Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Cell ; 33(1): 43-52, 2009 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-19150426

RESUMO

The glycine-rich G loop controls ATP binding and phosphate transfer in protein kinases. Here we show that the functions of Src family and Abl protein tyrosine kinases require an electrostatic interaction between oppositely charged amino acids within their G loops that is conserved in multiple other phylogenetically distinct protein kinases, from plants to humans. By limiting G loop flexibility, it controls ATP binding, catalysis, and inhibition by ATP-competitive compounds such as Imatinib. In WeeB mice, mutational disruption of the interaction results in expression of a Lyn protein with reduced catalytic activity, and in perturbed B cell receptor signaling. Like Lyn(-/-) mice, WeeB mice show profound defects in B cell development and function and succumb to autoimmune glomerulonephritis. This demonstrates the physiological importance of the conserved G loop salt bridge and at the same time distinguishes the in vivo requirement for the Lyn kinase activity from other potential functions of the protein.


Assuntos
Biocatálise , Sequência Conservada , Proteínas Quinases/química , Eletricidade Estática , Quinases da Família src/química , Quinases da Família src/metabolismo , Sequência de Aminoácidos , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/enzimologia , Benzamidas , Biocatálise/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática/efeitos dos fármacos , Proteínas de Fusão bcr-abl/metabolismo , Mesilato de Imatinib , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Mutação/genética , Filogenia , Piperazinas/farmacologia , Estabilidade Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Pirimidinas/farmacologia , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais/efeitos dos fármacos
2.
Bioorg Med Chem Lett ; 25(20): 4642-7, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26320624

RESUMO

Design and optimization of benzo- and pyrido-thiazoles/isothiazoles are reported leading to the discovery of the potent, orally bioavailable Syk inhibitor 5, which was found to be active in a rat PK/PD model. Compound 5 showed acceptable overall kinase selectivity. However, in addition to Syk it also inhibited Aurora kinase in enzymatic and cellular settings leading to findings in the micronucleus assay. As a consequence, compound 5 was not further pursued.


Assuntos
Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Tiazóis/farmacologia , Administração Oral , Animais , Disponibilidade Biológica , Relação Dose-Resposta a Droga , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Estrutura Molecular , Inibidores de Proteínas Quinases/química , Proteínas Tirosina Quinases/metabolismo , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Quinase Syk , Tiazóis/administração & dosagem , Tiazóis/química
4.
Biochem J ; 430(3): 425-37, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20632993

RESUMO

ALK (anaplastic lymphoma kinase) is an RTK (receptor tyrosine kinase) of the IRK (insulin receptor kinase) superfamily, which share an YXXXYY autophosphorylation motif within their A-loops (activation loops). A common activation and regulatory mechanism is believed to exist for members of this superfamily typified by IRK and IGF1RK (insulin-like growth factor receptor kinase-1). Chromosomal translocations involving ALK were first identified in anaplastic large-cell lymphoma, a subtype of non-Hodgkin's lymphoma, where aberrant fusion of the ALK kinase domain with the NPM (nucleophosmin) dimerization domain results in autophosphosphorylation and ligand-independent activation. Activating mutations within the full-length ALK kinase domain, most commonly R1275Q and F1174L, which play a major role in neuroblastoma, were recently identified. To provide a structural framework for understanding these mutations and to guide structure-assisted drug discovery efforts, the X-ray crystal structure of the unphosphorylated ALK catalytic domain was determined in the apo, ADP- and staurosporine-bound forms. The structures reveal a partially inactive protein kinase conformation distinct from, and lacking, many of the negative regulatory features observed in inactive IGF1RK/IRK structures in their unphosphorylated forms. The A-loop adopts an inhibitory pose where a short proximal A-loop helix (alphaAL) packs against the alphaC helix and a novel N-terminal beta-turn motif, whereas the distal portion obstructs part of the predicted peptide-binding region. The structure helps explain the reported unique peptide substrate specificity and the importance of phosphorylation of the first A-loop Tyr1278 for kinase activity and NPM-ALK transforming potential. A single amino acid difference in the ALK substrate peptide binding P-1 site (where the P-site is the phosphoacceptor site) was identified that, in conjunction with A-loop sequence variation including the RAS (Arg-Ala-Ser)-motif, rationalizes the difference in the A-loop tyrosine autophosphorylation preference between ALK and IGF1RK/IRK. Enzymatic analysis of recombinant R1275Q and F1174L ALK mutant catalytic domains confirms the enhanced activity and transforming potential of these mutants. The transforming ability of the full-length ALK mutants in soft agar colony growth assays corroborates these findings. The availability of a three-dimensional structure for ALK will facilitate future structure-function and rational drug design efforts targeting this receptor tyrosine kinase.


Assuntos
Domínio Catalítico , Proteínas Mutantes/química , Conformação Proteica , Proteínas Tirosina Quinases/química , Difosfato de Adenosina/química , Difosfato de Adenosina/metabolismo , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Quinase do Linfoma Anaplásico , Animais , Linhagem Celular , Cristalização , Cristalografia por Raios X , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese , Proteínas Mutantes/metabolismo , Neuroblastoma/enzimologia , Neuroblastoma/genética , Fosforilação , Ligação Proteica , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Receptores Proteína Tirosina Quinases , Homologia de Sequência de Aminoácidos , Spodoptera , Estaurosporina/química , Estaurosporina/metabolismo , Especificidade por Substrato
5.
Biophys J ; 97(1): 110-20, 2009 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-19580749

RESUMO

Voltage-gated K+ channels share a common voltage sensor domain (VSD) consisting of four transmembrane helices, including a highly mobile S4 helix that contains the major gating charges. Activation of ether-a-go-go (EAG) family K+ channels is sensitive to external divalent cations. We show here that divalent cations slow the activation rate of two EAG family channels (Kv12.1 and Kv10.2) by forming a bridge between a residue in the S4 helix and acidic residues in S2. Histidine 328 in the S4 of Kv12.1 favors binding of Zn2+ and Cd2+, whereas the homologous residue Serine 321 in Kv10.2 contributes to effects of Mg2+ and Ni2+. This novel finding provides structural constraints for the position of transmembrane VSD helices in closed, ion-bound EAG family channels. Homology models of Kv12.1 and Kv10.2 VSD structures based on a closed-state model of the Shaker family K+ channel Kv1.2 match these constraints. Our results suggest close conformational conservation between closed EAG and Shaker family channels, despite large differences in voltage sensitivity, activation rates, and activation thresholds.


Assuntos
Cátions Bivalentes/metabolismo , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/metabolismo , Sequência de Aminoácidos , Animais , Cádmio/metabolismo , Canais de Potássio Éter-A-Go-Go/genética , Canal de Potássio Kv1.2/metabolismo , Magnésio/metabolismo , Potenciais da Membrana/fisiologia , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Níquel/metabolismo , Técnicas de Patch-Clamp , Conformação Proteica , Homologia de Sequência de Aminoácidos , Xenopus , Zinco/metabolismo
6.
Structure ; 11(1): 99-108, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12517344

RESUMO

Neuropilin-1 (Npn-1) is a type I cell surface receptor involved in a broad range of developmental processes, including axon guidance, angiogenesis, and heterophilic cell adhesion. We have determined the crystal structure of the human Npn-1 b1 domain to 1.9 A. The overall structure resembles coagulation factor V and VIII (F5/8) C1 and C2 domains, exhibiting a distorted jellyroll fold. Details of the structure provide insight to b1 domain regions responsible for ligand binding and facilitate rationalization of existing biochemical binding data. A polar cleft formed by adjacent loops at one end of the molecule in conjunction with flanking electronegative surfaces may represent the binding site for the positively charged tails of semaphorins and VEGF(165). The nature of the cell adhesion binding site of the b1 domain can be visualized in context of the structure.


Assuntos
Neuropilina-1/química , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Sítios de Ligação , Fatores de Coagulação Sanguínea/química , Fatores de Coagulação Sanguínea/genética , Adesão Celular/fisiologia , Cristalografia por Raios X , Humanos , Dados de Sequência Molecular , Neovascularização Fisiológica , Neuropilina-1/genética , Neuropilina-1/metabolismo , Dobramento de Proteína , Alinhamento de Sequência , Propriedades de Superfície
7.
Structure ; 12(3): 487-94, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15016364

RESUMO

AvrB is a Pseudomonas syringae type III effector protein that is translocated into host plant cells during attempted pathogenesis. Arabidopsis harboring the corresponding resistance protein RPM1 can detect AvrB and mount a rapid host defense response, thus avoiding active infection. In the plant cell, AvrB induces phosphorylation of RIN4, a key component in AvrB/RPM1 recognition. Although the AvrB/RPM1 system is among the best characterized of the numerous bacterial effector/plant resistance protein systems involved in plant disease resistance and pathogenesis, the details of the molecular recognition mechanism are still unclear. To gain further insights, the crystal structure of AvrB was determined. The 2.2 A structure exhibits a novel mixed alpha/beta bilobal fold. Aided by the structural information, we demonstrate that one lobe is the determinant of AvrB/RPM1 recognition specificity. This structural information and preliminary structure-function studies provide a framework for the future understanding of AvrB function on the molecular level.


Assuntos
Proteínas de Bactérias/química , Sequência de Aminoácidos , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Modelos Moleculares , Dados de Sequência Molecular , Dobramento de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Pseudomonas syringae/química , Pseudomonas syringae/genética , Pseudomonas syringae/metabolismo , Alinhamento de Sequência
8.
J Med Chem ; 58(4): 1950-63, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25633741
9.
Mol Biosyst ; 10(6): 1490-5, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24675991

RESUMO

Anaplastic Lymphoma Kinase (ALK) plays a major role in developing tumor processes and therefore has emerged as a validated therapeutic target. Applying atomistic molecular dynamics simulations on the wild type enzyme and the nine most frequently occurring and clinically important activation mutants we revealed important conformational effects on key interactions responsible for the activation of the enzyme.


Assuntos
Domínio Catalítico , Conformação Proteica , Receptores Proteína Tirosina Quinases/química , Quinase do Linfoma Anaplásico , Biologia Computacional , Cristalografia por Raios X , Humanos , Simulação de Dinâmica Molecular , Mutação , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores Proteína Tirosina Quinases/genética
10.
Cancer Discov ; 4(6): 662-673, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24675041

RESUMO

UNLABELLED: Non-small cell lung cancers (NSCLC) harboring anaplastic lymphoma kinase (ALK) gene rearrangements invariably develop resistance to the ALK tyrosine kinase inhibitor (TKI) crizotinib. Herein, we report the first preclinical evaluation of the next-generation ALK TKI, ceritinib (LDK378), in the setting of crizotinib resistance. An interrogation of in vitro and in vivo models of acquired resistance to crizotinib, including cell lines established from biopsies of patients with crizotinib-resistant NSCLC, revealed that ceritinib potently overcomes crizotinib-resistant mutations. In particular, ceritinib effectively inhibits ALK harboring L1196M, G1269A, I1171T, and S1206Y mutations, and a cocrystal structure of ceritinib bound to ALK provides structural bases for this increased potency. However, we observed that ceritinib did not overcome two crizotinib-resistant ALK mutations, G1202R and F1174C, and one of these mutations was identified in 5 of 11 biopsies from patients with acquired resistance to ceritinib. Altogether, our results demonstrate that ceritinib can overcome crizotinib resistance, consistent with clinical data showing marked efficacy of ceritinib in patients with crizotinib-resistant disease. SIGNIFICANCE: The second-generation ALK inhibitor ceritinib can overcome several crizotinib-resistant mutations and is potent against several in vitro and in vivo laboratory models of acquired resistance to crizotinib. These findings provide the molecular basis for the marked clinical activity of ceritinib in patients with ALK-positive NSCLC with crizotinib-resistant disease. Cancer Discov; 4(6); 662-73. ©2014 AACR. See related commentary by Ramalingam and Khuri, p. 634 This article is highlighted in the In This Issue feature, p. 621.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Sulfonas/uso terapêutico , Quinase do Linfoma Anaplásico , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Crizotinibe , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos SCID , Mutação , Pirazóis/uso terapêutico , Piridinas/uso terapêutico , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Carga Tumoral
11.
J Med Chem ; 56(14): 5675-90, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23742252

RESUMO

The synthesis, preclinical profile, and in vivo efficacy in rat xenograft models of the novel and selective anaplastic lymphoma kinase inhibitor 15b (LDK378) are described. In this initial report, preliminary structure-activity relationships (SARs) are described as well as the rational design strategy employed to overcome the development deficiencies of the first generation ALK inhibitor 4 (TAE684). Compound 15b is currently in phase 1 and phase 2 clinical trials with substantial antitumor activity being observed in ALK-positive cancer patients.


Assuntos
Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/síntese química , Pirimidinas/síntese química , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Sulfonas/síntese química , Quinase do Linfoma Anaplásico , Animais , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Cães , Humanos , Macaca fascicularis , Masculino , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/farmacocinética , Pirimidinas/uso terapêutico , Ratos , Relação Estrutura-Atividade , Sulfonas/farmacocinética , Sulfonas/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Bioorg Med Chem Lett ; 16(10): 2689-92, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16524731

RESUMO

A series of 2-amino-9-aryl-7H-pyrrolo[2,3-d]pyrimidines were designed and synthesized as focal adhesion kinase (FAK) inhibitors using molecular modeling in conjunction with a co-crystal structure. Chemistry was developed to introduce functionality onto the 9-aryl ring, which resulted in the identification of potent FAK inhibitors. In particular, compound 32 possessed single-digit nanomolar IC(50) and represents one of the most potent FAK inhibitors discovered to date.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/síntese química , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Pirimidinas/química , Pirimidinas/síntese química , Inibidores Enzimáticos/farmacologia , Estrutura Molecular , Pirimidinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA