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










Base de dados
Intervalo de ano de publicação
1.
Cell Signal ; 44: 43-50, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29329779

RESUMO

The signalling functions of many G protein-coupled receptors (GPCRs) expressed in the myocardium are incompletely understood. Among these are the endothelin receptor (ETR) family and α1-adrenergic receptor (α1-AR), which are thought to couple to the G protein Gαq. In this study, we used transcriptome analysis to compare the signalling networks downstream of these receptors in primary neonatal rat cardiomyocytes. This analysis indicated increased expression of target genes of cAMP responsive element modulator (CREM) after 24 h treatment with the α1-AR agonist phenylephrine, but not the ETR agonist endothelin-1, suggesting a specific role for the α1-AR in promoting cAMP production in cardiomyocytes. To validate the difference observed between these two GPCRs, we used heterologous expression of the receptors and genetically encoded biosensors in HEK 293 cell lines. We validated that both α1A- and α1B-AR subtypes were able to lead to the accumulation of cAMP in response to phenylephrine in both the nucleus and cytoplasm in a Gαs-dependent manner. However, the ETR subtype ETA did not affect cAMP levels in either compartment. All three receptors were coupled to Gαq signalling as expected. Further, we showed that activation of PKA in different compartments was α1-AR subtype specific, with α1B-AR able to activate PKA in the cytoplasm and nucleus and α1A-AR only able to in the nucleus. We provide evidence for a pathway downstream of the α1-AR, and show that distinct pools of a receptor lead to differential activation of downstream effector proteins dependent on their cellular compartment.


Assuntos
Modulador de Elemento de Resposta do AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Miócitos Cardíacos/citologia , Receptor de Endotelina A/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Células HEK293 , Humanos , Fenilefrina/farmacologia , Ratos
2.
J Cardiovasc Pharmacol ; 71(4): 193-204, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28858907

RESUMO

There is significant evidence that internal pools of G protein-coupled receptors (GPCRs) exist and may be affected by both endogenous signaling molecules and hydrophobic pharmaceutical ligands, once assumed to only affect cell surface versions of these receptors. Here, we discuss evidence that the biology of nuclear GPCRs in particular is complex, rich, and highly interactive with GPCR signaling from the cell surface. Caging existing GPCR ligands may be an excellent means of further stratifying the phenotypic effects of known pharmacophores such as ß-adrenergic, angiotensin II, and type B endothelin receptor ligands in the cardiovascular system. We describe some synthetic strategies we have used to design ligands to go from in cellulo to in vivo experiments. We also consider how surface and intracellular GPCR signaling might be integrated and ways to dissect this. If they could be selectively targeted, nuclear GPCRs and their associated nucleoligands would represent a completely novel area for exploration by Pharma.


Assuntos
Fármacos Cardiovasculares/farmacologia , Doenças Cardiovasculares/tratamento farmacológico , Sistema Cardiovascular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Desenho de Fármacos , Reposicionamento de Medicamentos/métodos , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Animais , Fármacos Cardiovasculares/síntese química , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatologia , Núcleo Celular/genética , Humanos , Ligantes , Estrutura Molecular , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
3.
Cell Signal ; 30: 50-58, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27887991

RESUMO

GPCRs form signalling complexes with other receptors as part of dimers, G proteins and effector partners. A proteomic screen to identify proteins that associate with the ß2-adrenergic receptor (ß2AR) identified many of components of the Endoplasmic-Reticulum-Associated Degradation (ERAD) quality control system [1], including the valosin-containing protein (VCP/p97). Here, we validated the interaction of VCP with co-expressed FLAG-ß2AR, demonstrating, using an inducible expression system, that the interaction of FLAG-ß2AR and VCP is not an artifact of overexpression of the ß2AR per se. We knocked down VCP and noted that levels of FLAG-ß2AR were increased in cells with lower VCP levels. This increase in the level of FLAG-ß2AR did not lead to an increase in the level of functional receptor observed at the cell surface. Similarly, inhibition of the proteasome lead to a dramatic increase in the abundance of TAP-ß2AR, while cellular responses again remained unchanged. Taken together, our data suggests that a substantial proportion of the ß2AR produced is non-functional and VCP plays a key role in the maturation and trafficking of the ß2AR as part of the ERAD quality control process.


Assuntos
Biossíntese de Proteínas , Receptores Adrenérgicos beta 2/biossíntese , Proteína com Valosina/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Ligação Proteica/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos dos fármacos
4.
Methods Cell Biol ; 132: 319-37, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26928550

RESUMO

The use of biosensors either individually or as part of panels has now become a common technique to capturing signaling events in living cells. Such biosensors have become particularly important for probing biased signaling and allostery in G protein-coupled receptor drug screening efforts. However, assumptions about the portability of such biosensors between cell types may lead to misinterpretation of drug effects on specific signaling pathways in a given cellular context. Further, the output of a particular biosensor may be different depending on where it is localized in a cell. Here, we discuss strategies to mitigate these concerns which should feed into future biosensor design and usage.


Assuntos
Técnicas Biossensoriais , Transdução de Sinais , Núcleo Celular/enzimologia , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases , Receptores Acoplados a Proteínas G/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
5.
J Cardiovasc Pharmacol ; 65(2): 110-22, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25590750

RESUMO

According to the standard model of G protein-coupled receptor (GPCR) signaling, GPCRs are localized to the cell membrane where they respond to extracellular signals. Stimulation of GPCRs leads to the activation of heterotrimeric G proteins and their intracellular signaling pathways. However, this model fails to accommodate GPCRs, G proteins, and their downstream effectors that are found on the nuclear membrane or in the nucleus. Evidence from isolated nuclei indicates the presence of GPCRs on the nuclear membrane that can activate similar G protein-dependent signaling pathways in the nucleus as at the cell surface. These pathways also include activation of cyclic adenosine monophosphate, calcium and nitric oxide synthase signaling in cardiomyocytes. In addition, a number of distinct heterotrimeric and monomeric G proteins have been found in the nucleus of various cell types. This review will focus on understanding the function of nuclear G proteins with a focus on cardiac signaling where applicable.


Assuntos
Núcleo Celular/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Miócitos Cardíacos/fisiologia , Proteínas Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Cálcio/metabolismo , AMP Cíclico/metabolismo , Humanos , Óxido Nítrico Sintase/metabolismo , Transdução de Sinais/fisiologia
6.
Methods Mol Biol ; 1234: 161-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25304356

RESUMO

It has become clear in recent years that the Gßγ subunits of heterotrimeric proteins serve broad roles in the regulation of cellular activity and interact with many proteins in different subcellular locations including the nucleus. Protein affinity purification is a common method to identify and confirm protein interactions. When used in conjugation with mass spectrometry it can be used to identify novel protein interactions with a given bait protein. The tandem affinity purification (TAP) technique identifies partner proteins bound to tagged protein bait. Combined with protocols to enrich the nuclear fraction of whole cell lysate through sucrose cushions, TAP allows for purification of interacting proteins found specifically in the nucleus. Here we describe the use of the TAP technique on cytosolic and nuclear lysates to identify candidate proteins, through mass spectrometry, that bind to Gß1 subunits.


Assuntos
Proteínas de Transporte/isolamento & purificação , Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Cromatografia de Afinidade , Citosol/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Espectrometria de Massas em Tandem , Linhagem Celular , Cromatografia de Afinidade/métodos , Humanos , Espectrometria de Massas em Tandem/métodos
7.
Mol Pharmacol ; 83(2): 416-28, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23175530

RESUMO

This study assessed how conformational information encoded by ligand binding to δ-opioid receptors (DORs) is transmitted to Kir3.1/Kir3.2 channels. Human embryonic kidney 293 cells were transfected with bioluminescence resonance energy transfer (BRET) donor/acceptor pairs that allowed us to evaluate independently reciprocal interactions among signaling partners. These and coimmunoprecipitation studies indicated that DORs, Gßγ, and Kir3 subunits constitutively interacted with one another. GαoA associated with DORs and Gßγ, but despite being part of the complex, no evidence of its direct association with the channel was obtained. DOR activation by different ligands left DOR-Kir3 interactions unmodified but modulated BRET between DOR-GαoA, DOR-Gßγ, GαoA-Gßγ, and Gßγ-Kir3 interfaces. Ligand-induced BRET changes assessing Gßγ-Kir3.1 subunit interaction 1) followed similar kinetics to those monitoring the GαoA-Gßγ interface, 2) displayed the same order of efficacy as those observed at the DOR-Gßγ interface, 3) were sensitive to pertussis toxin, and 4) were predictive of whether a ligand could evoke channel currents. Conformational changes at the Gßγ/Kir3 interface were lost when Kir3.1 subunits were replaced by a mutant lacking essential sites for Gßγ-mediated activation. Thus, conformational information encoded by agonist binding to the receptor is relayed to the channel via structural rearrangements that involve repositioning of Gßγ with respect to DORs, GαoA, and channel subunits. Further, the fact that BRET changes at the Gßγ-Kir3 interface are predictive of a ligand's ability to induce channel currents points to these conformational biosensors as screening tools for identifying GPCR ligands that induce Kir3 channel activation.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Receptores Opioides delta/metabolismo , Sítios de Ligação , Linhagem Celular , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/química , Células HEK293 , Humanos , Cinética , Ligantes , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas , Receptores Opioides delta/química
8.
J Neurosci ; 32(14): 4827-40, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22492038

RESUMO

Opioid tendency to generate analgesic tolerance has been previously linked to biased internalization. Here, we assessed an alternative possibility; whether tolerance of delta opioid receptor agonists (DORs) could be related to agonist-specific recycling. A first series of experiments revealed that DOR internalization by DPDPE and SNC-80 was similar, but only DPDPE induced recycling. We then established that the non-recycling agonist SNC-80 generated acute analgesic tolerance that was absent in mice treated with DPDPE. Furthermore, both agonists stabilized different conformations, whose distinct interaction with Gßγ subunits led to different modalities of ß-arrestin2 (ßarr2) recruitment. In particular, bioluminescence resonance energy transfer (BRET) assays revealed that sustained activation by SNC-80 drew the receptor C terminus in close proximity of the N-terminal domain of Gγ2, causing ßarr2 to interact with receptors and Gßγ subunits. DPDPE moved the receptor C-tail away from the Gßγ dimer, resulting in ßarr2 recruitment to the receptor but not in the vicinity of Gγ2. These differences were associated with stable DOR-ßarr2 association, poor recycling, and marked desensitization following exposure to SNC-80, while DPDPE promoted transient receptor interaction with ßarr2 and effective recycling, which conferred protection from desensitization. Together, these data indicate that DORs may adopt ligand-specific conformations whose distinct recycling properties determine the extent of desensitization and are predictive of analgesic tolerance. Based on these findings, we propose that the development of functionally selective DOR ligands that favor recycling could constitute a valid strategy for the production of longer acting opioid analgesics.


Assuntos
Analgésicos Opioides/metabolismo , Arrestinas/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Analgésicos Opioides/farmacologia , Animais , Animais Recém-Nascidos , Arrestinas/fisiologia , Linhagem Celular Transformada , Células Cultivadas , Tolerância a Medicamentos/fisiologia , Subunidades beta da Proteína de Ligação ao GTP/fisiologia , Subunidades gama da Proteína de Ligação ao GTP/fisiologia , Células HEK293 , Humanos , Masculino , Camundongos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , beta-Arrestinas
9.
Methods Mol Biol ; 756: 149-63, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21870224

RESUMO

Bioluminescence energy transfer (BRET) has become a powerful tool to study protein-protein interactions and conformational changes among interacting proteins. In particular, BRET assays performed in living cells have revealed that heptahelical receptors (7TMRs), heterotrimeric G proteins and their proximal effectors form constitutive signalling complexes. BRET technology has also allowed us to demonstrate that these multimeric protein arrays remain intact throughout initial stages of receptor signalling, thus providing a platform for direct transmission of conformational information from activated receptors to downstream signalling partners. A clear example of the latter are the distinct intermolecular re-arrangements undergone by 7TMRs and G protein subunits following activation of the receptor by different ligands. Here we present protocols describing the type of BRET assay that has been used to reveal the existence of constitutive signalling arrays formed by 7TMRs and proximal signalling partners as well as the ability of complex components to undergo ligand-specific conformational changes.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Proteínas Heterotriméricas de Ligação ao GTP/química , Humanos , Ligantes , Conformação Proteica , Transdução de Sinais
10.
Med Sci (Paris) ; 26(8-9): 734-9, 2010.
Artigo em Francês | MEDLINE | ID: mdl-20819711

RESUMO

Opiates are the most effective analgesics available for the treatment of severe pain. However, their clinical use is restricted by unwanted side effects such as tolerance, physical dependence and respiratory depression. The strategy to develop new opiates with reduced side effects has mainly focused on the study and production of ligands that specifically bind to different opiate receptors subtypes. However, this strategy has not allowed the production of novel therapeutic ligands with a better side effects profile. Thus, other research strategies need to be explored. One which is receiving increasing attention is the possibility of exploiting ligand ability to stabilize different receptor conformations with distinct signalling profiles. This newly described property, termed functional selectivity, provides a potential means of directing the stimulus generated by an activated receptor towards a specific cellular response. Here we summarize evidence supporting the existence of ligand-specific active conformations for two opioid receptors subtypes (delta and mu), and analyze how functional selectivity may contribute in the production of longer lasting, better tolerated opiate analgesics. double dagger.


Assuntos
Analgésicos Opioides/metabolismo , Receptores Opioides/fisiologia , Aclimatação , Analgésicos/uso terapêutico , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Animais , Tolerância a Medicamentos , Humanos , Receptores Opioides/química , Receptores Opioides delta/química , Receptores Opioides delta/fisiologia , Receptores Opioides mu/química , Receptores Opioides mu/fisiologia , Transdução de Sinais
11.
J Cell Mol Med ; 13(1): 147-63, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18363847

RESUMO

Abstract An important limitation in the clinical use of opiates is progressive loss of analgesic efficacy over time. Development of analgesic tolerance is tightly linked to receptor desensitization. In the case of delta opioid receptors (DOR), desensitization is especially swift because receptors are rapidly internalized and are poorly recycled to the membrane. In the present study, we investigated whether Src activity contributed to this sorting pattern and to functional desensitization of DORs. A first series of experiments demonstrated that agonist binding activates Src and destabilizes a constitutive complex formed by the spontaneous association of DORs with the kinase. Src contribution to DOR desensitization was then established by showing that pre-treatment with Src inhibitor PP2 (20 microM; 1 hr) or transfection of a dominant negative Src mutant preserved DOR signalling following sustained exposure to an agonist. This protection was afforded without interfering with endocytosis, but suboptimal internalization interfered with PP2 ability to preserve DOR signalling, suggesting a post-endocytic site of action for the kinase. This assumption was confirmed by demonstrating that Src inhibition by PP2 or its silencing by siRNA increased membrane recovery of internalized DORs and was further corroborated by showing that inhibition of recycling by monensin or dominant negative Rab11 (Rab11S25N) abolished the ability of Src blockers to prevent desensitization. Finally, Src inhibitors accelerated recovery of DOR-Galphal3 coupling after desensitization. Taken together, these results indicate that Src dynamically regulates DOR recycling and by doing so contributes to desensitization of these receptors.


Assuntos
Tolerância a Medicamentos/fisiologia , Endocitose/fisiologia , Receptores Opioides delta/metabolismo , Quinases da Família src/metabolismo , Analgésicos Opioides/metabolismo , Animais , Linhagem Celular , D-Penicilina (2,5)-Encefalina/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Pirimidinas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Opioides delta/genética , Transdução de Sinais/fisiologia , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética
12.
J Biol Chem ; 283(22): 15078-88, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18381293

RESUMO

Heptahelical receptors communicate extracellular information to the cytosolic compartment by binding an extensive variety of ligands. They do so through conformational changes that propagate to intracellular signaling partners as the receptor switches from a resting to an active conformation. This active state has been classically considered unique and responsible for regulation of all signaling pathways controlled by a receptor. However, recent functional studies have challenged this notion and called for a paradigm where receptors would exist in more than one signaling conformation. This study used bioluminescence resonance energy transfer assays in combination with ligands of different functional profiles to provide in vivo physical evidence of conformational diversity of delta-opioid receptors (DORs). DORs and alpha(i1)beta(1)gamma(2) G protein subunits were tagged with Luc or green fluorescent protein to produce bioluminescence resonance energy transfer pairs that allowed monitoring DOR-G protein interactions from different vantage points. Results showed that DORs and heterotrimeric G proteins formed a constitutive complex that underwent structural reorganization upon ligand binding. Conformational rearrangements could not be explained by a two-state model, supporting the idea that DORs adopt ligand-specific conformations. In addition, conformational diversity encoded by the receptor was conveyed to the interaction among heterotrimeric subunits. The existence of multiple active receptor states has implications for the way we conceive specificity of signal transduction.


Assuntos
Transferência Ressonante de Energia de Fluorescência , Proteínas Heterotriméricas de Ligação ao GTP/química , Modelos Biológicos , Receptores Opioides delta/química , Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas Heterotriméricas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Humanos , Ligantes , Ligação Proteica/fisiologia , Estrutura Quaternária de Proteína/fisiologia , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais/fisiologia
13.
J Biol Chem ; 280(9): 7808-16, 2005 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-15632168

RESUMO

The present study showed that delta opioid receptor (deltaOR) ligands Tyr-Ticpsi [CH(2)-NH]Cha-Phe-OH (TICP) and ICI174864 behaved as inverse agonists in the cyclase pathway but induced agonist responses in the ERK cascade. Unlike ligands that behaved as agonists in both pathways, and whose stimulation of ERK was marked but transient (10 min), ERK activation by ICI174864 and TICP was moderate and sustained, lasting for more than 1 h in the case of TICP. Biochemical experiments showed that duration of ERK activation by agonists and "dual efficacy ligands" was inversely correlated with their ability to trigger receptor phosphorylation and degradation. Thus, although TICP stabilized deltaORs in a conformation that did not incorporate (32)P, was not a substrate for tyrosine kinase Src, and was not down-regulated following prolonged exposure to the drug, the conformation stabilized by D-Pen-2,5-enkephalin (DPDPE) incorporated (32)P, was phosphorylated by Src, and suffered degradation within the first 2 h of treatment. Inhibition of endocytosis by sucrose prolonged ERK activation by DPDPE increasing the decay half-life of the response to values that resembled those of dual efficacy ligands (from a 2-min decay t((1/2)) increased to 12 min). Src inhibitor PP2 also prolonged ERK stimulation by DPDPE. It did so by maintaining a sustained activation of the kinase at approximately 20% of maximum following an initial rapid reduction in the response. These results show that specific kinetics of ERK activation by agonists and dual efficacy ligands are determined, at least in part, by the differential ability of the two types of drugs to trigger mechanisms regulating deltaOR responsiveness.


Assuntos
Encefalina Leucina/análogos & derivados , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Receptores Opioides delta/metabolismo , Western Blotting , Linhagem Celular , AMP Cíclico/metabolismo , DNA/química , DNA Complementar/metabolismo , Eletroforese em Gel de Poliacrilamida , Endocitose , Encefalina Leucina/farmacologia , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Vetores Genéticos , Humanos , Imunoprecipitação , Cinética , Ligantes , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/farmacologia , Fosforilação , Transdução de Sinais , Sacarose/farmacologia , Tetra-Hidroisoquinolinas/farmacologia , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...