Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Med Chem ; 64(15): 11129-11147, 2021 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-34291633

RESUMO

Both previous and additional genetic knockdown studies reported herein implicate G protein-coupled receptor kinase 6 (GRK6) as a critical kinase required for the survival of multiple myeloma (MM) cells. Therefore, we sought to develop a small molecule GRK6 inhibitor as an MM therapeutic. From a focused library of known kinase inhibitors, we identified two hits with moderate biochemical potencies against GRK6. From these hits, we developed potent (IC50 < 10 nM) analogues with selectivity against off-target kinases. Further optimization led to the discovery of an analogue (18) with an IC50 value of 6 nM against GRK6 and selectivity against a panel of 85 kinases. Compound 18 has potent cellular target engagement and antiproliferative activity against MM cells and is synergistic with bortezomib. In summary, we demonstrate that targeting GRK6 with small molecule inhibitors represents a promising approach for MM and identify 18 as a novel, potent, and selective GRK6 inhibitor.


Assuntos
Antineoplásicos/farmacologia , Desenho de Fármacos , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Quinases de Receptores Acoplados a Proteína G/metabolismo , Humanos , Camundongos , Modelos Moleculares , Estrutura Molecular , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Quinazolinas/síntese química , Quinazolinas/química , Relação Estrutura-Atividade
2.
Cells ; 10(1)2020 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-33396400

RESUMO

Many receptors for neurotransmitters, such as dopamine, norepinephrine, acetylcholine, and neuropeptides, belong to the superfamily of G protein-coupled receptors (GPCRs). A general model posits that GPCRs undergo two-step homologous desensitization: the active receptor is phosphorylated by kinases of the G protein-coupled receptor kinase (GRK) family, whereupon arrestin proteins specifically bind active phosphorylated receptors, shutting down G protein-mediated signaling, facilitating receptor internalization, and initiating distinct signaling pathways via arrestin-based scaffolding. Here, we review the mechanisms of GRK-dependent regulation of neurotransmitter receptors, focusing on the diverse modes of GRK-mediated phosphorylation of receptor subtypes. The immediate signaling consequences of GRK-mediated receptor phosphorylation, such as arrestin recruitment, desensitization, and internalization/resensitization, are equally diverse, depending not only on the receptor subtype but also on phosphorylation by GRKs of select receptor residues. We discuss the signaling outcome as well as the biological and behavioral consequences of the GRK-dependent phosphorylation of neurotransmitter receptors where known.


Assuntos
Arrestinas/metabolismo , Quinases de Receptores Acoplados a Proteína G/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neurotransmissores/metabolismo , Animais , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/química , Humanos , Fosforilação , Transdução de Sinais/genética
3.
Nat Rev Cardiol ; 16(10): 612-622, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31186538

RESUMO

G protein-coupled receptors (GPCRs) are critical cellular sensors that mediate numerous physiological processes. In the heart, multiple GPCRs are expressed on various cell types, where they coordinate to regulate cardiac function by modulating critical processes such as contractility and blood flow. Under pathological settings, these receptors undergo aberrant changes in expression levels, localization and capacity to couple to downstream signalling pathways. Conventional therapies for heart failure work by targeting GPCRs, such as ß-adrenergic receptor and angiotensin II receptor antagonists. Although these treatments have improved patient survival, heart failure remains one of the leading causes of mortality worldwide. GPCR kinases (GRKs) are responsible for GPCR phosphorylation and, therefore, desensitization and downregulation of GPCRs. In this Review, we discuss the GPCR signalling pathways and the GRKs involved in the pathophysiology of heart disease. Given that increased expression and activity of GRK2 and GRK5 contribute to the loss of contractile reserve in the stressed and failing heart, inhibition of overactive GRKs has been proposed as a novel therapeutic approach to treat heart failure.


Assuntos
Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/metabolismo , Cardiopatias/tratamento farmacológico , Cardiopatias/fisiopatologia , Antagonistas Adrenérgicos beta/uso terapêutico , Animais , Catecolaminas/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/fisiopatologia , Humanos , Contração Muscular , Miócitos Cardíacos , Fragmentos de Peptídeos/genética , Receptores Adrenérgicos/metabolismo , Proteínas Recombinantes/genética , Transdução de Sinais/genética , beta-Arrestinas/metabolismo
4.
Int J Biol Sci ; 14(2): 189-203, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29483837

RESUMO

G protein-coupled receptors (GPCRs) are the largest family of plasma membrane receptors. Emerging evidence demonstrates that signaling through GPCRs affects numerous aspects of cancer biology such as vascular remolding, invasion, and migration. Therefore, development of GPCR-targeted drugs could provide a new therapeutic strategy to treating a variety of cancers. G protein-coupled receptor kinases (GRKs) modulate GPCR signaling by interacting with the ligand-activated GPCR and phosphorylating its intracellular domain. This phosphorylation initiates receptor desensitization and internalization, which inhibits downstream signaling pathways related to cancer progression. GRKs can also regulate non-GPCR substrates, resulting in the modulation of a different set of pathophysiological pathways. In this review, we will discuss the role of GRKs in modulating cell signaling and cancer progression, as well as the therapeutic potential of targeting GRKs.


Assuntos
Quinases de Receptores Acoplados a Proteína G/fisiologia , Neoplasias/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Animais , Comunicação Celular , Linhagem Celular Tumoral , Progressão da Doença , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/metabolismo , Humanos , Neoplasias/genética , Neoplasias/patologia , Fosforilação , Domínios Proteicos , Transdução de Sinais
5.
PLoS Pathog ; 14(1): e1006718, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29346437

RESUMO

Schistosomes are blood-dwelling trematodes with global impact on human and animal health. Because medical treatment is currently based on a single drug, praziquantel, there is urgent need for the development of alternative control strategies. The Schistosoma mansoni genome project provides a platform to study and connect the genetic repertoire of schistosomes to specific biological functions essential for successful parasitism. G protein-coupled receptors (GPCRs) form the largest superfamily of transmembrane receptors throughout the Eumetazoan phyla, including platyhelminths. Due to their involvement in diverse biological processes, their pharmacological importance, and proven druggability, GPCRs are promising targets for new anthelmintics. However, to identify candidate receptors, a more detailed understanding of the roles of GPCR signalling in schistosome biology is essential. An updated phylogenetic analysis of the S. mansoni GPCR genome (GPCRome) is presented, facilitated by updated genome data that allowed a more precise annotation of GPCRs. Additionally, we review the current knowledge on GPCR signalling in this parasite and provide new insights into the potential roles of GPCRs in schistosome reproduction based on the findings of a recent tissue-specific transcriptomic study in paired and unpaired S. mansoni. According to the current analysis, GPCRs contribute to gonad-specific functions but also to nongonad, pairing-dependent processes. The latter may regulate gonad-unrelated functions during the multifaceted male-female interaction. Finally, we compare the schistosome GPCRome to that of another parasitic trematode, Fasciola, and discuss the importance of GPCRs to basic and applied research. Phylogenetic analyses display GPCR diversity in free-living and parasitic platyhelminths and suggest diverse functions in schistosomes. Although their roles need to be substantiated by functional studies in the future, the data support the selection of GPCR candidates for basic and applied studies, invigorating the exploitation of this important receptor class for drug discovery against schistosomes but also other trematodes.


Assuntos
Quinases de Receptores Acoplados a Proteína G/metabolismo , Proteínas de Helminto/metabolismo , Modelos Biológicos , Schistosoma mansoni/metabolismo , Transdução de Sinais , Animais , Antiplatelmínticos/farmacologia , Fasciola/efeitos dos fármacos , Fasciola/genética , Fasciola/metabolismo , Fasciola/patogenicidade , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/química , Quinases de Receptores Acoplados a Proteína G/genética , Perfilação da Expressão Gênica , Genoma Helmíntico , Genômica/métodos , Proteínas de Helminto/antagonistas & inibidores , Proteínas de Helminto/química , Proteínas de Helminto/genética , Humanos , Especificidade de Órgãos , Filogenia , Inibidores de Proteínas Quinases/farmacologia , Schistosoma mansoni/efeitos dos fármacos , Schistosoma mansoni/genética , Schistosoma mansoni/patogenicidade , Transdução de Sinais/efeitos dos fármacos
6.
Sci Rep ; 6: 26812, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27245949

RESUMO

C-kit positive cardiac stem cells (CSCs) have been shown to contribute to myocardial regeneration after infarction. Previously, we have shown that the c-kit ligand stem cell factor (SCF) can induce CSC migration into the infarcted area during myocardial infarction (MI). However, the precise mechanism involved is not fully understood. In this study, we found that CSCs also express C-X-C chemokine receptor type 4 (CXCR4), which is a typical member of the seven transmembrane-spanning G protein-coupled receptor (GPCR). In vitro, activation of c-kit signalling by SCF promotes migration of CSCs with increased phosphorylation of CXCR4-serine 339, p38 mitogen-activated protein kinase (p38 MAPK) and extracellular regulated protein kinases 1/2 (ERK1/2). Knockdown of CXCR4 expression by siRNA reduces SCF/c-kit-induced migration and downstream signalling. As previously reported, CXCR4-serine 339 phosphorylation is mainly regulated by GPCR kinase 6 (GRK6); thus, silencing of GRK6 expression by siRNA impairs CXCR4-serine 339 phosphorylation and migration of CSCs caused by SCF. In vivo, knockdown of GRK6 impairs the ability of CSCs to migrate into peri-infarcted areas. These results demonstrate that SCF-induced CSC migration is regulated by the transactivation of CXCR4-serine 339 phosphorylation, which is mediated by GRK6.


Assuntos
Células-Tronco Adultas/fisiologia , Quimiotaxia/fisiologia , Quinases de Receptores Acoplados a Proteína G/fisiologia , Miocárdio/citologia , Proteínas Proto-Oncogênicas c-kit/fisiologia , Receptores CXCR4/metabolismo , Fator de Células-Tronco/fisiologia , Animais , Células Cultivadas , Ativação Enzimática , Feminino , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/genética , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Fosforilação , Fosfosserina/metabolismo , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno/genética , Ativação Transcricional , Transfecção
7.
Mol Pharmacol ; 89(5): 585-92, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26984025

RESUMO

The recent, unfortunate death of Alfred G. ("Al") Gilman, M.D., Ph.D., represents a sad signpost for an era spanning over 40 years in molecular pharmacology. Gilman's discoveries, influence, and persona were dominant forces in research and training in pharmacology. Here, we review the progression of ideas and knowledge that spawned early work by Gilman and collaborators (among them, one of the authors) and later efforts (including those of the other author) that have recently yielded a comprehensive and precise structural understanding of fundamental topics in pharmacology: the binding of ligands to G protein-coupled receptors (GPCRs) and the interaction of GPCRs with heterotrimeric G proteins and effector molecules. Those data provide new and important insights into the molecular basis that underlies affinity and efficacy, two of the most important features of drug action, which represent the latest chapter in the saga that Al Gilman's work helped launch.


Assuntos
Bioquímica/história , AMP Cíclico/fisiologia , Quinases de Receptores Acoplados a Proteína G/metabolismo , Modelos Biológicos , Medicina Molecular/história , Farmacologia/história , Sistemas do Segundo Mensageiro , Adenilil Ciclases/química , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Animais , Autoria , Bioquímica/educação , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/química , Quinases de Receptores Acoplados a Proteína G/genética , História do Século XX , História do Século XXI , Humanos , Cinética , Liderança , Ligantes , Medicina Molecular/educação , National Academy of Sciences, U.S. , Prêmio Nobel , Farmacocinética , Farmacologia/educação , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Estados Unidos
8.
Oncol Rep ; 35(2): 1027-33, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26718636

RESUMO

Hypopharyngeal squamous cell carcinoma (HSCC) is one of the most common head and neck cancers with high invasiveness and poor prognosis. To identify targeted therapeutics against metastasis, a better understanding of the regulation of HSCC cell invasion is needed. In recent years, G protein-coupled receptor kinases (GRKs) have been implicated in cancer metastasis through phosphorylation of the activated form of G protein coupled receptors (GPCRs). However, there is little information regarding GRKs expression in HSCC. In the present study, we examined GRK6 expression in HSCC and also assessed the possible cause of its aberrant expression, as well as its clinical significance. Real-time quantitative PCR (qPCR) and western blotting were performed to analyze the expression of GRK6 in HSCC tissues and corresponding non-malignant tissues. Subsequently, paired HSCC and corresponding non-malignant tissues were evaluated for the methylation status of GRK6 gene promoter using methylation-specific PCR (MSP). Furthermore, we investigated the methylation status and the clinicopathological significance of GRK6 in 45 paired HSCC and corresponding non-malignant tissues. The suppression of GRK6 in hypopharyngeal cell line FaDu by GRK6-shRNA lentivirus transfection was utilized to detect the role of GRK6 in hypopharyngeal cancer progression. Our results showed that the expression of GRK6 mRNA and protein was significantly lower in HSCC than in corresponding adjacent non-tumor tissues, and this downregulation was found to be in accordance with aberrant methylation of the gene. Hypermethylation of the gene was observed in 77.8% (35/45) of the HSCC tissues, while it was found in only 42.2% (19/45) of the corresponding non-malignant tissues. GRK6 methylation was related to depth of tumor invasion and TNM stage. Upon treatment with 5-aza-2'-deoxycytidine, GRK6 expression was upregulated in FaDu cells, and cell invasion was signinficantly inhibited. Furthermore, the suppression of GRK6 by shRNA transfection enhanced FaDu cells invasion. Our results indicate that the aberrant methylation of GRK6 gene promoter may underlie its downregulation in HSCC, and may play an important role in the metastasis of HSCC.


Assuntos
Carcinoma de Células Escamosas/genética , Metilação de DNA , Quinases de Receptores Acoplados a Proteína G/genética , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias Hipofaríngeas/genética , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Metilação de DNA/efeitos dos fármacos , Decitabina , Regulação para Baixo , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/biossíntese , Quinases de Receptores Acoplados a Proteína G/fisiologia , Humanos , Neoplasias Hipofaríngeas/mortalidade , Neoplasias Hipofaríngeas/patologia , Invasividade Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/fisiologia , Prognóstico , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , RNA Interferente Pequeno/genética
9.
Mol Cell Endocrinol ; 407: 57-66, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25766502

RESUMO

Bradykinin is associated with infections and inflammation, which given the strong correlation between uterine infection and preterm labour may imply that it could play a role in this process. Therefore, we investigated bradykinin signalling, and the roles that arrestin proteins play in their regulation in human myometrial cells. Bradykinin induced rapid, transient intracellular Ca(2+) increases that were inhibited following B2 receptor (B2R) antagonism. Arrestin2 or arrestin3 depletion enhanced and prolonged bradykinin-stimulated Ca(2+) responses, and attenuated B2R desensitisation. Knockdown of either arrestin enhanced B2R-stimulated ERK1/2 signals. Moreover, depletion of either arrestin elevated peak-phase p38-MAPK signalling, yet only arrestin3 depletion prolonged B2R-induced p38-MAPK signals. Arrestin2-knockdown augmented bradykinin-induced cell movement. Bradykinin stimulates pro-contractile signalling mechanisms in human myometrial cells and arrestin proteins play key roles in their regulation. Our data suggest bradykinin not only acts as an utertonin, but may also have the potential to enhance the contractile environment of the uterus.


Assuntos
Arrestinas/genética , Bradicinina/farmacologia , Cálcio/metabolismo , Células Musculares/efeitos dos fármacos , Arrestinas/antagonistas & inibidores , Bradicinina/metabolismo , Sinalização do Cálcio , Linhagem Celular Transformada , Movimento Celular , Feminino , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/genética , Quinases de Receptores Acoplados a Proteína G/metabolismo , Regulação da Expressão Gênica , Humanos , 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 , Células Musculares/citologia , Células Musculares/metabolismo , Contração Muscular/efeitos dos fármacos , Miométrio/citologia , Miométrio/efeitos dos fármacos , Miométrio/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor B2 da Bradicinina/genética , Receptor B2 da Bradicinina/metabolismo , beta-Arrestinas , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
ACS Chem Biol ; 10(1): 310-9, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25238254

RESUMO

Selective inhibitors of individual subfamilies of G protein-coupled receptor kinases (GRKs) would serve as useful chemical probes as well as leads for therapeutic applications ranging from heart failure to Parkinson's disease. To identify such inhibitors, differential scanning fluorimetry was used to screen a collection of known protein kinase inhibitors that could increase the melting points of the two most ubiquitously expressed GRKs: GRK2 and GRK5. Enzymatic assays on 14 of the most stabilizing hits revealed that three exhibit nanomolar potency of inhibition for individual GRKs, some of which exhibiting orders of magnitude selectivity. Most of the identified compounds can be clustered into two chemical classes: indazole/dihydropyrimidine-containing compounds that are selective for GRK2 and pyrrolopyrimidine-containing compounds that potently inhibit GRK1 and GRK5 but with more modest selectivity. The two most potent inhibitors representing each class, GSK180736A and GSK2163632A, were cocrystallized with GRK2 and GRK1, and their atomic structures were determined to 2.6 and 1.85 Å spacings, respectively. GSK180736A, developed as a Rho-associated, coiled-coil-containing protein kinase inhibitor, binds to GRK2 in a manner analogous to that of paroxetine, whereas GSK2163632A, developed as an insulin-like growth factor 1 receptor inhibitor, occupies a novel region of the GRK active site cleft that could likely be exploited to achieve more selectivity. However, neither compound inhibits GRKs more potently than their initial targets. This data provides the foundation for future efforts to rationally design even more potent and selective GRK inhibitors.


Assuntos
Descoberta de Drogas/métodos , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Inibidores de Proteínas Quinases , Bibliotecas de Moléculas Pequenas , Animais , Bovinos , Cristalografia por Raios X , Escherichia coli/genética , Quinases de Receptores Acoplados a Proteína G/química , Quinases de Receptores Acoplados a Proteína G/genética , Humanos , Cinética , Modelos Moleculares , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade
11.
ACS Chem Biol ; 10(1): 246-56, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24984143

RESUMO

Small molecules that inhibit the protein kinase A, G, and C (AGC) family of serine/threonine kinases can exert profound effects on cell homeostasis and thereby regulate fundamental processes such as heart rate, blood pressure, and metabolism, but there is not yet a clinically approved drug in the United States selective for a member of this family. One subfamily of AGC kinases, the G protein-coupled receptor (GPCR) kinases (GRKs), initiates the desensitization of active GPCRs. Of these, GRK2 has been directly implicated in the progression of heart failure. Thus, there is great interest in the identification of GRK2-specific chemical probes that can be further developed into therapeutics. Herein, we compare crystal structures of small molecule inhibitors in complex with GRK2 to those of highly selective compounds in complex with Rho-associated coiled-coil containing kinase 1 (ROCK1), a closely related AGC kinase. This analysis suggests that reduced hydrogen-bond formation with the hinge of the kinase domain, occupation of the hydrophobic subsite, and, consequently, higher buried surface area are key drivers of potency and selectivity among GRK inhibitors.


Assuntos
Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Descoberta de Drogas , Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinases de Receptores Acoplados a Proteína G/genética , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Inibidores de Proteínas Quinases/química , Estrutura Terciária de Proteína , Bibliotecas de Moléculas Pequenas/química , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética
12.
Curr Opin Cell Biol ; 27: 25-31, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24680427

RESUMO

The atomic structure of a protein can greatly advance our understanding of molecular recognition and catalysis, properties of fundamental importance in signal transduction. However, a single structure is incapable of fully describing how a protein functions, particularly when allostery is involved. Recent advances in the structure and function of G protein-coupled receptor (GPCR) kinases (GRKs) have concentrated on the mechanism of their inhibition by small and large molecules. These studies have generated a wealth of new information on the conformational flexibility of these enzymes, which opens new avenues for the development of selective chemical probes and provides deeper insights into the molecular basis for activation of these enzymes by GPCRs and phospholipids.


Assuntos
Quinases de Receptores Acoplados a Proteína G/química , Quinases de Receptores Acoplados a Proteína G/metabolismo , Animais , Sítios de Ligação , Ativação Enzimática , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Humanos , Modelos Moleculares , Fosfolipídeos/metabolismo , Conformação Proteica , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
13.
Expert Rev Hematol ; 7(1): 9-11, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24405278

RESUMO

Keith Stewart is the Dean for Research at Mayo Clinic (AZ, USA). He holds the Vasek and Anna Maria Polak Endowed Professorship in Cancer Research. He received his medical degree at Aberdeen University Medical School and trained in internal medicine and hematology in Glasgow, Kingston, Toronto and Boston. He was a consultant at the Toronto General and Princess Margaret Hospitals from 1992 to 2005 and a Professor in the Faculty of Medicine at the University of Toronto. He joined Mayo Clinic in Arizona in 2005. His lab-based research has focused on the genomics and developmental therapeutics of multiple myeloma. He has led numerous clinical trials from 'first in man', through to large international Phase III studies. Funding for his research includes the National Cancer Institute, the Leukemia and Lymphoma Society (LLS) and the Multiple Myeloma Research Foundation (MMRF). In addition, he is an associate editor of Blood, the journal of the American Society of Hematology (ASH).


Assuntos
Genoma Humano , Mieloma Múltiplo/genética , Anticorpos Monoclonais/uso terapêutico , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/metabolismo , Humanos , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Oligopeptídeos/uso terapêutico , Medicina de Precisão , Inibidores de Proteassoma/uso terapêutico , Interferência de RNA , Análise de Sequência de DNA
14.
Mol Pharmacol ; 85(2): 237-48, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24220010

RESUMO

Recently we identified the serotonin reuptake inhibitor paroxetine as an inhibitor of G protein-coupled receptor kinase 2 (GRK2) that improves cardiac performance in live animals. Paroxetine exhibits up to 50-fold selectivity for GRK2 versus other GRKs. A better understanding of the molecular basis of this selectivity is important for the development of even more selective and potent small molecule therapeutics and chemical genetic probes. We first sought to understand the molecular mechanisms underlying paroxetine selectivity among GRKs. We directly measured the K(D) for paroxetine and assessed its mechanism of inhibition for each of the GRK subfamilies and then determined the atomic structure of its complex with GRK1, the most weakly inhibited GRK tested. Our results suggest that the selectivity of paroxetine for GRK2 largely reflects its lower affinity for adenine nucleotides. Thus, stabilization of off-pathway conformational states unique to GRK2 will likely be key for the development of even more selective inhibitors. Next, we designed a benzolactam derivative of paroxetine that has optimized interactions with the hinge of the GRK2 kinase domain. The crystal structure of this compound in complex with GRK2 confirmed the predicted interactions. Although the benzolactam derivative did not significantly alter potency of inhibition among GRKs, it exhibited 20-fold lower inhibition of serotonin reuptake. However, there was an associated increase in the potency for inhibition of other AGC kinases, suggesting that the unconventional hydrogen bond formed by the benzodioxole ring of paroxetine is better accommodated by GRKs.


Assuntos
Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Paroxetina/análogos & derivados , Paroxetina/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Trifosfato de Adenosina/metabolismo , Cristalografia , Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Quinase 2 de Receptor Acoplado a Proteína G/química , Quinases de Receptores Acoplados a Proteína G/química , Ligação de Hidrogênio , Paroxetina/química , Fosforilação , Conformação Proteica
15.
Curr Pharm Des ; 19(28): 5127-34, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23607665

RESUMO

With a constant focus on the primary tumor, the current approaches in drug development in oncology yield dismal results. However over 90 percent of cancer deaths today are due to metastasis formation and yet there is no anti-metastatic drug on the market. Tumor cell migration is the essential prerequisite for invasion and metastasis formation. It is regulated by signal substances in terms of the grade of activity and in terms of direction (chemotaxis). The latter is important for the organotropism, the localization of metastasis in certain organs. Ligands to G protein-coupled receptors, mainly chemokines and neurotransmitters, as well as ligands to receptor kinases, mainly cytokines and growth factors, form the most important group of such regulators. We provide an overview of currently available agonists and antagonists to these receptors, which have a potential as anti-metastatic targets. Moreover we provide with the example of beta-blockers, how established drugs in other indications are possibly effective and can be co-opted as such anti-metastatics. The increasing knowledge of such regulators opens new opportunities to target cancer spreading and may put forth the development of antimetastatic drugs for oncological therapy.


Assuntos
Antineoplásicos/uso terapêutico , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Terapia de Alvo Molecular , Metástase Neoplásica/prevenção & controle , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Movimento Celular/efeitos dos fármacos , Quinases de Receptores Acoplados a Proteína G/metabolismo , Humanos , Metástase Neoplásica/tratamento farmacológico , Proteínas de Neoplasias/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Receptores Acoplados a Proteínas G/metabolismo
16.
Mol Biol Cell ; 24(11): 1649-60, S1-3, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23576548

RESUMO

Most α-synuclein (α-syn) deposited in Lewy bodies, the pathological hallmark of Parkinson disease (PD), is phosphorylated at Ser-129. However, the physiological and pathological roles of this modification are unclear. Here we investigate the effects of Ser-129 phosphorylation on dopamine (DA) uptake in dopaminergic SH-SY5Y cells expressing α-syn. Subcellular fractionation of small interfering RNA (siRNA)-treated cells shows that G protein-coupled receptor kinase 3 (GRK3), GRK5, GRK6, and casein kinase 2 (CK2) contribute to Ser-129 phosphorylation of membrane-associated α-syn, whereas cytosolic α-syn is phosphorylated exclusively by CK2. Expression of wild-type α-syn increases DA uptake, and this effect is diminished by introducing the S129A mutation into α-syn. However, wild-type and S129A α-syn equally increase the cell surface expression of dopamine transporter (DAT) in SH-SY5Y cells and nonneuronal HEK293 cells. In addition, siRNA-mediated knockdown of GRK5 or GRK6 significantly attenuates DA uptake without altering DAT cell surface expression, whereas knockdown of CK2 has no effect on uptake. Taken together, our results demonstrate that membrane-associated α-syn enhances DA uptake capacity of DAT by GRKs-mediated Ser-129 phosphorylation, suggesting that α-syn modulates intracellular DA levels with no functional redundancy in Ser-129 phosphorylation between GRKs and CK2.


Assuntos
Dopamina/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/genética , Quinases de Receptores Acoplados a Proteína G/genética , Neurônios/metabolismo , Serina/metabolismo , alfa-Sinucleína/genética , Transporte Biológico , Caseína Quinase II/antagonistas & inibidores , Caseína Quinase II/genética , Caseína Quinase II/metabolismo , Linhagem Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Neurônios/citologia , Fosforilação , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , alfa-Sinucleína/metabolismo
17.
Brain Res Bull ; 95: 70-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23352984

RESUMO

In the present study, we investigated the mechanisms of brain derived neurotrophic factor (BDNF) in regulating cortical neuron premature synapse formation. KN-93, a specific inhibitor of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), and G-protein-coupled receptor kinase interactor-1 (G1T1) siRNA were utilized, and the premature synapse formation of cortical neurons was detected under BDNF stimulation. Plasmids HA-GIT1, HA-GIT1 (ΔSLD), HA-GIT1 (S419A) and Flag-CaMKIIß were constructed. The interaction between GIT1 and CaMKIIß, and their influence on the premature synapse formation of BDNF-stimulated cortical neurons were examined. BDNF-stimulated cortical neurons were associated with increased premature synapse formation, the enhancement of phosphorylation for CaMKIIß, and the combination of GIT1 and p-CaMKII(thr286). G1T1 siRNA and KN-93 inhibited premature synapse formation in cortical neurons. The interaction between GIT1 and CaMKIIß required SLD domain and serine 419 in GIT1. BDNF-induced CaMKIIß phosphorylation and premature synapse formation were suppressed in GIT1 (S419A) transfected cortical neurons. By interacting with CaMKIIß, G1T1 (S419) were shown to participate in BDNF-induced premature synapse formation within cortical neurons.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Calmodulina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Fosfoproteínas/metabolismo , Sinapses/metabolismo , Animais , Benzilaminas , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Neurônios/metabolismo , Ratos , Serina/metabolismo , Transdução de Sinais/fisiologia , Sulfonamidas
18.
J Immunol ; 189(6): 2824-32, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22869904

RESUMO

The chemokine receptors, CXCR1 and CXCR2, couple to Gαi to induce leukocyte recruitment and activation at sites of inflammation. Upon activation by CXCL8, these receptors become phosphorylated, desensitized, and internalized. In this study, we investigated the role of different G protein-coupled receptor kinases (GRKs) in CXCR1- and CXCR2-mediated cellular functions. To that end, short hairpin RNA was used to inhibit GRK2, 3, 5, and 6 in RBL-2H3 cells stably expressing CXCR1 or CXCR2, and CXCL8-mediated receptor activation and regulation were assessed. Inhibition of GRK2 and GRK6 increased CXCR1 and CXCR2 resistance to phosphorylation, desensitization, and internalization, respectively, and enhanced CXCL8-induced phosphoinositide hydrolysis and exocytosis in vitro. GRK2 depletion diminished CXCR1-induced ERK1/2 phosphorylation but had no effect on CXCR2-induced ERK1/2 phosphorylation. GRK6 depletion had no significant effect on CXCR1 function. However, peritoneal neutrophils from mice deficient in GRK6 (GRK6(-/-)) displayed an increase in CXCR2-mediated G protein activation but in vitro exhibited a decrease in chemotaxis, receptor desensitization, and internalization relative to wild-type (GRK6(+/+)) cells. In contrast, neutrophil recruitment in vivo in GRK6(-/-) mice was increased in response to delivery of CXCL1 through the air pouch model. In a wound-closure assay, GRK6(-/-) mice showed enhanced myeloperoxidase activity, suggesting enhanced neutrophil recruitment, and faster wound closure compared with GRK6(+/+) animals. Taken together, the results indicate that CXCR1 and CXCR2 couple to distinct GRK isoforms to mediate and regulate inflammatory responses. CXCR1 predominantly couples to GRK2, whereas CXCR2 interacts with GRK6 to negatively regulate receptor sensitization and trafficking, thus affecting cell signaling and angiogenesis.


Assuntos
Quinases de Receptores Acoplados a Proteína G/metabolismo , Neutrófilos/imunologia , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo , Animais , Linhagem Celular Tumoral , Exocitose/genética , Exocitose/imunologia , Feminino , Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Quinase 2 de Receptor Acoplado a Proteína G/deficiência , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/deficiência , Humanos , Interleucina-8/fisiologia , Leucemia Basofílica Aguda/genética , Leucemia Basofílica Aguda/imunologia , Leucemia Basofílica Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/imunologia , Neutrófilos/enzimologia , Neutrófilos/metabolismo , Fosforilação/genética , Ratos , Receptores de Interleucina-8A/fisiologia , Receptores de Interleucina-8B/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
19.
Curr Pharm Des ; 18(31): 4839-53, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22632864

RESUMO

Traditional Japanese herbal, or Kampo medicine was developed and modified from Chinese herbal medicine. After the Japanese government approved Kampo for clinical use, much attention has been paid to establishing scientific evidence for the effectiveness of these medicines. Recent progress has been made in elucidating the mechanisms of action of some types of Kampo medicine, including rikkunshito (RKT), daikenchuto, and yokukansan. In this review, we focused on identifying the target molecules and the active ingredients of RKT. Thus far, many target molecules have been implicated in the mechanism of action of Kampo medicines, such as ion channels, enzymes, and receptors. In particular, G protein-coupled receptors are attractive candidates for explaining herbal medicine activity. This is particularly true of RKT, which is composed of 8 independent, crude drug extracts. Recent reports have shown that RKT elicits its effects through dual action to the G protein-coupled receptors: inhibition of serotonergic 5-HT2C and 5-HT2B receptors and activation of ghrelin receptors via specific ingredients of RKT. In addition, we suggest that the identification of the effective ingredients from Kampo medicines could contribute to the discovery and development of new drugs by means of modern high-throughput drug screening technology.


Assuntos
Medicina Kampo , Preparações de Plantas/farmacologia , Animais , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/química , Quinases de Receptores Acoplados a Proteína G/metabolismo , Grelina/agonistas , Grelina/metabolismo , Humanos , Panax , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Preparações de Plantas/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Receptores de Grelina/agonistas , Receptores de Grelina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Zanthoxylum , Zingiberaceae
20.
Proc Natl Acad Sci U S A ; 109(18): 7055-60, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22509025

RESUMO

ß-Arrestins are multifunctional proteins that play central roles in G protein-coupled receptor (GPCR) trafficking and signaling. ß-Arrestin1 is also recruited to the insulin-like growth factor-1 receptor (IGF-1R), a receptor tyrosine kinase (RTK), mediating receptor degradation and signaling. Because GPCR phosphorylation by GPCR-kinases (GRKs) governs interactions of the receptors with ß-arrestins, we investigated the regulatory roles of the four widely expressed GRKs on IGF-1R signaling/degradation. By suppressing GRK expression with siRNA, we demonstrated that lowering GRK5/6 abolishes IGF1-mediated ERK and AKT activation, whereas GRK2 inhibition increases ERK activation and partially inhibits AKT signaling. Conversely, ß-arrestin-mediated ERK signaling is enhanced by overexpression of GRK6 and diminished by GRK2. Similarly, we demonstrated opposing effects of GRK2 and -6 on IGF-1R degradation: GRK2 decreases whereas GRK6 enhances ligand-induced degradation. GRK2 and GRK6 coimmunoprecipitate with IGF-1R and increase IGF-1R serine phosphorylation, promoting ß-arrestin1 association. Using immunoprecipitation, confocal microscopy, and FRET analysis, we demonstrated ß-arrestin/IGF-1R association to be transient for GRK2 and stable for GRK6. Using bioinformatic studies we identified serines 1248 and 1291 as the major serine phosphorylation sites of the IGF-1R, and subsequent mutation analysis demonstrated clear effects on IGF-1R signaling and degradation, mirroring alterations by GRKs. Targeted mutation of S1248 recapitulates GRK2 modulation, whereas S1291 mutation resembles GRK6 effects on IGF-1R signaling/degradation, consistent with GRK isoform-specific serine phosphorylation. This study demonstrates distinct roles for GRK isoforms in IGF-1R signaling through ß-arrestin binding with divergent functional outcomes.


Assuntos
Quinases de Receptores Acoplados a Proteína G/metabolismo , Receptor IGF Tipo 1/metabolismo , Sequência de Aminoácidos , Animais , Arrestinas/metabolismo , Sequência de Bases , Linhagem Celular , Transferência Ressonante de Energia de Fluorescência , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/genética , Células HEK293 , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , RNA Interferente Pequeno/genética , Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/genética , Serina/química , Transdução de Sinais , Especificidade por Substrato , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA