Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Biochemistry ; 55(20): 2841-8, 2016 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-27088923

RESUMO

Interactions between proteins and cell membranes are critical for biological processes such as transmembrane signaling, and specific components of the membrane may play roles in helping to organize or mandate particular conformations of both integral and peripheral membrane proteins. One example of a signaling enzyme whose function is dependent on membrane binding and whose activity is affected by specific lipid components is G protein-coupled receptor (GPCR) kinase 2 (GRK2). Efficient GRK2-mediated phosphorylation of activated GPCRs is dependent not only on its recruitment to the membrane by heterotrimeric Gßγ subunits but also on the presence of highly negatively charged lipids, in particular phosphatidylinositol 4',5'-bisphosphate (PIP2). We hypothesized that PIP2 may favor a distinct orientation of the GRK2-Gßγ complex on the membrane that is more optimal for function. In this study, we compared the possible orientations of the GRK2-Gßγ complex and Gßγ alone on model cell membranes prepared with various anionic phospholipids as deduced from sum frequency generation vibrational and attenuated total reflectance Fourier transform infrared spectroscopic methods. Our results indicate that PIP2 affects the membrane orientation of the GRK2-Gß1γ2 complex but not that of complexes formed with anionic phospholipid binding deficient mutations in the GRK2 pleckstrin homology (PH) domain. Gß1γ2 exhibits a similar orientation on the lipid bilayer regardless of its lipid composition. The PIP2-induced orientation of the GRK2-Gß1γ2 complex is therefore most likely caused by specific interactions between PIP2 and the GRK2 PH domain. Thus, PIP2 not only helps recruit GRK2 to the membrane but also "fine tunes" the orientation of the GRK2-Gßγ complex so that it is better positioned to phosphorylate activated GPCRs.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/química , Subunidades beta da Proteína de Ligação ao GTP/química , Subunidades gama da Proteína de Ligação ao GTP/química , Bicamadas Lipídicas/química , Complexos Multienzimáticos/química , Animais , Bovinos , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Domínios Proteicos
2.
J Biol Chem ; 290(34): 20649-20659, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26032411

RESUMO

G protein-coupled receptor kinases (GRKs) regulate cell signaling by initiating the desensitization of active G protein-coupled receptors. The two most widely expressed GRKs (GRK2 and GRK5) play a role in cardiovascular disease and thus represent important targets for the development of novel therapeutic drugs. In the course of a GRK2 structure-based drug design campaign, one inhibitor (CCG215022) exhibited nanomolar IC50 values against both GRK2 and GRK5 and good selectivity against other closely related kinases such as GRK1 and PKA. Treatment of murine cardiomyocytes with CCG215022 resulted in significantly increased contractility at 20-fold lower concentrations than paroxetine, an inhibitor with more modest selectivity for GRK2. A 2.4 Å crystal structure of the GRK5·CCG215022 complex was determined and revealed that the inhibitor binds in the active site similarly to its parent compound GSK180736A. As designed, its 2-pyridylmethyl amide side chain occupies the hydrophobic subsite of the active site where it forms three additional hydrogen bonds, including one with the catalytic lysine. The overall conformation of the GRK5 kinase domain is similar to that of a previously determined structure of GRK6 in what is proposed to be its active state, but the C-terminal region of the enzyme adopts a distinct conformation. The kinetic properties of site-directed mutants in this region are consistent with the hypothesis that this novel C-terminal structure is representative of the membrane-bound conformation of the enzyme.


Assuntos
Fármacos Cardiovasculares/química , Inibidores Enzimáticos/química , Quinase 5 de Receptor Acoplado a Proteína G/química , Miócitos Cardíacos/efeitos dos fármacos , Piridinas/química , Animais , Fármacos Cardiovasculares/síntese química , Fármacos Cardiovasculares/farmacologia , Domínio Catalítico , Bovinos , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Quinase 5 de Receptor Acoplado a Proteína G/genética , Quinase 5 de Receptor Acoplado a Proteína G/isolamento & purificação , Expressão Gênica , Septos Cardíacos/química , Septos Cardíacos/citologia , Septos Cardíacos/efeitos dos fármacos , Septos Cardíacos/enzimologia , Ventrículos do Coração/química , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/enzimologia , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Cinética , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/química , Miócitos Cardíacos/citologia , Miócitos Cardíacos/enzimologia , Paroxetina/química , Paroxetina/farmacologia , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Piridinas/síntese química , Piridinas/farmacologia , Alinhamento de Sequência
3.
J Biol Chem ; 290(17): 10775-90, 2015 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-25770216

RESUMO

G protein-coupled receptor (GPCR) kinases (GRKs) play a key role in homologous desensitization of GPCRs. It is widely assumed that most GRKs selectively phosphorylate only active GPCRs. Here, we show that although this seems to be the case for the GRK2/3 subfamily, GRK5/6 effectively phosphorylate inactive forms of several GPCRs, including ß2-adrenergic and M2 muscarinic receptors, which are commonly used as representative models for GPCRs. Agonist-independent GPCR phosphorylation cannot be explained by constitutive activity of the receptor or membrane association of the GRK, suggesting that it is an inherent ability of GRK5/6. Importantly, phosphorylation of the inactive ß2-adrenergic receptor enhanced its interactions with arrestins. Arrestin-3 was able to discriminate between phosphorylation of the same receptor by GRK2 and GRK5, demonstrating preference for the latter. Arrestin recruitment to inactive phosphorylated GPCRs suggests that not only agonist activation but also the complement of GRKs in the cell regulate formation of the arrestin-receptor complex and thereby G protein-independent signaling.


Assuntos
Quinase 4 de Receptor Acoplado a Proteína G/metabolismo , Quinases de Receptores Acoplados a Proteína G/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Arrestinas/metabolismo , Bovinos , Quinase 4 de Receptor Acoplado a Proteína G/genética , Quinases de Receptores Acoplados a Proteína G/genética , Células HEK293 , Humanos , Mutagênese Sítio-Dirigida , Fosforilação , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
Biochemistry ; 54(28): 4320-9, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26120872

RESUMO

G protein-coupled receptor kinases (GRKs) play an important role in the desensitization of G protein-mediated signaling of G protein-coupled receptors (GPCRs). The level of interest in mapping their phosphorylation sites has increased because recent studies suggest that the differential pattern of receptor phosphorylation has distinct biological consequences. In vitro phosphorylation experiments using well-controlled systems are useful for deciphering the complexity of these physiological reactions and understanding the targeted event. Here, we report on the phosphorylation of the class A GPCR neurotensin receptor 1 (NTSR1) by GRKs under defined experimental conditions afforded by nanodisc technology. Phosphorylation of NTSR1 by GRK2 was agonist-dependent, whereas phosphorylation by GRK5 occurred in an activation-independent manner. In addition, the negatively charged lipids in the immediate vicinity of NTSR1 directly affect phosphorylation by GRKs. Identification of phosphorylation sites in agonist-activated NTSR1 revealed that GRK2 and GRK5 target different residues located on the intracellular receptor elements. GRK2 phosphorylates only the C-terminal Ser residues, whereas GRK5 phosphorylates Ser and Thr residues located in intracellular loop 3 and the C-terminus. Interestingly, phosphorylation assays using a series of NTSR1 mutants show that GRK2 does not require acidic residues upstream of the phospho-acceptors for site-specific phosphorylation, in contrast to the ß2-adrenergic and µ-opioid receptors. Differential phosphorylation of GPCRs by GRKs is thought to encode a particular signaling outcome, and our in vitro study revealed NTSR1 differential phosphorylation by GRK2 and GRK5.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Receptores de Neurotensina/química , Receptores de Neurotensina/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Ratos
5.
EMBO Rep ; 14(6): 520-6, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23579341

RESUMO

We present active-state structures of the G protein-coupled receptor (GPCRs) rhodopsin carrying the disease-causing mutation G90D. Mutations of G90 cause either retinitis pigmentosa (RP) or congenital stationary night blindness (CSNB), a milder, non-progressive form of RP. Our analysis shows that the CSNB-causing G90D mutation introduces a salt bridge with K296. The mutant thus interferes with the E113Q-K296 activation switch and the covalent binding of the inverse agonist 11-cis-retinal, two interactions that are crucial for the deactivation of rhodopsin. Other mutations, including G90V causing RP, cannot promote similar interactions. We discuss our findings in context of a model in which CSNB is caused by constitutive activation of the visual signalling cascade.


Assuntos
Oftalmopatias Hereditárias/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Mutação de Sentido Incorreto , Miopia/genética , Cegueira Noturna/genética , Rodopsina/química , Arrestina/química , Cristalografia por Raios X , Células HEK293 , Humanos , Modelos Moleculares , Ligação Proteica , Estabilidade Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Rodopsina/genética , Bases de Schiff , Homologia Estrutural de Proteína , Temperatura de Transição
6.
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
7.
Molecules ; 19(10): 16937-49, 2014 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-25340299

RESUMO

G protein-coupled receptor kinases (GRKs) have been implicated in human diseases ranging from heart failure to diabetes. Previous studies have identified several compounds that selectively inhibit GRK2, such as paroxetine and balanol. Far fewer selective inhibitors have been reported for GRK5, a target for the treatment of cardiac hypertrophy, and the mechanism of action of reported compounds is unknown. To identify novel scaffolds that selectively inhibit GRK5, a differential scanning fluorometry screen was used to probe a library of 4480 compounds. The best hit was amlexanox, an FDA-approved anti-inflammatory, anti-allergic immunomodulator. The crystal structure of amlexanox in complex with GRK1 demonstrates that its tricyclic aromatic ring system forms ATP-like interactions with the hinge of the kinase domain, which is likely similar to how this drug binds to IκB kinase ε (IKKε), another kinase known to be inhibited by this compound. Amlexanox was also able to inhibit myocyte enhancer factor 2 transcriptional activity in neonatal rat ventricular myocytes in a manner consistent with GRK5 inhibition. The GRK1 amlexanox structure thus serves as a springboard for the rational design of inhibitors with improved potency and selectivity for GRK5 and IKKε.


Assuntos
Aminopiridinas/farmacologia , Antialérgicos/farmacologia , Descoberta de Drogas , Quinase 5 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Aminopiridinas/química , Animais , Animais Recém-Nascidos , Antialérgicos/química , Bovinos , Células Cultivadas , Cristalografia por Raios X , Receptor Quinase 1 Acoplada a Proteína G/química , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Cinética , Modelos Moleculares , Inibidores de Proteínas Quinases/química , Ratos , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
8.
J Am Chem Soc ; 135(13): 5044-51, 2013 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-23461393

RESUMO

The manner in which the heterotrimeric G protein complexes Gß1γ2 and Gαiß1γ2 interact with membranes is likely related to their biological function. We combined complementary measurements from sum frequency generation (SFG) vibrational and attenuated total reflectance Fourier transform infrared (ATR-FTIR) spectroscopy to determine the possible membrane orientations of Gß1γ2 and the Gαiß1γ2 heterotrimer more precisely than could be achieved using SFG alone. The most likely orientations of Gß1γ2 and the Gαiß1γ2 heterotrimer were both determined to fall within a similar narrow range of twist and tilt angles, suggesting that Gß1γ2 may bind to Gαi without a significant change in orientation. This "basal" orientation seems to depend primarily on the geranylgeranylated C-terminus of Gγ2 along with basic residues at the N-terminus of Gαi, and suggests that activated G protein-coupled receptors (GPCRs) must reorient G protein heterotrimers at lipid bilayers to catalyze nucleotide exchange. The innovative methodologies developed in this paper can be widely applied to study the membrane orientation of other proteins in situ.


Assuntos
Bicamadas Lipídicas/química , Receptores Acoplados a Proteínas G/química , Análise Espectral , Proteínas rac de Ligação ao GTP/química , Animais , Modelos Moleculares , Estrutura Secundária de Proteína , Ratos , Espectroscopia de Infravermelho com Transformada de Fourier , Vibração
9.
Mol Imaging Biol ; 25(2): 314-323, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-35906512

RESUMO

PURPOSE: To image colon-expressed alternatively spliced D domain of tenascin C in preclinical colitis models using near infrared (NIR)-labeled targeted molecular imaging agents. PROCEDURES: A human IgG1 with nanomolar binding affinity specific to the alternatively spliced D domain of tenascin C was generated. Immunohistochemistry identified disease-specific expression of this extracellular matrix protein in the colon of mice given dextran sulfate sodium in the drinking water. The antibody reagent was labeled with the NIR fluorophore IRDye 800CW via amine chemistry and intravenously dosed to evaluate in vivo targeting specificity. Increasing doses of imaging agent were given to estimate the saturating dose. RESULTS: The NIR-labeled proteins successfully targeted colonic lesions in a murine model of colitis. Co-administration of a molar excess competing unlabeled dose reduced normalized uptake in diseased colon by > 70%. Near infrared ex vivo images of colon resected from diseased animals showed saturation at doses exceeding 1 nmol and was confirmed with additional quantitative ex vivo biodistribution. Cellular-level specificity and protein stability were assessed via microscopy. CONCLUSIONS: Our imaging data suggest the alternatively spliced D domain of tenascin C is a promising target for delivery-based applications in inflammatory bowel diseases.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Humanos , Animais , Camundongos , Tenascina , Distribuição Tecidual , Colite/patologia
10.
Biochemistry ; 51(16): 3404-11, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22480180

RESUMO

G protein-coupled receptor (GPCR) kinases (GRKs) instigate the desensitization of activated GPCRs via phosphorylation that promotes interaction with arrestins, thereby preventing the interaction of GPCRs with heterotrimeric G proteins. A current proposed model of GRK1 activation involves the binding of activated rhodopsin (Rho*) to the N-terminal region of GRK1. Perhaps concomitantly, this N-terminal region also stabilizes a closed, active conformation of the kinase domain. To further probe this model, we mapped changes in the backbone flexibility of GRK1 as it binds to its two substrates, adenosine triphosphate (Mg(2+)·ATP) and Rho*. We found that the conformational flexibility of GRK1 was reduced in the presence of either Mg(2+)·ATP or Rho*, with Mg(2+)·ATP having the greatest effect. In a truncated form of GRK1 lacking the N-terminal region (ΔN-GRK1), peptides that directly interact with ATP were not as dramatically stabilized by adding Mg(2+)·ATP, and dynamics were greater in the interface between the large lobe of the kinase domain and the regulator of the G protein signaling homology domain. In the presence of Mg(2+)·ATP, the influence of Rho* versus Rho on GRK1 dynamics was negligible.


Assuntos
Receptor Quinase 1 Acoplada a Proteína G/química , Trifosfato de Adenosina/química , Sequência de Aminoácidos , Animais , Bovinos , Células Cultivadas , Receptor Quinase 1 Acoplada a Proteína G/genética , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Conformação Molecular , Simulação de Dinâmica Molecular , Fosforilação , Estrutura Terciária de Proteína , Rodopsina/química , Segmento Externo da Célula Bastonete/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais
11.
Bioorg Med Chem ; 18(14): 5449-56, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20538467

RESUMO

The human cytoplasmic protein tyrosine phosphatase (HCPTP) has been identified as a potential target for inhibition in order to downregulate metastatic transformation in several human epithelial cancers such as breast, prostate and colon cancer. Docking with two scoring functions on both isoforms of HCPTP was employed as an initial virtual screen to identify potential inhibitors. Compounds identified as potential inhibitors via this in silico screen were subjected to kinetic analysis in order to validate their selection as improved inhibitors. Eleven compounds with IC50's of less than 100 microM were identified in a single concentration screen. Five of these compounds were determined to have an IC50 of less than 10 microM; however, all but one of these compounds inhibited via non-specific aggregation. The validated effective inhibitor, which is based on a naphthyl sulfonic acid, strongly resembles a previously synthesized rationally designed azaindole phosphonic acid. This similarity suggests subsequent inhibitor optimization based on this scaffold may generate effective inhibitors of HCPTP. The structural elements of the computationally identified inhibitors are discussed to analyze the combined use of rational design and virtual screening to reduce false negatives in the identification of multiple strong inhibitors of HCPTP.


Assuntos
Desenho de Fármacos , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Tirosina Fosfatases/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Humanos , Concentração Inibidora 50 , Modelos Moleculares
12.
J Med Chem ; 60(7): 3052-3069, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28323425

RESUMO

In heart failure, the ß-adrenergic receptors (ßARs) become desensitized and uncoupled from heterotrimeric G proteins. This process is initiated by G protein-coupled receptor kinases (GRKs), some of which are upregulated in the failing heart, making them desirable therapeutic targets. The selective serotonin reuptake inhibitor, paroxetine, was previously identified as a GRK2 inhibitor. Utilizing a structure-based drug design approach, we modified paroxetine to generate a small compound library. Included in this series is a highly potent and selective GRK2 inhibitor, 14as, with an IC50 of 30 nM against GRK2 and greater than 230-fold selectivity over other GRKs and kinases. Furthermore, 14as showed a 100-fold improvement in cardiomyocyte contractility assays over paroxetine and a plasma concentration higher than its IC50 for over 7 h. Three of these inhibitors, including 14as, were additionally crystallized in complex with GRK2 to give insights into the structural determinants of potency and selectivity of these inhibitors.


Assuntos
Desenho de Fármacos , Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Paroxetina/análogos & derivados , Paroxetina/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Animais , Cristalografia por Raios X , Quinase 2 de Receptor Acoplado a Proteína G/química , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Humanos , Camundongos , Microssomos Hepáticos/metabolismo , Simulação de Acoplamento Molecular , Paroxetina/sangue , Paroxetina/metabolismo , Inibidores de Proteínas Quinases/sangue , Inibidores de Proteínas Quinases/metabolismo
13.
J Med Chem ; 59(8): 3793-807, 2016 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-27050625

RESUMO

G protein-coupled receptors (GPCRs) are central to many physiological processes. Regulation of this superfamily of receptors is controlled by GPCR kinases (GRKs), some of which have been implicated in heart failure. GSK180736A, developed as a Rho-associated coiled-coil kinase 1 (ROCK1) inhibitor, was identified as an inhibitor of GRK2 and co-crystallized in the active site. Guided by its binding pose overlaid with the binding pose of a known potent GRK2 inhibitor, Takeda103A, a library of hybrid inhibitors was developed. This campaign produced several compounds possessing high potency and selectivity for GRK2 over other GRK subfamilies, PKA, and ROCK1. The most selective compound, 12n (CCG-224406), had an IC50 for GRK2 of 130 nM, >700-fold selectivity over other GRK subfamilies, and no detectable inhibition of ROCK1. Four of the new inhibitors were crystallized with GRK2 to give molecular insights into the binding and kinase selectivity of this class of inhibitors.


Assuntos
Inibidores de Proteínas Quinases/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Animais , Bovinos , Células Cultivadas , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Conformação Proteica , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Relação Estrutura-Atividade , Quinases Associadas a rho/antagonistas & inibidores
14.
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
15.
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
16.
Elife ; 42015 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-25910054

RESUMO

Rod photoreceptors generate measurable responses to single-photon activation of individual molecules of the G protein-coupled receptor (GPCR), rhodopsin. Timely rhodopsin desensitization depends on phosphorylation and arrestin binding, which quenches G protein activation. Rhodopsin phosphorylation has been measured biochemically at C-terminal serine residues, suggesting that these residues are critical for producing fast, low-noise responses. The role of native threonine residues is unclear. We compared single-photon responses from rhodopsin lacking native serine or threonine phosphorylation sites. Contrary to expectation, serine-only rhodopsin generated prolonged step-like single-photon responses that terminated abruptly and randomly, whereas threonine-only rhodopsin generated responses that were only modestly slower than normal. We show that the step-like responses of serine-only rhodopsin reflect slow and stochastic arrestin binding. Thus, threonine sites play a privileged role in promoting timely arrestin binding and rhodopsin desensitization. Similar coordination of phosphorylation and arrestin binding may more generally permit tight control of the duration of GPCR activity.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Rodopsina/metabolismo , Serina/metabolismo , Treonina/metabolismo , Animais , Arrestina/metabolismo , Sítios de Ligação/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Células Fotorreceptoras Retinianas Bastonetes/citologia , Rodopsina/genética
17.
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
19.
Cell Signal ; 25(11): 2155-62, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23872075

RESUMO

The effects of activating mutations associated with night blindness on the stoichiometry of rhodopsin interactions with G protein-coupled receptor kinase 1 (GRK1) and arrestin-1 have not been reported. Here we show that the monomeric form of WT rhodopsin and its constitutively active mutants M257Y, G90D, and T94I, reconstituted into HDL particles are effectively phosphorylated by GRK1, as well as two more ubiquitously expressed subtypes, GRK2 and GRK5. All versions of arrestin-1 tested (WT, pre-activated, and constitutively monomeric mutants) bind to monomeric rhodopsin and show the same selectivity for different functional forms of rhodopsin as in native disc membranes. Rhodopsin phosphorylation by GRK1 and GRK2 promotes arrestin-1 binding to a comparable extent, whereas similar phosphorylation by GRK5 is less effective, suggesting that not all phosphorylation sites on rhodopsin are equivalent in promoting arrestin-1 binding. The binding of WT arrestin-1 to phospho-opsin is comparable to the binding to its preferred target, P-Rh*, suggesting that in photoreceptors arrestin-1 only dissociates after opsin regeneration with 11-cis-retinal, which converts phospho-opsin into inactive phospho-rhodopsin that has lower affinity for arrestin-1. Reduced binding of arrestin-1 to the phospho-opsin form of G90D mutant likely contributes to night blindness caused by this mutation in humans.


Assuntos
Arrestina/genética , Receptor Quinase 1 Acoplada a Proteína G/genética , Mutação , Cegueira Noturna/genética , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Rodopsina/genética , Animais , Arrestina/metabolismo , Bovinos , HDL-Colesterol/química , HDL-Colesterol/metabolismo , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Regulação da Expressão Gênica , Isoenzimas/genética , Isoenzimas/metabolismo , Cegueira Noturna/metabolismo , Cegueira Noturna/patologia , Opsinas/genética , Opsinas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerização Proteica , Células Fotorreceptoras Retinianas Bastonetes/patologia , Rodopsina/metabolismo , Transdução de Sinais
20.
ACS Chem Biol ; 7(11): 1830-9, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22882301

RESUMO

G protein-coupled receptor kinase 2 (GRK2) is a well-established therapeutic target for the treatment of heart failure. Herein we identify the selective serotonin reuptake inhibitor (SSRI) paroxetine as a selective inhibitor of GRK2 activity both in vitro and in living cells. In the crystal structure of the GRK2·paroxetine-Gßγ complex, paroxetine binds in the active site of GRK2 and stabilizes the kinase domain in a novel conformation in which a unique regulatory loop forms part of the ligand binding site. Isolated cardiomyocytes show increased isoproterenol-induced shortening and contraction amplitude in the presence of paroxetine, and pretreatment of mice with paroxetine before isoproterenol significantly increases left ventricular inotropic reserve in vivo with no significant effect on heart rate. Neither is observed in the presence of the SSRI fluoxetine. Our structural and functional results validate a widely available drug as a selective chemical probe for GRK2 and represent a starting point for the rational design of more potent and specific GRK2 inhibitors.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Coração/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Paroxetina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Domínio Catalítico/efeitos dos fármacos , Células Cultivadas , Quinase 2 de Receptor Acoplado a Proteína G/química , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Coração/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Fosforilação/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína/efeitos dos fármacos , Hormônio Liberador de Tireotropina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA