Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1834(1): 46-52, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22999981

RESUMO

BACKGROUND: Bacterial resistance to antibiotic therapies is increasing and new treatment options are badly needed. There is an overlap between these resistant bacteria and organisms classified as likely bioterror weapons. For example, Bacillus anthracis is innately resistant to the anti-folate trimethoprim due to sequence changes found in the dihydrofolate reductase enzyme. Development of new inhibitors provides an opportunity to enhance the current arsenal of anti-folate antibiotics while also expanding the coverage of the anti-folate class. METHODS: We have characterized inhibitors of B. anthracis dihydrofolate reductase by measuring the K(i) and MIC values and calculating the energetics of binding. This series contains a core diaminopyrimidine ring, a central dimethoxybenzyl ring, and a dihydrophthalazine moiety. We have altered the chemical groups extended from a chiral center on the dihydropyridazine ring of the phthalazine moiety. The interactions for the most potent compounds were visualized by X-ray structure determination. RESULTS: We find that the potency of individual enantiomers is divergent with clear preference for the S-enantiomer, while maintaining a high conservation of contacts within the binding site. The preference for enantiomers seems to be predicated largely by differential interactions with protein residues Leu29, Gln30 and Arg53. CONCLUSIONS: These studies have clarified the activity of modifications and of individual enantiomers, and highlighted the role of the less-active R-enantiomer in effectively diluting the more active S-enantiomer in racemic solutions. This directly contributes to the development of new antimicrobials, combating trimethoprim resistance, and treatment options for potential bioterrorism agents.


Assuntos
Bacillus anthracis/enzimologia , Proteínas de Bactérias/antagonistas & inibidores , Desenho de Fármacos , Antagonistas do Ácido Fólico/química , Simulação de Acoplamento Molecular , Tetra-Hidrofolato Desidrogenase/química , Antraz/tratamento farmacológico , Antraz/enzimologia , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Antagonistas do Ácido Fólico/uso terapêutico , Relação Estrutura-Atividade , Tetra-Hidrofolato Desidrogenase/metabolismo
2.
ChemMedChem ; 7(11): 1974-82, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22930550

RESUMO

(±)-6-Alkyl-2,4-diaminopyrimidine-based inhibitors of bacterial dihydrofolate reductase (DHFR) have been prepared and evaluated for biological potency against Bacillus anthracis and Staphylococcus aureus. Biological studies revealed attenuated activity relative to earlier structures lacking substitution at C6 of the diaminopyrimidine moiety, though minimum inhibitory concentration (MIC) values are in the 0.125-8 µg mL(-1) range for both organisms. This effect was rationalized from three- dimensional X-ray structure studies that indicate the presence of a side pocket containing two water molecules adjacent to the main binding pocket. Because of the hydrophobic nature of the substitutions at C6, the main interactions are with protein residues Leu 20 and Leu 28. These interactions lead to a minor conformational change in the protein, which opens the pocket containing these water molecules such that it becomes continuous with the main binding pocket. These water molecules are reported to play a critical role in the catalytic reaction, highlighting a new area for inhibitor expansion within the limited architectural variation at the catalytic site of bacterial DHFR.


Assuntos
Antibacterianos/farmacologia , Bacillus anthracis/efeitos dos fármacos , Proteínas de Bactérias/metabolismo , Antagonistas do Ácido Fólico/farmacologia , Pirimidinas/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Tetra-Hidrofolato Desidrogenase/metabolismo , Antraz/tratamento farmacológico , Antraz/microbiologia , Antibacterianos/química , Bacillus anthracis/enzimologia , Proteínas de Bactérias/antagonistas & inibidores , Antagonistas do Ácido Fólico/química , Humanos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Pirimidinas/química , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/enzimologia
3.
J Antibiot (Tokyo) ; 65(5): 237-43, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22377538

RESUMO

Der is an essential and widely conserved GTPase that assists assembly of a large ribosomal subunit in bacteria. Der associates specifically with the 50S subunit in a GTP-dependent manner and the cells depleted of Der accumulate the structurally unstable 50S subunit, which dissociates into an aberrant subunit at a lower Mg(2+) concentration. As Der is an essential and ubiquitous protein in bacteria, it may prove to be an ideal cellular target against which new antibiotics can be developed. In the present study, we describe our attempts to identify novel antibiotics specifically targeting Der GTPase. We performed the structure-based design of Der inhibitors using the X-ray crystal structure of Thermotoga maritima Der (TmDer). Virtual screening of commercially available chemical library retrieved 257 small molecules that potentially inhibit Der GTPase activity. These 257 chemicals were tested for their in vitro effects on TmDer GTPase and in vivo antibacterial activities. We identified three structurally diverse compounds, SBI-34462, -34566 and -34612, that are both biologically active against bacterial cells and putative enzymatic inhibitors of Der GTPase homologs. We also presented the possible interactions of each compound with the Der GTP-binding site to understand the mechanism of inhibition. Therefore, our lead compounds inhibiting Der GTPase provide scaffolds for the development of novel antibiotics against antibiotic-resistant pathogenic bacteria.


Assuntos
Antibacterianos/farmacologia , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , GTP Fosfo-Hidrolases/antagonistas & inibidores , Thermotoga maritima/enzimologia , Antibacterianos/química , Sítios de Ligação , Cristalografia por Raios X , Sistemas de Liberação de Medicamentos , Farmacorresistência Bacteriana , GTP Fosfo-Hidrolases/metabolismo , Relação Estrutura-Atividade , Thermotoga maritima/efeitos dos fármacos
4.
Protein Sci ; 14(7): 1870-8, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15937277

RESUMO

We have identified a rare HIV-1 protease (PR) mutation, I47A, associated with a high level of resistance to the protease inhibitor lopinavir (LPV) and with hypersusceptibility to the protease inhibitor saquinavir (SQV). The I47A mutation was found in 99 of 112,198 clinical specimens genotyped after LPV became available in late 2000, but in none of 24,426 clinical samples genotyped from 1998 to October 2000. Phenotypic data obtained for five I47A mutants showed unexpected resistance to LPV (86- to >110-fold) and hypersusceptibility to SQV (0.1- to 0.7-fold). Molecular modeling and energy calculations for these mutants using our structural phenotyping methodology showed an increase in the binding energy of LPV by 1.9-3.1 kcal/mol with respect to the wild type complex, corresponding to a 20- to >100-fold decrease in binding affinity, consistent with the observed high levels of LPV resistance. In the WT PR-LPV complex, the Ile 47 side chain is positioned close to the phenoxyacetyl moiety of LPV and its van der Waals interactions contribute significantly to the ligand binding. These interactions are lost for the smaller Ala 47 residue. Calculated binding energy changes for SQV ranged from -0.4 to -1.2 kcal/mol. In the mutant I47A PR-SQV complexes, the PR flaps are packed more tightly around SQV than in the WT complex, resulting in the formation of additional hydrogen bonds that increase binding affinity of SQV consistent with phenotypic hypersusceptibility. The emergence of mutations at PR residue 47 strongly correlates with increasing prescriptions of LPV (Spearman correlation r(s) = 0.96, P < .0001).


Assuntos
Farmacorresistência Viral/genética , Inibidores da Protease de HIV/farmacologia , Protease de HIV/química , HIV-1/enzimologia , Mutação/genética , Pirimidinonas/farmacologia , Genótipo , Protease de HIV/genética , Protease de HIV/metabolismo , Inibidores da Protease de HIV/química , HIV-1/efeitos dos fármacos , HIV-1/genética , Humanos , Cinética , Lopinavir , Modelos Moleculares , Estrutura Molecular , Fenótipo , Ligação Proteica , Conformação Proteica , Pirimidinonas/química , Saquinavir/química , Saquinavir/farmacologia , Saquinavir/uso terapêutico
5.
Protein Sci ; 12(8): 1706-18, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12876320

RESUMO

Mutations in HIV-1 drug targets lead to resistance and consequent therapeutic failure of antiretroviral drugs. Phenotypic resistance assays are time-consuming and costly, and genotypic rules-based interpretations may fail to predict the effects of multiple mutations. We have developed a computational procedure that rapidly evaluates changes in the binding energy of inhibitors to mutant HIV-1 PR variants. Models of WT complexes were produced from crystal structures. Mutant complexes were built by amino acid substitutions in the WT complexes with subsequent energy minimization of the ligand and PR binding site residues. Accuracy of the models was confirmed by comparison with available crystal structures and by prediction of known resistance-related mutations. PR variants from clinical isolates were modeled in complex with six FDA-approved PIs, and changes in the binding energy (DeltaE(bind)) of mutant versus WT complexes were correlated with the ratios of phenotypic 50% inhibitory concentration (IC(50)) values. The calculated DeltaE(bind) of five PIs showed significant correlations (R(2) = 0.7-0.8) with IC(50) ratios from the Virco Antivirogram assay, and the DeltaE(bind) of six PIs showed good correlation (R(2) = 0.76-0.85) with IC(50) ratios from the Virologic PhenoSense assay. DeltaE(bind) cutoffs corresponding to a four-fold increase in IC(50) were used to define the structure-based phenotype as susceptible, resistant, or equivocal. Blind predictions for 78 PR variants gave overall agreement of 92% (kappa = 0.756) and 86% (kappa = 0.666) with PhenoSense and Antivirogram phenotypes, respectively. The structural phenotyping predicted drug resistance of clinical HIV-1 PR variants with an accuracy approaching that of frequently used cell-based phenotypic assays.


Assuntos
Fármacos Anti-HIV/farmacologia , Farmacorresistência Viral/genética , Inibidores da Protease de HIV/farmacologia , Protease de HIV/química , Protease de HIV/metabolismo , HIV-1/efeitos dos fármacos , HIV-1/enzimologia , Algoritmos , Fármacos Anti-HIV/química , Fármacos Anti-HIV/metabolismo , Desenho de Fármacos , Protease de HIV/genética , Inibidores da Protease de HIV/química , Inibidores da Protease de HIV/metabolismo , HIV-1/genética , Modelos Moleculares , Fenótipo , Conformação Proteica , Relação Estrutura-Atividade
6.
J Comput Aided Mol Des ; 17(8): 495-513, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-14703121

RESUMO

The goals were (1) to understand the driving forces in the binding of small molecule inhibitors to the active site of PTP1B and (2) to develop a molecular mechanics-based empirical free energy function for compound potency prediction. A set of compounds with known activities was docked onto the active site. The related energy components and molecular surface areas were calculated. The bridging water molecules were identified and their contributions were considered. Linear relationships were explored between the above terms and the binding free energies of compounds derived based on experimental inhibition constants. We found that minimally three terms are required to give rise to a good correlation (0.86) with predictive power in five-group cross-validation test (q2 = 0.70). The dominant terms are the electrostatic energy and non-electrostatic energy stemming from the intra- and intermolecular interactions of solutes and from those of bridging water molecules in complexes.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacocinética , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Sítios de Ligação , Calorimetria , Desenho de Fármacos , Inibidores Enzimáticos/química , Modelos Moleculares , Conformação Molecular , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Reprodutibilidade dos Testes , Relação Estrutura-Atividade , Termodinâmica
7.
Drug Discov Today ; 7(24): 1214-20, 2002 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-12547004

RESUMO

Large-scale comparative analysis of drug-target polymorphism structures enables the rational design of next generation 'super drugs'--drugs that are less prone to development of drug resistance or that work for the largest possible fraction of the patient population. Furthermore, knowledge of the drug-target-shape repertoire that exists within the patient population enables predictions of likely clinical trial outcomes and response rates for drug efficacy. This gives information on the optimal drug candidates before the initiation of clinical trials. The economic impact of incorporating pharmacogenomics insights early on in the drug discovery process will be substantial and will afford significant competitive advantages to companies that successfully incorporate this technology.


Assuntos
Fármacos Anti-HIV/síntese química , Desenho de Fármacos , Farmacorresistência Viral Múltipla , Farmacogenética/métodos , Animais , Farmacorresistência Viral Múltipla/fisiologia , Humanos , Modelos Químicos , Modelos Moleculares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...