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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Curr Opin Chem Biol ; 5(4): 409-15, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11470604

RESUMO

The past two years have witnessed a number of significant advances in the design of SH2 inhibitors of both Src and Grb2. For Src, several non-peptide templates have been developed with high affinity, and one case, in the context of bone-binding phosphotyrosine bioisostere, has yielded an in vivo active antiresorptive agent. Similarly, high-affinity Grb2 SH2 inhibitors with novel phosphotyrosine replacements have now been reported that demonstrate, for the first time, cellular activities consistent with an anticancer agent.


Assuntos
Domínios de Homologia de src , Modelos Moleculares , Conformação Proteica
2.
Chem Biol ; 5(10): 529-38, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9818146

RESUMO

BACKGROUND: Many intracellular signal-transduction pathways are regulated by specific protein-protein interactions. These interactions are mediated by structural domains within signaling proteins that modulate a protein's cellular location, stability or activity. For example, Src-homology 2 (SH2) domains mediate protein-protein interactions through short contiguous amino acid motifs containing phosphotyrosine. As SH2 domains have been recognized as key regulatory molecules in a variety of cellular processes, they have become attractive drug targets. RESULTS: We have developed a novel mechanism-based cellular assay to monitor specific SH2-domain-dependent protein-protein interactions. The assay is based on a two-hybrid system adapted to function in mammalian cells where the SH2 domain ligand is phosphorylated, and binding to a specific SH2 domain can be induced and easily monitored. As examples, we have generated a series of mammalian cell lines that can be used to monitor SH2-domain-dependent activity of the signaling proteins ZAP-70 and Src. We are utilizing these cell lines to screen for immunosuppressive and anti-osteoclastic compounds, respectively, and demonstrate here the utility of this system for the identification of small-molecule, cell-permeant SH2 domain inhibitors. CONCLUSIONS: A mechanism-based mammalian cell assay has been developed to identify inhibitors of SH2-domain-dependent protein-protein interactions. Mechanism-based assays similar to that described here might have general use as screens for cell-permeant, nontoxic inhibitors of protein-protein interactions.


Assuntos
Proteínas/antagonistas & inibidores , Domínios de Homologia de src , Animais , Humanos , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/metabolismo , Proteínas/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Proteína-Tirosina Quinase ZAP-70
3.
Chem Biol ; 7(3): 225-35, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10712930

RESUMO

BACKGROUND: The observations that Src(-/-) mice develop osteopetrosis and Src family tyrosine kinase inhibitors decrease osteoclast-mediated resorption of bone have implicated Src in the regulation of osteoclast-resorptive activity. We have designed and synthesized a compound, AP22161, that binds selectively to the Src SH2 domain and demonstrated that it inhibits Src-dependent cellular activity and inhibits osteoclast-mediated resorption. RESULTS: AP22161 was designed to bind selectively to the Src SH2 domain by targeting a cysteine residue within the highly conserved phosphotyrosine-binding pocket. AP22161 was tested in vitro for binding to SH2 domains and was found to bind selectively and with high affinity to the Src SH2 domain. AP22161 was further tested in mechanism-based cellular assays and found to block Src SH2 binding to peptide ligands, inhibit Src-dependent cellular activity and diminish osteoclast resorptive activity. CONCLUSIONS: These results indicate that a compound that selectively inhibits Src SH2 binding can be used to inhibit osteoclast resorption. Furthermore, AP22161 has the potential to be further developed for treating osteoporosis.


Assuntos
Benzoatos/farmacologia , Reabsorção Óssea/prevenção & controle , Inibidores Enzimáticos/farmacologia , Osteoclastos/efeitos dos fármacos , Quinases da Família src/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Benzoatos/síntese química , Benzoatos/química , Sítios de Ligação/genética , Reabsorção Óssea/etiologia , Reabsorção Óssea/fisiopatologia , Células Cultivadas , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Técnicas In Vitro , Ligantes , Camundongos , Dados de Sequência Molecular , Osteoclastos/fisiologia , Coelhos , Ratos , Homologia de Sequência de Aminoácidos , Transformação Genética , Técnicas do Sistema de Duplo-Híbrido , Domínios de Homologia de src/genética , Quinases da Família src/genética , Quinases da Família src/metabolismo
4.
Med Chem ; 1(3): 293-319, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-16787325

RESUMO

Protein phosphorylation has been exploited by Nature in profound ways to control various aspects of cell proliferation, differentiation, metabolism, survival, motility and gene transcription. Cellular signal transduction pathways involve protein kinases, protein phosphatases, and phosphoprotein-interacting domain (e.g., SH2, PTB, WW, FHA, 14-3-3) containing cellular proteins to provide multidimensional, dynamic and reversible regulation of many biological activities. Knowledge of cellular signal transduction pathways has led to the identification of promising therapeutic targets amongst these superfamilies of enzymes and adapter proteins which have been linked to various cancers as well as inflammatory, immune, metabolic and bone diseases. This review focuses on protein kinase, protein phosphatase and phosphoprotein-interacting cellular protein therapeutic targets with an emphasis on small-molecule drug discovery from a chemistry perspective. Noteworthy studies related to molecular genetics, signal transduction pathways, structural biology, and drug design for several of these therapeutic targets are highlighted. Some exemplary proof-of-concept lead compounds, clinical candidates and/or breakthrough medicines are further detailed to illustrate achievements as well as challenges in the generation, optimization and development of small-molecule inhibitors of protein kinases, protein phosphatases or phosphoprotein-interacting domain containing cellular proteins.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/química , Fosfoproteínas Fosfatases/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Proteínas Quinases/efeitos dos fármacos , Proteínas/metabolismo , Inibidores Enzimáticos/farmacologia , Fosfoproteínas Fosfatases/química , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/química , Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína , Transdução de Sinais , Relação Estrutura-Atividade
5.
J Cereb Blood Flow Metab ; 16(4): 599-604, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8964798

RESUMO

We tested the effects of administration of a selective neuronal nitric oxide synthase (nNOS) inhibitor, ARL 17477, on ischemic cell damage and regional cerebral blood flow (rCBF), in rats subjected to transient (2 h) middle cerebral artery (MCA) occlusion and 166 h of reperfusion (n = 48) and in rats without MCA occlusion (n = 25), respectively. Animals were administered ARL 17477 (i.v.): 10 mg/kg; 1 mg/kg; 3mg/kg; N-nitro-L-arginine (L-NA) 10 mg/kg L-NA 1 mg/kg; and Vehicle. Administration of ARL 17477 1 mg/kg, 3 mg/kg and 10 mg/kg reduced ischemic infarct volume by 53 (p < 0.05), 23, and 6.5%, respectively. L-NA 1 mg/kg and 10 mg/kg increased infarct volume by 2 and 15%, respectively (p > 0.05). Administration of ARL 17477 (10 mg/kg) significantly (p < 0.05) decreased rCBF by 27 +/- 5.3 and 24 +/- 14.08% and cortical NOS activity by 86 +/- 14.9 and 91 +/- 8.9% at 10 min or 3 h, respectively, and did not alter mean arterial blood pressure (MABP). L-NA (10 mg/kg) significantly reduced rCBF by 23 +/- 9.8% and NOS activity by 81 +/- 7% and significantly (p < 0.05) increased MABP. Treatment with 3 mg/kg and 1 mg/kg ARL 17477 reduced rCBF by only 2.4 +/- 4.5 and 0%, respectively, even when NOS activity was reduced by 63 +/- 13.4 and 45 +/- 15.7% at 3 h, respectively, (p < 0.05). The data demonstrate that ARL 17477 inhibits nNOS in the rat brain and causes a dose-dependent reduction in infarct volume after transient MCA occlusion.


Assuntos
Amidinas/farmacologia , Arteriopatias Oclusivas/patologia , Artérias Cerebrais , Infarto Cerebral/patologia , Inibidores Enzimáticos/farmacologia , Neurônios/enzimologia , Óxido Nítrico Sintase/antagonistas & inibidores , Animais , Arteriopatias Oclusivas/fisiopatologia , Pressão Sanguínea , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Infarto Cerebral/fisiopatologia , Circulação Cerebrovascular/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Reperfusão
6.
Bone ; 28(1): 54-64, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11165943

RESUMO

Src, a nonreceptor tyrosine kinase, is an important regulator of osteoclast-mediated resorption. We have investigated whether compounds that bind to the Src SH2 domain inhibit Src activity in cells and decrease osteoclast-mediated resorption. Compounds were examined for binding to the Src SH2 domain in vitro using a fluorescence polarization binding assay. Experiments were carried out with compounds demonstrating in vitro binding activity (nmol/L range) to determine if they inhibit Src SH2 binding and Src function in cells, demonstrate blockade of Src signaling, and lack cellular toxicity. Cell-based assays included: (1) a mammalian two-hybrid assay; (2) morphological reversion and growth inhibition of cSrcY527F-transformed cells; and (3) inhibition of cortactin phosphorylation in csk-/- cells. The Src SH2 binding compounds inhibit Src activity in all three of these mechanism-based assays. The compounds described were synthesized to contain nonhydrolyzable phosphotyrosine mimics that bind to bone. These compounds were further tested and found to inhibit rabbit osteoclast-mediated resorption of dentine. These results indicate that compounds that bind to the Src SH2 domain can inhibit Src activity in cells and inhibit osteoclast-mediated resorption.


Assuntos
Reabsorção Óssea/metabolismo , Difosfonatos/metabolismo , Osteoclastos/metabolismo , Domínios de Homologia de src/fisiologia , Quinases da Família src/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Transformada , Dentina/metabolismo , Difosfonatos/química , Difosfonatos/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Humanos , Ligantes , Mamíferos , Camundongos , Dados de Sequência Molecular , Osteoclastos/citologia , Osteoporose/metabolismo , Coelhos , Ensaio Radioligante , Ratos , Trítio , Técnicas do Sistema de Duplo-Híbrido , Quinases da Família src/antagonistas & inibidores
7.
J Med Chem ; 41(9): 1382-91, 1998 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-9554871

RESUMO

We show that carbohydrates constitute an attractive source of readily available, stereochemically defined scaffolds for the facile attachment of side chains contained in genetically encoded and other amino acids. beta-D- and beta-L-glucose, L-mannose, and the 6-deoxy-6-N-analogue of beta-D-glucose have been employed to synthesize peptidomimetics that bind the SRIF receptors on AtT-20 mouse pituitary cells, five cloned human receptor subtypes (hSSTRs), and the NK-1 receptor. The affinity profile of various sugar-based ligands at the hSSTRs is compared with that of SRIF. Compound 19 bound hSSTR4 with a Ki of 100 nM. Subtle structural changes affect affinities. Evidence is presented that suggests that one compound (8) binds both the AtT-20 cell receptors and the five hSSTRs via a unique mode. The SARs of the glycosides at SRIF receptors differ markedly from those at the NK-1 receptor. For example a 4-benzyl substituent is important for SRIF receptor binding, but the 4-desbenzyl analogue 27 was highly potent (IC50 of 27 nM) at the NK-1 receptor. A new, nonbasic method for the synthesis of base-sensitive ethers from primary and secondary alcohols is also described.


Assuntos
Éteres/metabolismo , Glucosídeos/metabolismo , Mimetismo Molecular , Monossacarídeos/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Somatostatina/agonistas , Animais , Células CHO , Linhagem Celular , Cricetinae , Éteres/síntese química , Éteres/química , Glucosídeos/síntese química , Glucosídeos/química , Humanos , Ligantes , Lisina/metabolismo , Camundongos , Modelos Moleculares , Monossacarídeos/síntese química , Monossacarídeos/química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Hipófise/citologia , Hipófise/metabolismo , Receptores da Neurocinina-1/metabolismo , Somatostatina/análogos & derivados , Somatostatina/química , Somatostatina/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade
10.
Br J Cancer ; 94(11): 1710-7, 2006 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-16685275

RESUMO

Vascular endothelial growth factor (VEGF) is the predominant pro-angiogenic cytokine in human malignancy, and its expression correlates with disease recurrence and poor outcomes in patients with colorectal cancer. Recently, expression of vascular endothelial growth factor receptors (VEGFRs) has been observed on tumours of epithelial origin, including those arising in the colon, but the molecular mechanisms governing potential VEGF-driven biologic functioning in these tumours are not well characterised. In this report, we investigated the role of Src family kinases (SFKs) in VEGF-mediated signalling in human colorectal carcinoma (CRC) cell lines. Vascular endothelial growth factor specifically activated SFKs in HT29 and KM12L4 CRC cell lines. Further, VEGF stimulation resulted in enhanced cellular migration, which was effectively blocked by pharmacologic inhibition of VEGFR-1 or Src kinase. Correspondingly, migration studies using siRNA clones with reduced Src expression confirmed the requirement for Src in VEGF-induced migration in these cells. Furthermore, VEGF treatment enhanced VEGFR-1/SFK complex formation and increased tyrosine phosphorylation of focal adhesion kinase, p130 cas and paxillin. Finally, we demonstrate that VEGF-induced migration is not due, at least in part, to VEGF acting as a mitogen. These results suggest that VEGFR-1 promotes migration of tumour cells through a Src-dependent pathway linked to activation of focal adhesion components that regulate this process.


Assuntos
Adenocarcinoma/fisiopatologia , Movimento Celular/fisiologia , Neoplasias do Colo/fisiopatologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Quinases da Família src/genética , Quinases da Família src/metabolismo , Adenocarcinoma/enzimologia , Linhagem Celular Tumoral , Neoplasias do Colo/enzimologia , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Immunoblotting , Plasmídeos , RNA Interferente Pequeno/genética
11.
Curr Opin Drug Discov Devel ; 3(5): 549-64, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19649883

RESUMO

Signal transduction targets include catalytic and/or non-catalytic domains, which are critical to various aspects of cell growth, differentiation, metabolism and function, mitogenesis, motility and gene transcription. Specific examples of molecular targets include the catalytic domains of protein tyrosine kinases (PTKs) and of protein tyrosine phosphatases (PTPases), as well as related protein-protein interaction motifs (eg, SH2, PTB and SH3 domains). From the relationship of tyrosine phosphorylation to intracellular pathway regulation by PTKs and PTPases, the dynamic and reversible binding interactions of SH2 and PTB domain-containing proteins with their cognate phosphotyrosine (pTyr)-containing proteins, provide an additional dimension to the modulation of signal transduction pathways which exist as a result of pTyr formation, degradation and molecular recognition events. This review focuses on our current understanding of key relative to recent reports which have provided further insight into their three-dimensional structure and mechanism. This review also highlights recent progress in the design and optimization of molecular mechanism-based signal transduction inhibitors.

12.
Bioorg Med Chem Lett ; 9(21): 3109-12, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10560734

RESUMO

A CuBr-mediated, regioselective cross-coupling between methyl 2,5-diiodobenzoate (4) and [(diethoxyphosphinyl)difluoromethyl]zinc bromide is reported. Palladium-catalyzed incorporation of an amino acid side chain, followed by subsequent modifications resulted in the rapid construction of 2. Compound 2 was designed to engage Cys188 of the Src SH2 domain, however, this was not observed spectroscopically.


Assuntos
Compostos de Flúor/síntese química , Organofosfonatos/síntese química , Fenilalanina/análogos & derivados , Domínios de Homologia de src , Sítios de Ligação , Cisteína/química , Compostos de Flúor/farmacologia , Ligantes , Estrutura Molecular , Organofosfonatos/farmacologia , Ligação Proteica , Proteínas Tirosina Quinases/química
13.
Bioorg Med Chem Lett ; 11(17): 2319-23, 2001 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-11527723

RESUMO

The novel phosphotyrosine (pTyr) mimetic 4'-carboxymethyloxy-3'-phosphonophenylalanine (Cpp) has been designed and incorporated into a series of nonpeptide inhibitors of the SH2 domain of pp60(c-Src) (Src) tyrosine kinase. A 2.2 A X-ray crystal structure of 1a bound to a mutant form of Lck SH2 domain provides insight regarding the structure-activity relationships and supports the design concept of this new pTyr mimetic.


Assuntos
Benzamidas/química , Benzamidas/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Organofosfonatos/química , Organofosfonatos/farmacologia , Fosfotirosina/química , Proteínas Proto-Oncogênicas pp60(c-src)/antagonistas & inibidores , Domínios de Homologia de src/efeitos dos fármacos , Animais , Sítios de Ligação , Reabsorção Óssea , Cristalografia por Raios X , Dentina/efeitos dos fármacos , Desenho de Fármacos , Inibidores Enzimáticos/metabolismo , Espectroscopia de Ressonância Magnética , Mimetismo Molecular , Mutação , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/química , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Coelhos , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA