Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Biochemistry ; 58(52): 5320-5328, 2019 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-31095371

RESUMO

Acyl phosphates of ATP (ATPAc) and related nucleotides have proven to be useful for the interrogation of known nucleotide binding sites via specific acylation of conserved lysines (K). In addition, occasional K acylations are identified in proteins without such known sites. Here we present a robust and specific acylation of procaspase-6 by ATPAc at K133 in Jurkat cell lysates. The K133 acylation is dependent on π-π stacking interactions between the adenine moiety of ATPAc and a conserved Y198-Y198 site formed at the homodimeric interface of procaspase-6. Significantly, the Y198A mutation in procaspase-6 abolishes K133 acylation but has no effect on the proteolytic activity of the mature, active caspase-6 Y198A variant. Additional in vitro studies show that ATP can inhibit the autoproteolytic activation of procaspase-6. These observations suggest that ATP, and possibly other nucleotides, may serve as the endogenous ligands for the allosteric site at the procaspase-6 dimer interface, a site that has persisted in its "orphan" status for more than a decade.


Assuntos
Trifosfato de Adenosina/metabolismo , Caspase 6/química , Caspase 6/metabolismo , Precursores Enzimáticos/química , Precursores Enzimáticos/metabolismo , Proteômica , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Células Jurkat , Modelos Moleculares , Conformação Proteica
2.
Proc Natl Acad Sci U S A ; 113(42): 11865-11870, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27679845

RESUMO

Unlike other members of the MAPK family, ERK5 contains a large C-terminal domain with transcriptional activation capability in addition to an N-terminal canonical kinase domain. Genetic deletion of ERK5 is embryonic lethal, and tissue-restricted deletions have profound effects on erythroid development, cardiac function, and neurogenesis. In addition, depletion of ERK5 is antiinflammatory and antitumorigenic. Small molecule inhibition of ERK5 has been shown to have promising activity in cell and animal models of inflammation and oncology. Here we report the synthesis and biological characterization of potent, selective ERK5 inhibitors. In contrast to both genetic depletion/deletion of ERK5 and inhibition with previously reported compounds, inhibition of the kinase with the most selective of the new inhibitors had no antiinflammatory or antiproliferative activity. The source of efficacy in previously reported ERK5 inhibitors is shown to be off-target activity on bromodomains, conserved protein modules involved in recognition of acetyl-lysine residues during transcriptional processes. It is likely that phenotypes reported from genetic deletion or depletion of ERK5 arise from removal of a noncatalytic function of ERK5. The newly reported inhibitors should be useful in determining which of the many reported phenotypes are due to kinase activity and delineate which can be pharmacologically targeted.


Assuntos
Imunidade Celular , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Animais , Biomarcadores , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Citocinas/genética , Citocinas/metabolismo , Ativação Enzimática , Expressão Gênica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células HeLa , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imunidade Celular/efeitos dos fármacos , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Concentração Inibidora 50 , Camundongos , Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 7 Ativada por Mitógeno/genética , Estrutura Molecular , Fosforilação , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Relação Estrutura-Atividade , Transcriptoma
3.
Blood ; 128(2): 239-48, 2016 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-27151888

RESUMO

Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma, yet 40% to 50% of patients will eventually succumb to their disease, demonstrating a pressing need for novel therapeutic options. Gene expression profiling has identified messenger RNAs that lead to transformation, but critical events transforming cells are normally executed by kinases. Therefore, we hypothesized that previously unrecognized kinases may contribute to DLBCL pathogenesis. We performed the first comprehensive analysis of global kinase activity in DLBCL, to identify novel therapeutic targets, and discovered that germinal center kinase (GCK) was extensively activated. GCK RNA interference and small molecule inhibition induced cell-cycle arrest and apoptosis in DLBCL cell lines and primary tumors in vitro and decreased the tumor growth rate in vivo, resulting in a significantly extended lifespan of mice bearing DLBCL xenografts. GCK expression was also linked to adverse clinical outcome in a cohort of 151 primary DLBCL patients. These studies demonstrate, for the first time, that GCK is a molecular therapeutic target in DLBCL tumors and that inhibiting GCK may significantly extend DLBCL patient survival. Because the majority of DLBCL tumors (∼80%) exhibit activation of GCK, this therapy may be applicable to most patients.


Assuntos
Apoptose , Pontos de Checagem do Ciclo Celular , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Linfoma Difuso de Grandes Células B/etnologia , Proteínas de Neoplasias/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Animais , Linhagem Celular Tumoral , Quinases do Centro Germinativo , Xenoenxertos , Humanos , Linfoma Difuso de Grandes Células B/mortalidade , Linfoma Difuso de Grandes Células B/patologia , Camundongos , Transplante de Neoplasias
4.
Biochemistry ; 55(38): 5434-41, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27571378

RESUMO

Palbociclib is a cyclin-dependent kinase (CDK) 4/CDK6 inhibitor approved for breast cancer that is estrogen receptor (ER)-positive and human epidermal growth factor receptor 2 (HER2)-negative. We profiled palbociclib in cells either sensitive or resistant to the drug using an ATP/ADP probe-based chemoproteomics platform. Palbociclib only engaged CDK4 or CDK6 in sensitive cells. In resistant cells, no inhibition of CDK4 or CDK6 was observed, although the off-target profiles were similar in both cell types. Prolonged incubation of sensitive cells with the compound (24 h) resulted in the downregulation of additional kinases, including kinases critical for cell cycle progression. This downregulation is consistent with cell cycle arrest caused by palbociclib treatment. Both the direct and indirect targets were also observed in a human tumor xenograft study using the COLO-205 cell line in which phosphorylation of the retinoblastoma protein was tracked as the pharmacodyanamic marker. Together, these results suggest that this probe-based approach could be an important strategy toward predicting patient responsiveness to palbociclib.


Assuntos
Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Neoplasias/patologia , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteômica , Piridinas/farmacologia , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Neoplasias/enzimologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Biochemistry ; 54(19): 3024-36, 2015 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-25905789

RESUMO

Hsp90 is an ATP-dependent chaperone of widespread interest as a drug target. Here, using an LC-MS/MS chemoproteomics platform based on a lysine-reactive ATP acyl phosphate probe, several Hsp90 inhibitors were profiled in native cell lysates. Inhibitor specificities for all four human paralogs of Hsp90 were simultaneously monitored at their endogenous relative abundances. Equipotent inhibition of probe labeling in each paralog occurred at sites both proximal to and distal from bound ATP observed in Hsp90 cocrystal structures, suggesting that the ATP probe is assaying a native conformation not predicted by available structures. Inhibitor profiling against a comprehensive panel of protein kinases and other ATP-binding proteins detected in native cell lysates identified PMS2, a member of the GHKL ATPase superfamily as an off-target of NVP-AUY922 and radicicol. Because of the endogenously high levels of Hsp90 paralogs in typical cell lysates, the measured potency of inhibitors was weaker than published IC50 values. Significant inhibition of Hsp90 required inhibitor concentrations above a threshold where off-target activity was detectable. Direct on- and off-target engagement was measured by profiling lysates derived from cells treated with Hsp90 inhibitors. These studies also assessed the downstream cellular pathway effects of Hsp90 inhibition, including the down regulation of several known Hsp90 client proteins and some previously unknown client proteins. Overall, the ATP probe-based assay methodology enabled a broad characterization of Hsp90 inhibitor activity and specificity in native cell lysates.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Trifosfato de Adenosina/metabolismo , Linhagem Celular , Proteínas de Choque Térmico HSP90/química , Humanos , Transdução de Sinais , Espectrometria de Massas em Tandem
6.
Nat Chem Biol ; 7(4): 203-5, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21378983

RESUMO

Mutations in leucine-rich repeat kinase 2 (LRRK2) are strongly associated with late-onset autosomal dominant Parkinson's disease. We employed a new, parallel, compound-centric approach to identify a potent and selective LRRK2 inhibitor, LRRK2-IN-1, and demonstrated that inhibition of LRRK2 induces dephosphorylation of Ser910 and Ser935 and accumulation of LRRK2 within aggregate structures. LRRK2-IN-1 will serve as a versatile tool to pharmacologically interrogate LRRK2 biology and study its role in Parkinson's disease.


Assuntos
Benzodiazepinonas/farmacologia , Inibidores Enzimáticos/farmacologia , Doença de Parkinson/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática , Células HEK293 , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Mutação , Doença de Parkinson/genética , Doença de Parkinson/patologia , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Serina/química , Serina/metabolismo
7.
J Pharmacol Exp Ther ; 338(1): 114-24, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21505060

RESUMO

The endogenous cannabinoid (endocannabinoid) anandamide is principally degraded by the integral membrane enzyme fatty acid amide hydrolase (FAAH). Pharmacological blockade of FAAH has emerged as a potentially attractive strategy for augmenting endocannabinoid signaling and retaining the beneficial effects of cannabinoid receptor activation, while avoiding the undesirable side effects, such as weight gain and impairments in cognition and motor control, observed with direct cannabinoid receptor 1 agonists. Here, we report the detailed mechanistic and pharmacological characterization of N-pyridazin-3-yl-4-(3-{[5-(trifluoromethyl)pyridin-2-yl]oxy}benzylidene)piperidine-1-carboxamide (PF-04457845), a highly efficacious and selective FAAH inhibitor. Mechanistic studies confirm that PF-04457845 is a time-dependent, covalent FAAH inhibitor that carbamylates FAAH's catalytic serine nucleophile. PF-04457845 inhibits human FAAH with high potency (k(inact)/K(i) = 40,300 M(-1)s(-1); IC(50) = 7.2 nM) and is exquisitely selective in vivo as determined by activity-based protein profiling. Oral administration of PF-04457845 produced potent antinociceptive effects in both inflammatory [complete Freund's adjuvant (CFA)] and noninflammatory (monosodium iodoacetate) pain models in rats, with a minimum effective dose of 0.1 mg/kg (CFA model). PF-04457845 displayed a long duration of action as a single oral administration at 1 mg/kg showed in vivo efficacy for 24 h with a concomitant near-complete inhibition of FAAH activity and maximal sustained elevation of anandamide in brain. Significantly, PF-04457845-treated mice at 10 mg/kg elicited no effect in motility, catalepsy, and body temperature. Based on its exceptional selectivity and in vivo efficacy, combined with long duration of action and optimal pharmacokinetic properties, PF-04457845 is a clinical candidate for the treatment of pain and other nervous system disorders.


Assuntos
Amidoidrolases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Dor/tratamento farmacológico , Dor/enzimologia , Piridazinas/farmacologia , Piridazinas/uso terapêutico , Ureia/análogos & derivados , Amidoidrolases/metabolismo , Animais , Inibidores Enzimáticos/química , Humanos , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piperidinas/química , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Piridazinas/química , Ratos , Ratos Sprague-Dawley , Ureia/química , Ureia/farmacologia , Ureia/uso terapêutico
8.
Bioorg Med Chem Lett ; 19(10): 2865-9, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19386497

RESUMO

The synthesis and structure-activity relationships (SAR) of a series of benzothiophene piperazine and piperidine urea FAAH inhibitors is described. These compounds inhibit FAAH by covalently modifying the enzyme's active site serine nucleophile. Activity-based protein profiling (ABPP) revealed that these urea inhibitors were completely selective for FAAH relative to other mammalian serine hydrolases. Several compounds showed in vivo activity in a rat complete Freund's adjuvant (CFA) model of inflammatory pain.


Assuntos
Amidoidrolases/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Piperazinas/química , Piperidinas/química , Tiofenos/química , Ureia/análogos & derivados , Amidoidrolases/metabolismo , Animais , Simulação por Computador , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Modelos Químicos , Piperazinas/síntese química , Piperazinas/farmacologia , Piperidinas/síntese química , Piperidinas/farmacologia , Ratos , Relação Estrutura-Atividade , Tiofenos/síntese química , Tiofenos/farmacologia , Ureia/síntese química , Ureia/farmacologia
9.
Commun Biol ; 2: 345, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31552298

RESUMO

Expression of human asparagine synthetase (ASNS) promotes metastatic progression and tumor cell invasiveness in colorectal and breast cancer, presumably by altering cellular levels of L-asparagine. Human ASNS is therefore emerging as a bona fide drug target for cancer therapy. Here we show that a slow-onset, tight binding inhibitor, which exhibits nanomolar affinity for human ASNS in vitro, exhibits excellent selectivity at 10 µM concentration in HCT-116 cell lysates with almost no off-target binding. The high-resolution (1.85 Å) crystal structure of human ASNS has enabled us to identify a cluster of negatively charged side chains in the synthetase domain that plays a key role in inhibitor binding. Comparing this structure with those of evolutionarily related AMP-forming enzymes provides insights into intermolecular interactions that give rise to the observed binding selectivity. Our findings demonstrate the feasibility of developing second generation human ASNS inhibitors as lead compounds for the discovery of drugs against metastasis.

11.
Assay Drug Dev Technol ; 1(1 Pt 2): 137-46, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15090140

RESUMO

In the latter stages of drug discovery and development, assays that establish drug selectivity and toxicity are important when side effects, which are often due to lack of specificity, determine drug candidate viability. There has been no comprehensive or systematic methodology to measure these factors outside of whole-animal assays, and such phenomenological assays generally fail to establish the additional targets of a given small molecule, or the molecular origin of toxicity. Consequently, small-molecule development programs destined for failure often reach advanced stages of testing, and the money and time invested in such programs could be saved if information on selectivity were available early in the process. Here, we present a methodology that utilizes chemical ABPs in combination with small-molecule inhibitors to selectively label small-molecule binding sites in whole proteomic samples. In principle, the ABP and small molecule will compete for similar binding sites, such that the small molecule will protect against modification by the ABP. Thus, after removal of the small molecule, the binding site for the ABP will be revealed, and a second probe can then be used to label the small-molecule binding sites selectively. To demonstrate this experimentally, we mapped the binding sites of the DPP4 inhibitor, IT, in a number of different tissue types.


Assuntos
Isoleucina/análogos & derivados , Preparações Farmacêuticas/química , Proteínas/química , Proteoma/química , Animais , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Células Cultivadas , Dipeptidil Peptidase 4/metabolismo , Desenho de Fármacos , Corantes Fluorescentes/química , Humanos , Técnicas In Vitro , Isoleucina/metabolismo , Rim/química , Rim/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Camundongos , Organofosfonatos/química , Preparações Farmacêuticas/metabolismo , Inibidores de Proteases/metabolismo , Proteínas/metabolismo , Relação Estrutura-Atividade , Tiazóis/metabolismo
12.
FEBS Lett ; 587(13): 1870-7, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23684650

RESUMO

The largest mammalian enzyme family is the kinases. Kinases and other nucleotide-binding proteins are key regulators of signal transduction pathways and the mutation or overexpression of these proteins is often the difference between health and disease. As a result, a massive research effort has focused on understanding how these proteins function and how to inhibit them for therapeutic benefit. Recent advances in chemical biological tools have enabled functional interrogation of these enzymes to provide a deeper understanding of their physiological roles. In addition, these innovative platforms have paved the way for a new generation of drugs whose properties have been guided by functional profiling.


Assuntos
Fosfotransferases/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Ensaios Enzimáticos , Humanos , Fosforilação , Fosfotransferases/química , Mapas de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Proteômica
13.
Curr Protoc Chem Biol ; 5(3): 213-26, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24391084

RESUMO

The protocols in this unit describe efficient and cost-effective approaches to determine the interaction of small-molecule inhibitors with native kinases, and also analyze the interactions between kinases and their binding partners in a cellular setting. The combined attributes of activity-based probes and western blotting procedures provide for quantitative measurement of inhibitor efficacy, isoform selectivity, and post-translational modifications. We further demonstrate the ability to identify protein-protein interactions between a probe-labeled protein and its noncovalent binding partners.


Assuntos
Técnicas Imunológicas , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/química , Proteínas Quinases/imunologia , Animais , Linhagem Celular , Células Cultivadas , Receptores ErbB/antagonistas & inibidores , Humanos , Indicadores e Reagentes , MAP Quinase Quinase 4/antagonistas & inibidores , Ligação Proteica , Inibidores de Proteínas Quinases/química , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas
14.
Chem Biol ; 19(1): 140-54, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22284361

RESUMO

The mitogen-activated kinases JNK1/2/3 are key enzymes in signaling modules that transduce and integrate extracellular stimuli into coordinated cellular response. Here, we report the discovery of irreversible inhibitors of JNK1/2/3. We describe two JNK3 cocrystal structures at 2.60 and 2.97 Å resolution that show the compounds form covalent bonds with a conserved cysteine residue. JNK-IN-8 is a selective JNK inhibitor that inhibits phosphorylation of c-Jun, a direct substrate of JNK, in cells exposed to submicromolar drug in a manner that depends on covalent modification of the conserved cysteine residue. Extensive biochemical, cellular, and pathway-based profiling establish the selectivity of JNK-IN-8 for JNK and suggests that the compound will be broadly useful as a pharmacological probe of JNK-dependent signal transduction. Potential lead compounds have also been identified for kinases, including IRAK1, PIK3C3, PIP4K2C, and PIP5K3.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Sítios de Ligação , Linhagem Celular Tumoral , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína
15.
Chem Biol ; 18(6): 699-710, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21700206

RESUMO

Protein kinases are intensely studied mediators of cellular signaling, yet important questions remain regarding their regulation and in vivo properties. Here, we use a probe-based chemoprotemics platform to profile several well studied kinase inhibitors against >200 kinases in native cell proteomes and reveal biological targets for some of these inhibitors. Several striking differences were identified between native and recombinant kinase inhibitory profiles, in particular, for the Raf kinases. The native kinase binding profiles presented here closely mirror the cellular activity of these inhibitors, even when the inhibition profiles differ dramatically from recombinant assay results. Additionally, Raf activation events could be detected on live cell treatment with inhibitors. These studies highlight the complexities of protein kinase behavior in the cellular context and demonstrate that profiling with only recombinant/purified enzymes can be misleading.


Assuntos
Proteínas Quinases/química , Trifosfato de Adenosina/química , Linhagem Celular Tumoral , Dasatinibe , Humanos , MAP Quinase Quinase 5/antagonistas & inibidores , MAP Quinase Quinase 5/metabolismo , Ligação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Pirimidinas/química , Pirimidinas/farmacologia , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tiazóis/química , Tiazóis/farmacologia , Quinases raf/antagonistas & inibidores , Quinases raf/genética , Quinases raf/metabolismo
16.
ACS Med Chem Lett ; 2(2): 91-96, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21666860

RESUMO

Fatty acid amide hydrolase (FAAH) is an integral membrane serine hydrolase that degrades the fatty acid amide family of signaling lipids, including the endocannabinoid anandamide. Genetic or pharmacological inactivation of FAAH leads to analgesic and anti-inflammatory phenotypes in rodents without showing the undesirable side effects observed with direct cannabinoid receptor agonists, indicating that FAAH may represent an attractive therapeutic target for the treatment of inflammatory pain and other nervous system disorders. Herein, we report the discovery and characterization of a highly efficacious and selective FAAH inhibitor PF-04457845 (23). Compound 23 inhibits FAAH by a covalent, irreversible mechanism involving carbamylation of the active-site serine nucleophile of FAAH with high in vitro potency (k(inact)/K(i) and IC(50) values of 40300 M(-1) s(-1) and 7.2 nM, respectively, for human FAAH). Compound 23 has exquisite selectivity for FAAH relative to other members of the serine hydrolase superfamily as demonstrated by competitive activity-based protein profiling. Oral administration of 23 at 0.1 mg/kg results in efficacy comparable to that of naproxen at 10 mg/kg in a rat model of inflammatory pain. Compound 23 is being evaluated in human clinical trials.

17.
Biochemistry ; 46(2): 350-8, 2007 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-17209545

RESUMO

The central role of protein kinases in signal transduction pathways has generated intense interest in targeting these enzymes for a wide range of therapeutic indications. Here we report a method for identifying and quantifying protein kinases in any biological sample or tissue from any species. The procedure relies on acyl phosphate-containing nucleotides, prepared from a biotin derivative and ATP or ADP. The acyl phosphate probes react selectively and covalently at the ATP binding sites of at least 75% of the known human protein kinases. Biotinylated peptide fragments from labeled proteomes are captured and then sequenced and identified using a mass spectrometry-based analysis platform to determine the kinases present and their relative levels. Further, direct competition between the probes and inhibitors can be assessed to determine inhibitor potency and selectivity against native protein kinases, as well as hundreds of other ATPases. The ability to broadly profile kinase activities in native proteomes offers an exciting prospect for both target discovery and inhibitor selectivity profiling.


Assuntos
Nucleotídeos de Adenina/metabolismo , Proteínas Quinases/metabolismo , Nucleotídeos de Adenina/química , Sítios de Ligação , Linhagem Celular , Sequência Conservada , Humanos , Modelos Moleculares , Técnicas de Sonda Molecular , Conformação Proteica , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/química , Proteínas Quinases/genética , Proteoma , Transdução de Sinais , Estaurosporina/farmacologia
18.
Biochemistry ; 46(45): 13019-30, 2007 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-17949010

RESUMO

Fatty acid amide hydrolase (FAAH) is an integral membrane enzyme that degrades the fatty acid amide family of signaling lipids, including the endocannabinoid anandamide. Genetic or pharmacological inactivation of FAAH leads to analgesic, anti-inflammatory, anxiolytic, and antidepressant phenotypes in rodents without showing the undesirable side effects observed with direct cannabinoid receptor agonists, indicating that FAAH may represent an attractive therapeutic target for treatment of pain, inflammation, and other central nervous system disorders. However, the FAAH inhibitors reported to date lack drug-like pharmacokinetic properties and/or selectivity. Herein we describe piperidine/piperazine ureas represented by N-phenyl-4-(quinolin-3-ylmethyl)piperidine-1-carboxamide (PF-750) and N-phenyl-4-(quinolin-2-ylmethyl)piperazine-1-carboxamide (PF-622) as a novel mechanistic class of FAAH inhibitors. PF-750 and PF-622 show higher in vitro potencies than previously established classes of FAAH inhibitors. Rather unexpectedly based on the high chemical stability of the urea functional group, PF-750 and PF-622 were found to inhibit FAAH in a time-dependent manner by covalently modifying the enzyme's active site serine nucleophile. Activity-based proteomic profiling revealed that PF-750 and PF-622 were completely selective for FAAH relative to other mammalian serine hydrolases. We hypothesize that this remarkable specificity derives, at least in part, from FAAH's special ability to function as a C(O)-N bond hydrolase, which distinguishes it from the vast majority of metabolic serine hydrolases in mammals that are restricted to hydrolyzing esters and/or thioesters. The piperidine/piperazine urea may thus represent a privileged chemical scaffold for the synthesis of FAAH inhibitors that display an unprecedented combination of potency and selectivity for use as potential analgesic and anxiolytic/antidepressant agents.


Assuntos
Amidoidrolases/antagonistas & inibidores , Anilidas/farmacologia , Inibidores Enzimáticos/farmacologia , Piperazinas/farmacologia , Piperidinas/farmacologia , Anilidas/química , Animais , Benzamidas/farmacologia , Sítios de Ligação/efeitos dos fármacos , Carbamatos/farmacologia , Inibidores Enzimáticos/química , Humanos , Concentração Inibidora 50 , Camundongos , Piperazinas/química , Piperidinas/química , Serina/química , Fatores de Tempo
19.
Proc Natl Acad Sci U S A ; 99(2): 585-90, 2002 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-11782529

RESUMO

The genetic code is established by the aminoacylation reactions of tRNA synthetases. Its accuracy depends on editing reactions that prevent amino acids from being assigned to incorrect codons. A group of class I synthetases share a common insertion that encodes a distinct site for editing that is about 30 A from the active site. Both misactivated aminoacyl adenylates and mischarged amino acids attached to tRNA are translocated to this site, which, in turn, is divided into subsites--one for the adenylate and one for the aminoacyl moiety attached to tRNA. Here we report that a specific mutation in isoleucyl-tRNA synthetase prevents editing by blocking translocation. The mutation alters a widely conserved residue that is believed to tether the amino group of mischarged tRNA to its subsite for editing. These and other data support a model where editing is initiated by translocation of the misacylated amino acid attached to tRNA to create an "editing complex" that facilitates subsequent rounds of editing by translocation of the misactivated adenylate.


Assuntos
Isoleucina-tRNA Ligase/química , Isoleucina-tRNA Ligase/genética , Sequência de Aminoácidos , Sítios de Ligação/genética , Transporte Biológico Ativo/genética , Isoleucina-tRNA Ligase/metabolismo , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Edição de RNA , RNA de Transferência de Isoleucina/metabolismo , Homologia de Sequência de Aminoácidos , Thermus thermophilus/enzimologia , Thermus thermophilus/genética
20.
Mol Cell ; 9(2): 353-62, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11864608

RESUMO

Aminoacyl tRNA synthetases (aaRSs) catalyze the first step in protein biosynthesis, establishing a connection between codons and amino acids. To maintain accuracy, aaRSs have evolved a second active site that eliminates noncognate amino acids. Isoleucyl tRNA synthetase edits valine by two tRNA(Ile)-dependent pathways: hydrolysis of valyl adenylate (Val-AMP, pretransfer editing) and hydrolysis of mischarged Val-tRNA(Ile) (posttransfer editing). Not understood is how a single editing site processes two distinct substrates--an adenylate and an aminoacyl tRNA ester. We report here distinct mutations within the center for editing that alter adenylate but not aminoacyl ester hydrolysis, and vice versa. These results are consistent with a molecular model that shows that the single editing active site contains two valyl binding pockets, one specific for each substrate.


Assuntos
Proteínas de Escherichia coli/fisiologia , Isoleucina-tRNA Ligase/fisiologia , Biossíntese de Proteínas/fisiologia , Monofosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Esterificação , Hidrólise , Isoleucina/química , Isoleucina-tRNA Ligase/química , Isoleucina-tRNA Ligase/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Aminoacil-RNA de Transferência/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/fisiologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Especificidade por Substrato , Valina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA