Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Nat Cell Biol ; 2(10): 677-85, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11025657

RESUMO

The p21-activated kinases (PAKs) are effectors for the Rho-family GTPase Cdc42p. Here we define the in vivo function of the kinase activity of the budding yeast PAK Cla4p, using cla4 alleles that are specifically inhibited by a cell-permeable compound that does not inhibit the wild-type kinase. CLA4 kinase inhibition in cells lacking the partially redundant PAK Ste20p causes reversible SWE1-dependent cell-cycle arrest and gives rise to narrow, highly elongated buds in which both actin and septin are tightly polarized to bud tips. Inhibition of Cla4p does not prevent polarization of F-actin, and cytokinesis is blocked only in cells that have not formed a bud before inhibitor treatment; cell polarization and bud emergence are not affected by Cla4p inhibition. Although localization of septin to bud necks is restored in swe1Delta cells, cytokinesis remains defective. Inhibition of Cla4p activity in swe1Delta cells causes a delay of bud emergence after cell polarization, indicating that this checkpoint may mediate an adaptive response that is capable of promoting budding when Cla4p function is reduced. Our data indicate that CLA4 PAK activity is required at an early stage of budding, after actin polarization and coincident with formation of the septin ring, for early bud morphogenesis and assembly of a cytokinesis site.


Assuntos
Ciclinas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae , Saccharomycetales/citologia , Alelos , Sequência de Aminoácidos , Compartimento Celular , Proteínas de Ciclo Celular , Divisão Celular/genética , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ativação Enzimática , Proteínas Fúngicas/isolamento & purificação , Peptídeos e Proteínas de Sinalização Intracelular , MAP Quinase Quinase Quinases , Dados de Sequência Molecular , Mutagênese , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Pirazóis/farmacologia , Pirimidinas/farmacologia
2.
Nat Cell Biol ; 3(3): 325-30, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11231586

RESUMO

Heterogeneous nuclear ribonucleoprotein K (hnRNP-K) is one of a family of 20 proteins that are involved in transcription and post-transcriptional messenger RNA metabolism. The mechanisms that underlie regulation of hnRNP-K activities remain largely unknown. Here we show that cytoplasmic accumulation of hnRNP-K is phosphorylation-dependent. Mitogen-activated protein kinase/extracellular-signal-regulated kinase (MAPK/ERK) efficiently phosphorylates hnRNP-K both in vitro and in vivo at serines 284 and 353. Serum stimulation or constitutive activation of ERK kinase (MEK1) results in phosphorylation and cytoplasmic accumulation of hnRNP-K. Mutation at ERK phosphoacceptor sites in hnRNP-K abolishes the ability to accumulate in the cytoplasm and renders the protein incapable of regulating translation of mRNAs that have a differentiation-control element (DICE) in the 3' untranslated region (UTR). Similarly, treatment with a pharmacological inhibitor of the ERK pathway abolishes cytoplasmic accumulation of hnRNP-K and attenuates inhibition of mRNA translation. Our results establish the role of MAPK/ERK in phosphorylation-dependent cellular localization of hnRNP-K, which is required for its ability to silence mRNA translation.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Biossíntese de Proteínas , Ribonucleoproteínas/metabolismo , Araquidonato 15-Lipoxigenase/genética , Western Blotting , Linhagem Celular , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Genes Reporter/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo K , Ribonucleoproteínas Nucleares Heterogêneas , Humanos , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/genética , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Transfecção
3.
J Exp Med ; 191(8): 1443-8, 2000 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-10770810

RESUMO

Signal transduction through the B cell antigen receptor (BCR) is altered in B cells that express a receptor that recognizes self-antigen. To understand the molecular basis for the change in signaling in autoreactive B cells, a transgenic model was used to isolate a homogeneous population of tolerant B lymphocytes. These cells were compared with a similar population of naive B lymphocytes. We show that the BCR from naive B cells enters a detergent-insoluble domain of the cell within 6 s after antigen binding, before a detectable increase in BCR phosphorylation. This fraction appears to be important for signaling because it is enriched for lyn kinase but lacks CD45 tyrosine phosphatase and because the BCR that moves into this domain becomes more highly phosphorylated. Partitioning of the BCR into this fraction is unaffected by src family kinase inhibition. Tolerant B cells do not efficiently partition the BCR into the detergent-insoluble domain, providing an explanation for their reduced tyrosine kinase activation and calcium flux in response to antigen. These results identify an early, regulated step in antigen receptor signaling and self-tolerance.


Assuntos
Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Tolerância a Antígenos Próprios , Animais , Autoimunidade , Galinhas , Camundongos , Camundongos Transgênicos , Muramidase/imunologia , Fosforilação , Receptores de Antígenos de Linfócitos B/genética , Transdução de Sinais
4.
J Exp Med ; 179(2): 425-38, 1994 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8294858

RESUMO

The specificity of antibody (Ab) responses depends on focusing helper T (Th) lymphocyte signals to suitable B lymphocytes capable of binding foreign antigens (Ags), and away from nonspecific or self-reactive B cells. To investigate the molecular mechanisms that prevent the activation of self-reactive B lymphocytes, the activation requirements of B cells specific for the Ag hen egg lysozyme (HEL) obtained from immunoglobulin (Ig)-transgenic mice were compared with those of functionally tolerant B cells isolated from Ig-transgenic mice which also express soluble HEL. To eliminate the need for surface (s)Ig-mediated Ag uptake and presentation and allow the effects of sIg signaling to be studied in isolation, we assessed the ability of allogeneic T cells from bm12 strain mice to provide in vivo help to C57BL/6 strain-transgenic B cells. Interestingly, non-tolerant Ig-transgenic B cells required both allogeneic Th cells and binding of soluble HEL for efficient activation and Ab production. By contrast, tolerant self-reactive B cells from Ig/HEL double transgenic mice responded poorly to the same combination of allogeneic T cells and soluble HEL. The tolerant B cells were nevertheless normally responsive to stimulation with interleukin 4 and anti-CD40 Abs in vitro, suggesting that they retained the capacity to respond to mediators of T cell help. However, the tolerant B cells exhibited a proximal block in the sIg signaling pathway which prevented activation of receptor-associated tyrosine kinases in response to the binding of soluble HEL. The functional significance of this sIg signaling defect was confirmed by using a more potent membrane-bound form of HEL capable of triggering sIg signaling in tolerant B cells, which markedly restored their ability to collaborate with allogeneic Th cells and produce Ab. These findings indicate that Ag-specific B cells require two signals for mounting a T cell-dependent Ab response and identify regulation of sIg signaling as a mechanism for controlling self-reactive B cells.


Assuntos
Linfócitos B/fisiologia , Tolerância Imunológica , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais , Linfócitos T/fisiologia , Animais , Antígenos/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Comunicação Celular , Membrana Celular/enzimologia , Células Cultivadas , Imunoglobulina D/imunologia , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Muramidase/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
5.
Trends Cell Biol ; 11(4): 167-72, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11306297

RESUMO

A chemical-genetic method for the generation of target-specific protein kinase inhibitors has been developed recently. This strategy utilizes a functionally silent active-site mutation to sensitize a target kinase to inhibition by a small molecule that does not inhibit wild-type kinases. Tyrosine and serine/threonine kinases are equally amenable to the drug-sensitization approach, which has been used to generate selective inhibitors of mutant Src-family kinases, Abl-family kinases, cyclin-dependent kinases, mitogen-activated kinases, p21-activated kinases and Ca(2+)/calmodulin-dependent kinases. The designed inhibitors are specific for the sensitized kinase in a cellular background where the wild-type kinase has been inactivated. By these means, kinase-sensitization has been used systematically to generate and analyze conditional alleles of several yeast protein kinases in vivo.


Assuntos
Proteínas de Transporte/síntese química , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Quinases/genética , Proteínas Quinases/uso terapêutico , Animais , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Sistemas de Liberação de Medicamentos/tendências , Desenho de Fármacos , Terapia Genética/métodos , Terapia Genética/tendências , Humanos , Mutagênese , Proteínas Quinases/efeitos dos fármacos
6.
Curr Biol ; 8(5): 257-66, 1998 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-9501066

RESUMO

BACKGROUND: Deconvoluting protein kinase signaling pathways using conventional genetic and biochemical approaches has been difficult because of the overwhelming number of closely related kinases. If cell-permeable inhibitors of individual kinases could be designed, the role of each kinase could be systematically assessed. RESULTS: We have devised an approach combining chemistry and genetics to develop the first highly specific cell-permeable inhibitor of the oncogenic tyrosine kinase v-Src. A functionally silent active-site mutation was made in v-Src to distinguish it from all other cellular kinases. A tight-binding cell-permeable inhibitor of this mutant kinase that does not inhibit wild-type kinases was designed and synthesized. In vitro and whole-cell assays established the unique specificity of the mutant v-Src-inhibitor pair. The inhibitor reversed cell transformation by the engineered but not the 'wild type' v-Src, establishing that changes in cellular signaling can be attributed to specific inhibition of the engineered kinase. The generality of the method was tested by engineering another tyrosine kinase, Fyn, to contain the corresponding active-site mutation to the one in v-Src. The same compound that inhibited mutant v-Src could also potently inhibit the engineered Fyn kinase. CONCLUSIONS: Allele-specific cell-permeable inhibitors of individual Src family kinases can be rapidly developed in an approach that should be applicable to all kinases. This approach will be useful for the deconvolution of kinase-mediated cellular pathways and for validating novel kinases as good targets for drug discovery both in vitro and in vivo.


Assuntos
Alelos , Proteína Oncogênica pp60(v-src)/antagonistas & inibidores , Proteínas Quinases/fisiologia , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/antagonistas & inibidores , Células 3T3 , Trifosfato de Adenosina/metabolismo , Animais , Desenho de Fármacos , Camundongos , Modelos Químicos , Modelos Moleculares , Engenharia de Proteínas , Especificidade por Substrato
7.
Pharmacol Ther ; 82(2-3): 337-46, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10454210

RESUMO

Protein phosphorylation is the major post-translational modification used by eukaryotic cells to control cellular signaling. Protein kinases have emerged as attractive drug targets because heightened protein kinase activity has been associated with several proliferative diseases, most notably cancer and restenosis. Until now, it has been very difficult to confirm the utility of protein kinases as inhibitor targets because very few small molecules that selectively inhibit one particular kinase are known. Discovery of highly specific kinase inhibitors has been slow because the protein family contains approximately 2000 members, all of which share a conserved active site fold. Recent work in several laboratories has sought to circumvent the problem of kinase structural degeneracy by engineering drug sensitivity into Src family tyrosine kinases and mitogen-activated protein kinases through site-directed mutagenesis. By introducing a unique non-naturally occurring amino acid into a conserved region of the enzyme's binding site, a target protein kinase can be rapidly sensitized to a small molecule. Introduction of the engineered kinase into a cell line or animal model should greatly expedite the investigation of protein kinase inhibition as a viable drug treatment. The purpose of this review is to summarize these recent advances in protein kinase drug sensitization.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/genética , Inibidores de Proteínas Quinases , Proteínas Quinases/genética , Proteínas/síntese química , Quinases da Família src/genética , Animais , Desenho de Fármacos , Fibroblastos/patologia , Previsões , Humanos , Técnicas In Vitro , Proteínas Tirosina Quinases/genética , Sensibilidade e Especificidade , Homologia de Sequência
8.
Curr Opin Chem Biol ; 5(4): 360-7, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11470597

RESUMO

New chemical methods that use small molecules to perturb cellular function in ways analogous to genetics have recently been developed. These approaches include both synthetic methods for discovering small molecules capable of acting like genetic mutations, and techniques that combine the advantages of genetics and chemistry to optimize the potency and specificity of small-molecule inhibitors. Both approaches have been used to study protein function in vivo and have provided insights into complex signaling cascades.


Assuntos
Química , Genética , Transdução de Sinais , Fenômenos Químicos , Mutação
9.
Chem Biol ; 2(8): 509-14, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9383454

RESUMO

Cytoplasmic tyrosine kinases are composed of modular domains; one (SH1) has catalytic activity, the other two (SH2 and SH3) do not. Kinase specificity is largely determined by the binding preferences of the SH2 domain. Attaching the SH1 domain to a new SH2 domain, via protein-protein association or mutation, can thus dramatically change kinase function.


Assuntos
Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/fisiologia , Transdução de Sinais/fisiologia , Transdução de Sinais/genética , Especificidade por Substrato , Domínios de Homologia de src/genética , Domínios de Homologia de src/fisiologia
10.
Chem Biol ; 5(2): 91-101, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9495830

RESUMO

BACKGROUND: Protein kinases play a central role in controlling diverse signal transduction pathways in all cells. The identification of the direct cellular substrates of individual protein kinases remains the key challenge in the field. RESULTS: We describe the protein engineering of v-Src to produce a kinase which preferentially uses an ATP analog, N6-(benzyl) ATP, as a substrate, rather than the natural v-Src substrate, ATP. The sidechain of a single residue (Ile338) controls specificity for N6-substituted ATP analogs in the binding pocket of v-Src. Elimination of this sidechain by mutation to glycine produces a v-Src kinase which preferentially utilizes N6-(benzyl) ATP as a phosphodonor substrate. Our engineering strategy is generally applicable to the Src family kinases: mutation of the corresponding residue (Thr339 to glycine) in the Fyn kinase confers specificity for N6-(benzyl) ATP on Fyn. CONCLUSIONS: The v-Src tyrosine kinase has been engineered to exhibit specificity for an unnatural ATP analog, N6-(benzyl) ATP, even in a cellular context where high concentrations of natural ATP are present (1-5 mM), where preferential use of the ATP analog by the mutant kinase is essential. The mutant v-Src transfers phosphate more efficiently with the designed unnatural analog than with ATP. As the identical mutation in the Src-family kinase Fyn confers on Fyn the ability to recognize the same unnatural ATP analog, our strategy is likely to be generally applicable to other protein kinases and may help to identify the direct targets of specific kinases.


Assuntos
Nucleotídeos/metabolismo , Proteína Oncogênica pp60(v-src)/metabolismo , Engenharia de Proteínas , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/genética , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Mutagênese Sítio-Dirigida/genética , Proteína Oncogênica pp60(v-src)/genética , Peptídeos/metabolismo , Fosforilação , Fosfotirosina/análise , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-fyn , Proteínas Proto-Oncogênicas c-hck , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Especificidade por Substrato
11.
Chem Biol ; 6(9): 671-8, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10467133

RESUMO

BACKGROUND: Small-molecule inhibitors that can target individual kinases are powerful tools for use in signal transduction research. It is difficult to find such compounds because of the enormous number of protein kinases and the highly conserved nature of their catalytic domains. Recently, a novel, potent, Src family selective tyrosine kinase inhibitor was reported (PP1). Here, we study the structural basis for this inhibitor's specificity for Src family kinases. RESULTS: A single residue corresponding to Ile338 (v-Src numbering; Thr338 in c-Src) in Src family tyrosine kinases largely controls PP1's ability to inhibit protein kinases. Mutation of Ile338 to a larger residue such as methionine or phenylalanine in v-Src makes this inhibitor less potent. Conversely, mutation of Ile338 to alanine or glycine increases PP1's potency. PP1 can inhibit Ser/Thr kinases if the residue corresponding to Ile338 in v-Src is mutated to glycine. We have accurately predicted several non-Src family kinases that are moderately (IC(50) approximately 1 microM) inhibited by PP1, including c-Abl and the MAP kinase p38. CONCLUSIONS: Our mutagenesis studies of the ATP-binding site in both tyrosine kinases and Ser/Thr kinases explain why PP1 is a specific inhibitor of Src family tyrosine kinases. Determination of the structural basis of inhibitor specificity will aid in the design of more potent and more selective protein kinase inhibitors. The ability to desensitize a particular kinase to PP1 inhibition of residue 338 or conversely to sensitize a kinase to PP1 inhibition by mutation should provide a useful basis for chemical genetic studies of kinase signal transduction.


Assuntos
Inibidores Enzimáticos/farmacologia , Proteínas Quinases Ativadas por Mitógeno , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas/farmacologia , Quinases da Família src/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Proteínas Quinases Dependentes de Cálcio-Calmodulina/química , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Inibidores Enzimáticos/química , Isoleucina/fisiologia , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese/fisiologia , Mutação , Conformação Proteica , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/genética , Proteínas/química , Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno , Quinases da Família src/química , Quinases da Família src/genética
12.
Oncogene ; 33(12): 1590-600, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23542178

RESUMO

The mammalian target of rapamycin (mTOR) regulates cell growth by integrating nutrient and growth factor signaling and is strongly implicated in cancer. But mTOR is not an oncogene, and which tumors will be resistant or sensitive to new adenosine triphosphate (ATP) competitive mTOR inhibitors now in clinical trials remains unknown. We screened a panel of over 600 human cancer cell lines to identify markers of resistance and sensitivity to the mTOR inhibitor PP242. RAS and phosphatidylinositol 3-kinase catalytic subunit alpha (PIK3CA) mutations were the most significant genetic markers for resistance and sensitivity to PP242, respectively; colon origin was the most significant marker for resistance based on tissue type. Among colon cancer cell lines, those with KRAS mutations were most resistant to PP242, whereas those without KRAS mutations most sensitive. Surprisingly, cell lines with co-mutation of PIK3CA and KRAS had intermediate sensitivity. Immunoblot analysis of the signaling targets downstream of mTOR revealed that the degree of cellular growth inhibition induced by PP242 was correlated with inhibition of phosphorylation of the translational repressor eIF4E-binding protein 1 (4E-BP1), but not ribosomal protein S6 (rpS6). In a tumor growth inhibition trial of PP242 in patient-derived colon cancer xenografts, resistance to PP242-induced inhibition of 4E-BP1 phosphorylation and xenograft growth was again observed in KRAS mutant tumors without PIK3CA co-mutation, compared with KRAS wild-type controls. We show that, in the absence of PIK3CA co-mutation, KRAS mutations are associated with resistance to PP242 and that this is specifically linked to changes in the level of phosphorylation of 4E-BP1.


Assuntos
Trifosfato de Adenosina/metabolismo , Antineoplásicos/farmacologia , Ligação Competitiva/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Indóis/farmacologia , Fosfoproteínas/metabolismo , Purinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Neoplasias do Colo/patologia , Fatores de Iniciação em Eucariotos , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Mutação , Fosfatidilinositol 3-Quinases/genética , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Proteína S6 Ribossômica/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/genética
14.
Leukemia ; 22(9): 1698-706, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18548104

RESUMO

The phosphatidylinositol 3-kinase (PI3K)/Akt and mammalian target of rapamycin complex 1 (mTORC1) signaling pathways are frequently activated in acute myelogenous leukemia (AML). mTORC1 inhibition with RAD001 induces PI3K/Akt activation and both pathways are activated independently, providing a rationale for dual inhibition of both pathways. PI-103 is a new potent PI3K/Akt and mTOR inhibitor. In human leukemic cell lines and in primary blast cells from AML patients, PI-103 inhibited constitutive and growth factor-induced PI3K/Akt and mTORC1 activation. PI-103 was essentially cytostatic for cell lines and induced cell cycle arrest in the G1 phase. In blast cells, PI-103 inhibited leukemic proliferation, the clonogenicity of leukemic progenitors and induced mitochondrial apoptosis, especially in the compartment containing leukemic stem cells. In contrast, apoptosis was not induced with RAD001 and IC87114 association, which specifically inhibits mTORC1 and p110delta activity, respectively. PI-103 had additive proapoptotic effects with etoposide in blast cells and in immature leukemic cells. Interestingly, PI-103 did not induce apoptosis in normal CD34(+) cells and had moderate effects on their clonogenic and proliferative properties. Here, we demonstrate that multitargeted therapy against PI3K/Akt and mTOR with PI-103 may be of therapeutic value in AML.


Assuntos
Antineoplásicos/farmacologia , Furanos/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/efeitos dos fármacos , Piridinas/farmacologia , Pirimidinas/farmacologia , Apoptose/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Furanos/uso terapêutico , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Pirimidinas/uso terapêutico , Serina-Treonina Quinases TOR , Células Tumorais Cultivadas
15.
Biochem Soc Trans ; 35(Pt 2): 245-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17371250

RESUMO

PI3K (phosphoinositide 3-kinase) is a key regulator of cell growth, metabolism and survival. The frequent activation of the PI3K pathway in cancer has stimulated widespread interest in identifying potent and selective inhibitors of PI3K isoforms. The present paper highlights recent progress in identifying such molecules and the challenges that remain for efforts to pharmacologically target the PI3K family.


Assuntos
Fosfatidilinositol 3-Quinases/metabolismo , Animais , Divisão Celular , Sobrevivência Celular , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Neoplasias/enzimologia , Inibidores de Fosfoinositídeo-3 Quinase
16.
Biochemistry ; 26(12): 3378-84, 1987 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-3651388

RESUMO

Uridylate synthase is a bifunctional protein that first forms orotidine 5'-phosphate (OMP) from orotate via its orotate phosphoribosyltransferase activity (EC 2.4.2.10) and then converts OMP to uridine 5'-phosphate (UMP) via the OMP decarboxylase activity (EC 4.1.1.23). A computer modeling analysis of the experiments that led to the proposal [Traut, T.W., & Jones, M.E. (1977) J. Biol. Chem. 252, 8374-8381] that uridylate synthase channels intermediate OMP suggests that the experimental results do not demonstrate preferential use of OMP generated in the bifunctional complex as against exogenous OMP. This analysis shows that the experimentally observed amounts of [6-14C]UMP from [6-14C]orotate in the presence of various amounts of exogenous [7-14C]OMP agree well with the amounts predicted by the computer simulations. Thus we conclude that uridylate synthase does not channel OMP. Additionally, the subsequent suggestion that channeling of OMP occurs to protect the intermediate from degradation by a nucleotidase [Traut, T.W. (1980) Arch. Biochem. Biophys. 200, 590-594] seems unlikely. The appropriate computer simulation demonstrates that low transient levels of OMP and protection of the intermediate are provided for strictly by the kinetic parameters of orotate phosphoribosyltransferase, OMP decarboxylase, and the nucleotidase. Additionally, calculations show that, in both sets of published experiments, the concentration of transient OMP greatly exceeded the concentration of OMP decarboxylase active sites. Thus, channeling of OMP by the bifunctional complex cannot be invoked to explain the evolution of uridylate synthase, and that event must be the result of some other selective pressure.


Assuntos
Carboxiliases/metabolismo , Complexos Multienzimáticos/metabolismo , Orotato Fosforribosiltransferase/metabolismo , Orotidina-5'-Fosfato Descarboxilase/metabolismo , Pentosiltransferases/metabolismo , Nucleotídeos de Uracila/metabolismo , Uridina Monofosfato/metabolismo , Cinética , Modelos Teóricos , Fatores de Tempo , Uridina Monofosfato/análogos & derivados
17.
Ciba Found Symp ; 159: 118-28; discussion 128-34, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1959444

RESUMO

To expand the scope of antibody-catalysed reactions to those involving rate-limiting proton abstraction, such as elimination, isomerization and condensation reactions, we developed a new strategy--hapten charge complementarity. A hapten containing a benzyl ammonium group was used to elicit a specific base, a carboxylate, in the combining site of an antibody that catalysed a beta-elimination reaction. This was the first example of the use of a hapten to elicit a specific catalytic residue in an antibody combining site. A variety of kinetic and chemical modification experiments strongly suggest that a specific Asp or Glu residue in the combining site is responsible for catalysis. Preliminary results indicate that in addition to charge-charge complementarity, the nucleophilic reactivity of amino acid residues (Ser, Thr, Lys, Asp, Glu, Cys) in antibodies can be used as a selection tool. Antibodies were raised against a reactive epoxide group to elicit an antibody containing a uniquely reactive carboxylate or thiol group. Antibodies which bind the epoxide do catalyse a beta-elimination reaction, indicating the presence of a specific base in the combining site. Antibodies elicited to two closely related haptens do not catalyse the beta-elimination reaction.


Assuntos
Anticorpos/metabolismo , Sítios de Ligação de Anticorpos/fisiologia , Antígenos/imunologia , Catálise , Eletroquímica , Haptenos/imunologia
18.
Nature ; 375(6529): 334-8, 1995 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-7753200

RESUMO

During an immune response, hypermutation of immunoglobulin genes in B cells proliferating within germinal centres (GCs) generates variant antibodies that react with higher affinity against either foreign or self antigens. Several experiments suggest that self-reactive B cells may be censored at this stage of the immune response, but the rarity of these cells and the dynamic nature of GC reactions have prevented direct analysis. We have developed a new approach to visualize the fate of antigen-specific B cells during GC reactions by seeding an ongoing immune response with lysozyme-specific B cells from immunoglobulin-gene transgenic animals. Administration of soluble antigen at the peak of the GC response rapidly eliminates lysozyme-specific GC B cells in two waves of apoptosis, one within the GC and a second in cells that have redistributed to lymphoid zones that are rich in T cells. Elimination of these cells is inhibited by constitutive expression of the follicular lymphoma proto-oncogene bcl-2. These findings reveal censoring steps that may normally prevent affinity maturation of autoantibodies to systemic autoantigens, and might be used by pathogenic microorganisms or in clinical strategies to interfere with antibody responses.


Assuntos
Antígenos/imunologia , Apoptose , Linfócitos B/imunologia , Sequência de Aminoácidos , Animais , Autoantígenos/imunologia , Patos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Muramidase/imunologia , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2 , Solubilidade , Baço/citologia , Baço/imunologia , Linfócitos T/imunologia
20.
Proc Natl Acad Sci U S A ; 94(8): 3565-70, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9108016

RESUMO

Protein phosphorylation plays a central role in controlling many diverse signal transduction pathways in all cells. Novel protein kinases are identified at a rapid rate using homology cloning methods and genetic screens or selections; however identification of the direct substrates of kinases has proven elusive to genetic methods because of the tremendous redundancy and overlapping of substrate specificities among protein kinases. We describe the development of a protein engineering-based method to identify the direct substrates of the prototypical protein tyrosine kinase v-Src, which controls fibroblast transformation by the Rous sarcoma virus. To differentiate the substrates of v-Src from all other kinase substrates, we mutated the ATP binding site of v-Src such that the engineered v-Src uniquely accepted an ATP analog. We show that the engineered v-Src kinase displayed catalytic efficiency with the ATP analog, N(6)-(cyclopentyl) ATP, which is similar to the wild-type kinase catalytic efficiency with ATP itself. However, the N(6)-(cyclopentyl) ATP analog was not accepted by the wild-type kinase. Furthermore, the engineered v-Src exhibited the same protein target specificity as wild-type v-Src despite the proximity of the reengineered nucleotide binding site to the phosphoacceptor binding site. The successful engineering of v-Src's active site to accept a unique nucleotide analog provides a unique handle by which the direct substrates of one kinase (v-Src) can be traced in the presence of any number of cellular kinases.


Assuntos
Vírus do Sarcoma Aviário , Nucleotídeos/metabolismo , Proteína Oncogênica pp60(v-src)/metabolismo , Sequência de Aminoácidos , Animais , Transformação Celular Viral , Feminino , Fibroblastos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação , Nucleotídeos/genética , Proteína Oncogênica pp60(v-src)/genética , Fosforilação , Engenharia de Proteínas , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA