Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Chem Inf Model ; 53(5): 1138-56, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23581389

RESUMO

FabH (Fatty acid biosynthesis, enzyme H, also referred to as ß-ketoacyl-ACP-synthase III) is a key condensing enzyme in the type II fatty acid synthesis (FAS) system. The FAS pathway in bacteria is essential for growth and survival and vastly differs from the human FAS pathway. Enzymes involved in this pathway have arisen as promising biomolecular targets for discovery of new antibacterial drugs. However, currently there are no clinical drugs that selectively target FabH, and known inhibitors of FabH all act within the active site. FabH exerts its catalytic function as a dimer, which could potentially be exploited in developing new strategies for inhibitor design. The aim of this study was to elucidate structural details of the dimer interface region by means of computational modeling, including molecular dynamics (MD) simulations, in order to derive information for the structure-based design of new FabH inhibitors. The dimer interface region was analyzed by MD simulations, trajectory snapshots were collected for further analyses, and docking studies were performed with potential small molecule disruptors. Alanine mutation and docking studies strongly suggest that the dimer interface could be a potential target for anti-infection drug discovery.


Assuntos
Acetiltransferases/química , Acetiltransferases/metabolismo , Alanina , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mutagênese , 3-Oxoacil-(Proteína de Transporte de Acila) Sintase , Acetiltransferases/antagonistas & inibidores , Acetiltransferases/genética , Sítios de Ligação , Domínio Catalítico , Análise por Conglomerados , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/genética , Ácido Graxo Sintase Tipo II/antagonistas & inibidores , Ácido Graxo Sintase Tipo II/química , Ácido Graxo Sintase Tipo II/genética , Ácido Graxo Sintase Tipo II/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Reprodutibilidade dos Testes , Solventes/química , Interface Usuário-Computador
2.
J Chem Inf Model ; 52(8): 2192-203, 2012 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-22747098

RESUMO

Computational methods involving virtual screening could potentially be employed to discover new biomolecular targets for an individual molecule of interest (MOI). However, existing scoring functions may not accurately differentiate proteins to which the MOI binds from a larger set of macromolecules in a protein structural database. An MOI will most likely have varying degrees of predicted binding affinities to many protein targets. However, correctly interpreting a docking score as a hit for the MOI docked to any individual protein can be problematic. In our method, which we term "Virtual Target Screening (VTS)", a set of small drug-like molecules are docked against each structure in the protein library to produce benchmark statistics. This calibration provides a reference for each protein so that hits can be identified for an MOI. VTS can then be used as tool for: drug repositioning (repurposing), specificity and toxicity testing, identifying potential metabolites, probing protein structures for allosteric sites, and testing focused libraries (collection of MOIs with similar chemotypes) for selectivity. To validate our VTS method, twenty kinase inhibitors were docked to a collection of calibrated protein structures. Here, we report our results where VTS predicted protein kinases as hits in preference to other proteins in our database. Concurrently, a graphical interface for VTS was developed.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Interface Usuário-Computador , Linhagem Celular Tumoral , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/química , Quinase 2 Dependente de Ciclina/metabolismo , Bases de Dados de Proteínas , Aprovação de Drogas , Humanos , Modelos Moleculares , Conformação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo , Proteínas Quinases/química , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia
3.
J Chem Inf Model ; 48(3): 639-45, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18266348

RESUMO

Consideration of stereochemistry early in the identification and optimization of lead compounds can improve the efficiency and efficacy of the drug discovery process and reduce the time spent on subsequent drug development. These improvements can result by focusing on specific enantiomers that have the desired potential therapeutic effect (eutomers), while removing from consideration enantiomers that may have no, or even undesirable, effects (distomers). A virtual screening campaign that correctly takes stereochemical information into account can, in theory, be utilized to provide information about the relative binding affinities of enantiomers. Thus, the proper enumeration of the relevant stereoisomers in general, and enantiomeric pairs in particular, of chiral compounds is crucial if one is to use virtual screening as an effective drug discovery tool. As is obvious, in cases where no stereochemical information is provided for chiral compounds in a 2D chemical database, then each possible stereoisomer should be generated for construction of the subsequent 3D database to be used for virtual screening. However, acute problems can arise in 3D database construction when relative stereochemistry is encoded in a 2D database for a chiral compound containing multiple stereogenic atoms but absolute stereochemistry is not implied. In this case, we report that generation of enantiomeric pairs is imperative in database development if one is to obtain accurate docking results. A study is described on the impact of the neglect of enantiomeric pairs on virtual screening using the human homolog of murine double minute 2 (MDM2) protein, the product of a proto-oncogene, as the target. Docking in MDM2 with GLIDE 4.0 was performed using the NCI Diversity Set 3D database and, for comparison, a set of enantiomers we created corresponding to mirror image structures of the single enantiomers of chiral compounds present in the NCI Diversity Set. Our results demonstrate that potential lead candidates may be overlooked when databases contain 3D structures representing only a single enantiomer of racemic chiral compounds.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Modelos Moleculares , Proto-Oncogene Mas , Estereoisomerismo
4.
J Chem Inf Model ; 47(5): 1897-905, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17676832

RESUMO

In silico chemical library screening (virtual screening) was used to identify a novel lead compound capable of inhibiting S-adenosylmethionine decarboxylase (AdoMetDC). AdoMetDC is intimately involved in the biosynthesis of polyamines, which are essential for tumor progression and are elevated in numerous types of tumors. Therefore, inhibition of this enzyme provides an attractive target for the discovery of novel anticancer drugs. We performed virtual screening using a computer model derived from the X-ray crystal structure of human AdoMetDC and the National Cancer Institute's Diversity Set (1990 compounds). Our docking study suggested several compounds that could serve as drug candidates since their docking modes and scores revealed potential inhibitory activity toward AdoMetDC. Experimental testing of the top-scoring compounds indicated that one of these compounds (NSC 354961) possesses an IC50 in the low micromolar range. A search of the entire NCI compound collection for compounds similar to NSC 354961 yielded two additional compounds that exhibited activity in the experimental assay but with significantly diminished potency relative to NSC 354961. In this report, we disclose the activity of NSC 354961 against AdoMetDC and its probable binding mode based on computational modeling. We also discuss the importance of virtual screening in the context of enzymes that are not readily amenable to high-throughput assays, thereby demonstrating the efficacy of virtual screening, combined with selective experimental testing, in identifying new potential drug candidates.


Assuntos
Adenosilmetionina Descarboxilase/antagonistas & inibidores , Aminacrina/química , Aminacrina/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Aminacrina/síntese química , Dióxido de Carbono/análise , Dióxido de Carbono/química , Simulação por Computador , Computadores , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/síntese química , Humanos , Proteínas Recombinantes/química , Reprodutibilidade dos Testes , Software , Relação Estrutura-Atividade
5.
Proc Natl Acad Sci U S A ; 104(18): 7391-6, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17463090

RESUMO

S3I-201 (NSC 74859) is a chemical probe inhibitor of Stat3 activity, which was identified from the National Cancer Institute chemical libraries by using structure-based virtual screening with a computer model of the Stat3 SH2 domain bound to its Stat3 phosphotyrosine peptide derived from the x-ray crystal structure of the Stat3beta homodimer. S3I-201 inhibits Stat3.Stat3 complex formation and Stat3 DNA-binding and transcriptional activities. Furthermore, S3I-201 inhibits growth and induces apoptosis preferentially in tumor cells that contain persistently activated Stat3. Constitutively dimerized and active Stat3C and Stat3 SH2 domain rescue tumor cells from S3I-201-induced apoptosis. Finally, S3I-201 inhibits the expression of the Stat3-regulated genes encoding cyclin D1, Bcl-xL, and survivin and inhibits the growth of human breast tumors in vivo. These findings strongly suggest that the antitumor activity of S3I-201 is mediated in part through inhibition of aberrant Stat3 activation and provide the proof-of-concept for the potential clinical use of Stat3 inhibitors such as S3I-201 in tumors harboring aberrant Stat3.


Assuntos
Ácidos Aminossalicílicos , Antineoplásicos/química , Antineoplásicos/metabolismo , Benzenossulfonatos/química , Benzenossulfonatos/metabolismo , Avaliação Pré-Clínica de Medicamentos , Fator de Transcrição STAT3/química , Fator de Transcrição STAT3/metabolismo , Ácido Aminossalicílico/química , Ácido Aminossalicílico/metabolismo , Ácido Aminossalicílico/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Apoptose , Benzenossulfonatos/uso terapêutico , Linhagem Celular , Biologia Computacional , DNA/química , DNA/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fosfotirosina/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Transcrição Gênica/genética , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Front Biosci ; 9: 2652-62, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15358588

RESUMO

The trace element copper is vital to the healthy functioning of organisms. Copper is used in a multitude of cellular activities including respiration, angiogenesis, and immune responses. Like other metals, copper homeostasis is a tightly regulated process. Copper is transported from dietary intake through the serum and into cells via a variety of transporters. There are a variety of copper chaperones designed to insure that copper is sequestered from interaction with cellular membranes, proteins, or DNA where its properties can result in oxidative damage. However, there are disease states in which copper transporters crucial to homeostasis are impaired resulting in potentially toxic copper accumulation. Wilsons and Menkes diseases are two such cases. Wilsons disease (hepatolenticular degeneration) is an autosomal recessive disorder resulting in extreme accumulation of copper in the liver with deposits elsewhere in the body. Menkes is characterized by a systemic copper deficiency (different from the liver specificity of Wilsons disease) and is the result of an X-linked recessive mutation in a copper transporter. Uptake of copper is impaired due to inability to remove existing copper from cells primarily in the small intestine. Though the causes are dramatically different, cancer also shares a similar diagnostic in the accumulation of copper in effected tissues. Studies have shown greatly elevated levels of copper in cancer tissues, and some diagnostics and treatments from Wilsons and Menkes diseases, such as copper chelation therapy, have been used in the treatment of cancer. Given the commonality of copper accumulation in these diseases and that common therapies exist between them, it may prove beneficial to study all three diseases in light of copper homeostasis. This review will examine the chemical nature and biological roles of copper, Wilsons and Menkes disease and their therapies, and the use of copper related therapies in cancer.


Assuntos
Cobre/metabolismo , Degeneração Hepatolenticular/diagnóstico , Síndrome dos Cabelos Torcidos/diagnóstico , Neoplasias/diagnóstico , Animais , Quelantes/farmacologia , Desenho de Fármacos , Degeneração Hepatolenticular/patologia , Humanos , Ligantes , Síndrome dos Cabelos Torcidos/patologia , Mutação , Neoplasias/patologia , Estresse Oxidativo , Oxigênio/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo
7.
Proteins ; 54(1): 58-70, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14705024

RESUMO

Previously, we demonstrated that natural and synthetic ester bond-containing green tea polyphenols were potent and specific non-peptide proteasome inhibitors. However, the molecular mechanism of inhibition is currently unknown. Here, we report that inhibition of the chymotrypsin activity of the 20S proteasome by (-)-epigallocatechin-3-gallate (EGCG) is time-dependent and irreversible, implicating acylation of the beta5-subunit's catalytic N-terminal threonine (Thr 1). This knowledge is used, along with in silico docking experiments, to aid in the understanding of binding and inhibition. On the basis of these docking experiments, we propose that (-)-EGCG binds the chymotrypsin site in an orientation and conformation that is suitable for a nucleophilic attack by Thr 1. Consistently, the distance from the electrophilic carbonyl carbon of (-)-EGCG to the hydroxyl group of Thr 1 was measured as 3.18 A. Furthermore, the A ring of (-)-EGCG acts as a tyrosine mimic, binding to the hydrophobic S1 pocket of the beta5-subunit. In the process, the (-)-EGCG scissile bond may become strained, which could lower the activation energy for attack by the hydroxyl group of Thr 1. This model is validated by comparison of predicted and actual activities of several EGCG analogs, either naturally occurring, previously synthesized, or rationally synthesized.


Assuntos
Catequina/análogos & derivados , Catequina/química , Cisteína Endopeptidases/efeitos dos fármacos , Inibidores de Cisteína Proteinase/química , Modelos Moleculares , Complexos Multienzimáticos/efeitos dos fármacos , Amidas/química , Sítios de Ligação , Camellia sinensis/química , Catequina/metabolismo , Catequina/farmacologia , Quimotripsina/metabolismo , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Desenho de Fármacos , Flavonoides/química , Flavonoides/metabolismo , Flavonoides/farmacologia , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Fenóis/química , Fenóis/metabolismo , Fenóis/farmacologia , Polifenóis , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA