de Genes , Humanos , Insulina/farmacologia , Resistência à Insulina/genética , Camundongos , Camundongos Knockout , Mutação/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais , beta-Arrestina 2 , beta-Arrestinas
18.
Nat Immunol ; 9(8): 898-907, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18604210

RESUMO

The inhibitory signaling of natural killer (NK) cells is crucial in the regulation of innate immune responses. Here we show that the association of KIR2DL1, an inhibitory receptor of NK cells, with beta-arrestin 2 mediated recruitment of the tyrosine phosphatases SHP-1 and SHP-2 to KIR2DL1 and facilitated 'downstream' inhibitory signaling. Consequently, the cytotoxicity of NK cells was higher in beta-arrestin 2-deficient mice but was inhibited in beta-arrestin 2-transgenic mice. Moreover, beta-arrestin 2-deficient mice were less susceptible than wild-type mice to mouse cytomegalovirus infection, an effect that was abolished by depletion of NK cells. Our findings identify a previously unknown mechanism by which the inhibitory signaling in NK cells is regulated.


Assuntos
Arrestinas/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Receptores Imunológicos/imunologia , Transdução de Sinais/imunologia , Animais , Células Cultivadas , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Receptores Imunológicos/química , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , beta-Arrestina 2 , beta-Arrestinas
19.
Trends Pharmacol Sci ; 28(8): 416-22, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17644195

RESUMO

Seven-transmembrane receptors (7TMRs), the most common molecular targets of modern drug therapy, are critically regulated by beta-arrestins, which both inhibit classic G-protein signaling and initiate distinct beta-arrestin signaling. The interplay of G-protein and beta-arrestin signals largely determines the cellular consequences of 7TMR-targeted drugs. Until recently, a drug's efficacy for beta-arrestin recruitment was believed to be proportional to its efficacy for G-protein activities. This paradigm restricts 7TMR drug effects to a linear spectrum of responses, ranging from inhibition of all responses to stimulation of all responses. However, it is now clear that 'biased ligands' can selectively activate G-protein or beta-arrestin functions and thus elicit novel biological effects from even well-studied 7TMRs. Here, we discuss the current state of beta-arrestin-biased ligand research and the prospects for beta-arrestin bias as a therapeutic target. Consideration of ligand bias might have profound influences on the way scientists approach 7TMR-targeted drug discovery.


Assuntos
Arrestinas/farmacologia , Ligantes , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação Alostérica , Animais , Arrestinas/química , Arrestinas/metabolismo , Desenho de Fármacos , Humanos , Modelos Biológicos , Ligação Proteica , Receptores Acoplados a Proteínas G/química , beta-Arrestinas
20.
J Biol Chem ; 282(15): 11329-38, 2007 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-17303558

RESUMO

Beta-arrestin1, which regulates many aspects of seven transmembrane receptor (7TMR) biology, has also been shown to serve as an adaptor, which brings Mdm2, an E3 ubiquitin ligase to the insulin-like growth factor-1 receptor (IGF-1R), leading to its proteasome-dependent destruction. Here we demonstrate that IGF-1R stimulation also leads to ubiquitination of beta-arrestin1, which regulates vesicular trafficking and activation of ERK1/2. This beta-arrestin1-dependent ERK activity can occur even when the classical tyrosine kinase signaling is impaired. siRNA-mediated suppression of beta-arrestin1 in human melanoma cells ablates IGF-1-stimulated ERK and prolongs the G1 phase of the cell cycle. These data suggest that beta-arrestin-dependent ERK signaling by the IGF-1R regulates cell cycle progression and may thus be an important regulator of the growth of normal and malignant cells.


Assuntos
Arrestinas/farmacologia , Ciclo Celular , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Receptor IGF Tipo 1/metabolismo , Animais , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Camundongos , Mutação/genética , Fosforilação , Transporte Proteico , Receptor IGF Tipo 1/genética , Transdução de Sinais , Ubiquitina/metabolismo , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...