Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Acta Crystallogr F Struct Biol Commun ; 76(Pt 4): 192-193, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32254053

RESUMEN

The true identity of the protein found in the crystals reported by Bondoc et al. [(2019), Acta Cryst. F75, 646-651] is given.

2.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 10): 646-651, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31584013

RESUMEN

Acyl carrier proteins (ACPs) are important components in fatty-acid biosynthesis in prokaryotes. Rv0100 is predicted to be an essential ACP in Mycobacterium tuberculosis, the pathogen that is the causative agent of tuberculosis, and therefore has the potential to be a novel antituberculosis drug target. Here, the successful cloning and purification of Rv0100 using Mycobacterium smegmatis as a host is reported. Crystals of the purified protein were obtained that diffracted to a resolution of 1.9 Å. Overall, this work lays the foundation for the future pursuit of drug discovery and development against this potentially novel drug target.


Asunto(s)
Proteína Transportadora de Acilo/química , Proteínas Bacterianas/química , Cristalización , Mycobacterium tuberculosis/química , Proteína Transportadora de Acilo/genética , Proteína Transportadora de Acilo/aislamiento & purificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Cristalografía por Rayos X , Mycobacterium smegmatis/genética , Mycobacterium tuberculosis/genética , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación
3.
ACS Omega ; 3(10): 13876-13881, 2018 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-30411052

RESUMEN

Inositol monophosphatase (IMPase) is a crucial enzyme for the biosynthesis of phosphatidylinositol, an essential component in mycobacterial cell walls. IMPase A (ImpA) from Mycobacterium smegmatis is a bifunctional enzyme that also functions as a fructose-1,6-bisphosphatase (FBPase). To better understand the bifunctional nature of this enzyme, point mutagenesis was conducted on several key residues and their enzyme activity was tested. Our results along with active site models support the fact that ImpA is a bifunctional enzyme with residues Gly94, Thr95 hypothesized to be contributing to the FBPase activity and residues Trp220, Asp221 hypothesized to be contributing to the IMPase activity. Double mutants, W220A + D221A reduced both FBPase and IMPase activity drastically while the double mutant G94A + T95A surprisingly partially restored the IMPase activity compared to the single mutants. This study establishes the foundation toward obtaining a better understanding of the bifunctional nature of this enzyme.

4.
Bioorg Med Chem Lett ; 28(11): 2074-2079, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29730028

RESUMEN

We have previously reported benzimidazole-based compounds to be potent inhibitors of FabI for Francisella tularensis (FtFabI), making them promising antimicrobial hits. Optically active enantiomers exhibit markedly differing affinities toward FtFabI. The IC50 of benzimidazole (-)-1 is ∼100× lower than the (+)-enantiomer, with similar results for the 2 enantiomers. Determining the absolute configuration for these optical compounds and elucidating their binding modes is important for further design. Electronic circular dichroism (ECD) quantum calculations have become important in determining absolute configurations of optical compounds. We determined the absolute configuration of (-)/(+)-1 and (-)/(+)-2 by comparing experimental spectra and theoretical density functional theory (DFT) simulations of ECD spectra at the B3LYP/6-311+G(2d, p) level using Gaussian09. Comparison of experimental and calculated ECD spectra indicates that the S configuration corresponds to the (-)-rotation for both compounds 1 and 2, while the R configuration corresponds to the (+)-rotation. Further, molecular dynamics simulations and MM-GBSA binding energy calculations for these two pairs of enantiomers with FtFabI show much tighter binding MM-GBSA free energies for S-1 and S-2 than for their enantiomers, R-1 and R-2, consistent with the S configuration being the more active one, and with the ECD determination of the S configuration corresponding to (-) and the R configuration corresponding to (+). Thus, our computational studies allow us to assign (-) to (S)- and (+) to (R)- for compounds 1 and 2, and to further evaluate structural changes to improve efficacy.


Asunto(s)
Antibacterianos/farmacología , Bencimidazoles/farmacología , Enoil-CoA Hidratasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Francisella tularensis/efectos de los fármacos , Teoría Cuántica , Antibacterianos/química , Bencimidazoles/química , Sitios de Unión/efectos de los fármacos , Dicroismo Circular , Relación Dosis-Respuesta a Droga , Enoil-CoA Hidratasa/metabolismo , Inhibidores Enzimáticos/química , Francisella tularensis/enzimología , Enlace de Hidrógeno , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Estructura Molecular , Relación Estructura-Actividad
5.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 2): 105-112, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29400320

RESUMEN

Enoyl-acyl carrier protein (ACP) reductase II (FabK) is a critical rate-limiting enzyme in the bacterial type II fatty-acid synthesis (FAS II) pathway. FAS II pathway enzymes are markedly disparate from their mammalian analogs in the FAS I pathway in both structure and mechanism. Enzymes involved in bacterial fatty-acid synthesis represent viable drug targets for Gram-negative pathogens, and historical precedent exists for targeting them in the treatment of diseases of the oral cavity. The Gram-negative organism Porphyromonas gingivalis represents a key causative agent of the costly and highly prevalent disease known as chronic periodontitis, and exclusively expresses FabK as its enoyl reductase enzyme in the FAS-II pathway. Together, these characteristics distinguish P. gingivalis FabK (PgFabK) as an attractive and novel narrow-spectrum antibacterial target candidate. PgFabK is a flavoenzyme that is dependent on FMN and NADPH as cofactors for the enzymatic reaction, which reduces the enoyl substrate via a ping-pong mechanism. Here, the structure of the PgFabK enzyme as determined using X-ray crystallography is reported to 1.9 Šresolution with endogenous FMN fully resolved and the NADPH cofactor partially resolved. PgFabK possesses a TIM-barrel motif, and all flexible loops are visible. The determined structure has allowed insight into the structural basis for the NADPH dependence observed in PgFabK and the role of a monovalent cation that has been observed in previous studies to be stringently required for FabK activity. The PgFabK structure and the insights gleaned from its analysis will facilitate structure-based drug-discovery efforts towards the prevention and treatment of P. gingivalis infection.


Asunto(s)
Enoil-ACP Reductasa (NADH)/química , Enoil-ACP Reductasa (NADH)/genética , Porphyromonas gingivalis/enzimología , Porphyromonas gingivalis/genética , Secuencia de Aminoácidos , Línea Celular , Enoil-ACP Reductasa (NADH)/aislamiento & purificación , Humanos , Estructura Secundaria de Proteína , Difracción de Rayos X/métodos
6.
Bioorg Med Chem ; 26(1): 65-76, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29162308

RESUMEN

S. aureus and A. baumannii are among the ESKAPE pathogens that are increasingly difficult to treat due to the rise in the number of drug resistant strains. Novel therapeutics targeting these pathogens are much needed. The bacterial enoyl reductase (FabI) is as potentially significant drug target for developing pathogen-specific antibiotics due to the presence of alternate FabI isoforms in many other bacterial species. We report the identification and development of a novel N-carboxy pyrrolidine scaffold targeting FabI in S. aureus and A. baumannii, two pathogens for which FabI essentiality has been established. This scaffold is unrelated to other known antibiotic families, and FabI is not targeted by any currently approved antibiotic. Our data shows that this scaffold displays promising enzyme inhibitory activity against FabI from both S. aureus and A. baumannii, as well as encouraging antibacterial activity in S. aureus. Compounds also display excellent synergy when combined with colistin and tested against A. baumannii. In this combination the MIC of colistin is reduced by 10-fold. Our first generation compound displays promising enzyme inhibition, targets FabI in S. aureus with a favorable selectivity index (ratio of cytotoxicity to MIC), and has excellent synergy with colistin against A. baumannii, including a multidrug resistant strain.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/síntesis química , Antibacterianos/química , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Enoil-ACP Reductasa (NADH)/metabolismo , Escherichia coli/efectos de los fármacos , Células Hep G2 , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Relación Estructura-Actividad
7.
ACS Infect Dis ; 3(1): 54-61, 2017 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-27756129

RESUMEN

The enoyl-ACP reductase (FabI) enzyme is a well validated target for anti-staphylococcal drug discovery and development. With the goal of finding alternate therapeutics for drug-resistant strains of Staphylococcus aureus, such as methicillin-resistant S. aureus (MRSA), our previously published series of benzimidazole-based inhibitors of the FabI enzyme from Francisella tularensis (FtFabI) have been evaluated against FabI from S. aureus (SaFabI). We report here the preliminary structure-activity relationship of this series and the prioritization of compounds toward lead optimization. Mutational studies have identified key residues that contribute toward stabilizing the inhibitors in the active site of FabI. Mutations that do not significantly impact enzyme function but destabilize inhibitor binding are more likely to occur in nature as organisms evolve to evade the action of antibiotics leading to resistance. Identifying these residues provides guidance for minimizing susceptibility to resistance. Additionally, we have identified compounds that elicit antibacterial activity through off-target effects and observe that close analogs can display differing modes of action (on-target vs off-target) and need to be individually evaluated early on to prioritize compounds for lead optimization. Overall, our data suggest that the benzimidazole scaffold is a promising scaffold for anti-staphylococcal drug development.


Asunto(s)
Antibacterianos/farmacología , Bencimidazoles/química , Bencimidazoles/farmacología , Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Staphylococcus aureus/enzimología , Clonación Molecular , Diseño de Fármacos , Descubrimiento de Drogas , Enoil-ACP Reductasa (NADH)/genética , Enoil-ACP Reductasa (NADH)/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Concentración 50 Inhibidora , Estructura Molecular , Staphylococcus aureus/efectos de los fármacos , Relación Estructura-Actividad
8.
J Comput Chem ; 36(25): 1859-73, 2015 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-26216222

RESUMEN

To validate a method for predicting the binding affinities of FabI inhibitors, three implicit solvent methods, MM-PBSA, MM-GBSA, and QM/MM-GBSA were carefully compared using 16 benzimidazole inhibitors in complex with Francisella tularensis FabI. The data suggests that the prediction results are sensitive to radii sets, GB methods, QM Hamiltonians, sampling protocols, and simulation length, if only one simulation trajectory is used for each ligand. In this case, QM/MM-GBSA using 6 ns MD simulation trajectories together with GB(neck2) , PM3, and the mbondi2 radii set, generate the closest agreement with experimental values (r(2) = 0.88). However, if the three implicit solvent methods are averaged from six 1 ns MD simulations for each ligand (called "multiple independent sampling"), the prediction results are relatively insensitive to all the tested parameters. Moreover, MM/GBSA together with GB(HCT) and mbondi, using 600 frames extracted evenly from six 0.25 ns MD simulations, can also provide accurate prediction to experimental values (r(2) = 0.84). Therefore, the multiple independent sampling method can be more efficient than a single, long simulation method. Since future scaffold expansions may significantly change the benzimidazole's physiochemical properties (charges, etc.) and possibly binding modes, which may affect the sensitivities of various parameters, the relatively insensitive "multiple independent sampling method" may avoid the need of an entirely new validation study. Moreover, due to large fluctuating entropy values, (QM/)MM-P(G)BSA were limited to inhibitors' relative affinity prediction, but not the absolute affinity. The developed protocol will support an ongoing benzimidazole lead optimization program.


Asunto(s)
Bencimidazoles/química , Bencimidazoles/farmacología , Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Francisella tularensis/enzimología , Diseño de Fármacos , Enoil-ACP Reductasa (NADH)/metabolismo , Entropía , Francisella tularensis/química , Humanos , Ligandos , Simulación de Dinámica Molecular , Unión Proteica , Tularemia/tratamiento farmacológico , Tularemia/microbiología
9.
AMB Express ; 5(1): 125, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26112315

RESUMEN

The I100V isoform of toluene-4-monooxygenase was used to catalyze the oxidative polymerization of anthranil and various indoles under mildly acidic conditions, favoring the production of trimers. Compounds produced in sufficient yield were purified and tested for their ability to inhibit the growth of B. anthracis, E. faecalis, L. monocytogenes, S. aureus, and in some cases, F. tularensis. 15 of the compounds displayed promising antibacterial activity (MIC < 5 µg/ml) against one or more of the strains tested, with the best MIC values being <0.8 µg/ml. All of these compounds had good selectivity, showing minimal cytotoxicity towards HepG2 cells. The structure was solved for six of the compounds that could be crystallized, revealing that minimally two classes of indole based trimers were produced. One compound class produced was a group of substituted derivatives of the natural product 2,2-bis(3-indolyl) indoxyl. The other group of compounds identified was classified as tryptanthrin-like compounds, all having multi-ring pendant groups attached at position 11 of tryptanthrin. One compound of particular interest, SAB-J85, had a structure that suggests that any compound, with a ring structure that can be activated by an oxygenase, might serve as a substrate for combinatorial biocatalysis.

10.
Bioorg Med Chem Lett ; 25(6): 1292-6, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25677657

RESUMEN

Francisella tularensis, the causative agent of tularemia, presents a significant biological threat and is a Category A priority pathogen due to its potential for weaponization. The bacterial FASII pathway is a viable target for the development of novel antibacterial agents treating Gram-negative infections. Here we report the advancement of a promising series of benzimidazole FabI (enoyl-ACP reductase) inhibitors to a second-generation using a systematic, structure-guided lead optimization strategy, and the determination of several co-crystal structures that confirm the binding mode of designed inhibitors. These compounds display an improved low nanomolar enzymatic activity as well as promising low microgram/mL antibacterial activity against both F. tularensis and Staphylococcus aureus and its methicillin-resistant strain (MRSA). The improvements in activity accompanying structural modifications lead to a better understanding of the relationship between the chemical structure and biological activity that encompasses both enzymatic and whole-cell activity.


Asunto(s)
Antibacterianos/química , Bencimidazoles/química , Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Francisella tularensis/enzimología , Antibacterianos/síntesis química , Antibacterianos/farmacología , Bencimidazoles/síntesis química , Bencimidazoles/farmacología , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Enoil-ACP Reductasa (NADH)/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Escherichia coli/efectos de los fármacos , Francisella tularensis/efectos de los fármacos , Cinética , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Staphylococcus aureus/efectos de los fármacos , Relación Estructura-Actividad
11.
Methods Mol Biol ; 1140: 291-304, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24590725

RESUMEN

An important aspect of enzymatic assays is that the effect of a ligand on enzyme activity is readily apparent and quantifiable. Enzyme-based assays are, therefore, highly amenable to high-throughput ligand screening, which profiles the effect of a panel of small molecules on a designated target. In order for enzyme assays to provide useful screening data, the kinetics, assay components, readout signal, and overall stability of the assay are optimized and adapted to the equipment prior to the screen. For the screen itself, careful consideration is given to the number of replicates, the plate layout, the compound concentration, and the details of assay assembly. Lastly, in the post-screen stages, the ligand screening data is processed and analyzed using various strategies, and the resulting preliminary hits are subjected to a series of secondary and tertiary assays to eliminate false positives and poor quality hits. The various stages of screening are described, using a viral protease, NS3/4A from Hepatitis C virus, as an example of an enzyme target.


Asunto(s)
Hepacivirus/enzimología , Ensayos Analíticos de Alto Rendimiento/métodos , Biología Molecular/métodos , Genómica , Hepacivirus/química , Humanos , Cinética , Ligandos , Bibliotecas de Moléculas Pequeñas/química
12.
Xenobiotica ; 44(5): 404-16, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24171690

RESUMEN

1. FabI is a potential antibiotic target against Francisella tularensis, which has been classified as a Category A biowarfare agent of high risk to public health. Our previous work demonstrated that N-benzyl benzimidazole compounds possess promising FabI inhibitory activity, but their druggability properties, including metabolic stability, are unknown. 2. The objective of this study was to characterize structure-metabolism relationships of a series of N-benzyl benzimidazole compounds to guide chemical optimization for better metabolic stability. To this end, metabolic stability data were obtained for 22 initial lead compounds using mouse hepatic microsomes. 3. Metabolic hotspots on the benzimidazole core structure as well as the benzyl ring were identified and verified by metabolite identification studies of four model compounds. Interestingly, the proposed structure-metabolism relationships did not apply to nine newly synthesized cyclopentane or oxacyclopentane derivatives of N-benzyl benzimidazole. 4. Subsequently, in silico quantitative structure-property relationship models were developed. Four molecular descriptors representing molecular polarity/polarisability, symmetry and size were identified to best explain variability in metabolic stability of different compounds. Multi-linear and non-linear regression models based on the selected molecular descriptors were developed and validated. 5. The structure-metabolism relationships for N-benzyl benzimidazole compounds should help optimization of N-benzyl benzimidazole compounds for better pharmacokinetic behavior.


Asunto(s)
Bencimidazoles/metabolismo , Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Francisella tularensis/enzimología , Relación Estructura-Actividad Cuantitativa , Animales , Antibacterianos/química , Bencimidazoles/química , Proteínas Sanguíneas/metabolismo , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/metabolismo , Femenino , Ratones , Ratones Endogámicos BALB C
13.
Appl Microbiol Biotechnol ; 97(16): 7151-63, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23749163

RESUMEN

Two bacterial hosts expressing cloned aromatic oxygenases were used to catalyze the oxidation and polymerization of indole and related substrates, creating mixtures of indigoid compounds comprised of novel dimers and trimers. Crude extracts and purified compounds were tested for their ability to inhibit the growth of Gram-positive organisms, in general, and Mycobacterium tuberculosis (TB), in particular. Of the 74 compounds tested against M. tuberculosis, ~66 % had minimum inhibitory concentrations (MIC) of 5 µg/ml or less. The most effective antibiotic found was designated SAB-P1, a heterodimer of indole and anthranil, which had a MIC of 0.16 µg/ml, and did not inhibit kidney cells (IC(50)) at concentrations of >8 µg/ml. Combinatorial biocatalysis was used to create a series of halogenated derivatives of SAB-P1 with a wider therapeutic window. None of the derivatives had MIC values that were superior to SAB-P1, but some had a wider therapeutic window because of decreased kidney cell toxicity. Generally, the indigoid dimers that were effective against TB appeared to be specific for TB. Some of the trimers generated, however, had a broader spectrum of activity inhibiting not only TB (MIC = 1.1 µg/ml) but also the growth of Mycobacterium smegmatis MC2 155, Bacillus cereus, Enterococcus faecalis, Staphylococcus epidermidis, Bacillus subtilis 168, and Clostridium acetobutylicum. The structure of two of the novel dimers (SAB-C4 and SAB-P1) and a trimer (SAB-R1) were solved using X-ray crystallography.


Asunto(s)
Antituberculosos/metabolismo , Indoles/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Antituberculosos/aislamiento & purificación , Antituberculosos/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Humanos , Indoles/aislamiento & purificación , Indoles/toxicidad , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana
14.
Protein Expr Purif ; 85(1): 100-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22820244

RESUMEN

The rapid rise in bacterial drug resistance coupled with the low number of novel antimicrobial compounds in the discovery pipeline has led to a critical situation requiring the expedient discovery and characterization of new antimicrobial drug targets. Enzymes in the bacterial fatty acid synthesis pathway, FAS-II, are distinct from their mammalian counterparts, FAS-I, in terms of both structure and mechanism. As such, they represent attractive targets for the design of novel antimicrobial compounds. Enoyl-acyl carrier protein reductase II, FabK, is a key, rate-limiting enzyme in the FAS-II pathway for several bacterial pathogens. The organism, Porphyromonas gingivalis, is a causative agent of chronic periodontitis that affects up to 25% of the US population and incurs a high national burden in terms of cost of treatment. P. gingivalis expresses FabK as the sole enoyl reductase enzyme in its FAS-II cycle, which makes this a particularly appealing target with potential for selective antimicrobial therapy. Herein we report the molecular cloning, expression, purification and characterization of the FabK enzyme from P. gingivalis, only the second organism from which this enzyme has been isolated. Characterization studies have shown that the enzyme is a flavoprotein, the reaction dependent upon FMN and NADPH and proceeding via a Ping-Pong Bi-Bi mechanism to reduce the enoyl substrate. A sensitive assay measuring the fluorescence decrease of NADPH as it is converted to NADP(+) during the reaction has been optimized for high-throughput screening. Finally, protein crystallization conditions have been identified which led to protein crystals that diffract x-rays to high resolution.


Asunto(s)
Clonación Molecular , Enoil-ACP Reductasa (NADH)/genética , Enoil-ACP Reductasa (NADH)/metabolismo , Porphyromonas gingivalis/enzimología , Cristalización , Cristalografía por Rayos X , Enoil-ACP Reductasa (NADH)/química , Enoil-ACP Reductasa (NADH)/aislamiento & purificación , Mononucleótido de Flavina/metabolismo , Cinética , NADP/metabolismo , Porphyromonas gingivalis/química , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
15.
J Med Chem ; 55(12): 5933-41, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22642319

RESUMEN

Because of structural and mechanistic differences between eukaryotic and prokaryotic fatty acid synthesis enzymes, the bacterial pathway, FAS-II, is an attractive target for the design of antimicrobial agents. We have previously reported the identification of a novel series of benzimidazole compounds with particularly good antibacterial effect against Francisella tularensis, a Category A biowarfare pathogen. Herein we report the crystal structure of the F. tularensis FabI enzyme in complex with our most active benzimidazole compound bound with NADH. The structure reveals that the benzimidazole compounds bind to the substrate site in a unique conformation that is distinct from the binding motif of other known FabI inhibitors. Detailed inhibition kinetics have confirmed that the compounds possess a novel inhibitory mechanism that is unique among known FabI inhibitors. These studies could have a strong impact on future antimicrobial design efforts and may reveal new avenues for the design of FAS-II active antibacterial compounds.


Asunto(s)
Antibacterianos/metabolismo , Antibacterianos/farmacología , Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Enoil-ACP Reductasa (NADH)/metabolismo , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Francisella tularensis/enzimología , Antibacterianos/síntesis química , Antibacterianos/química , Bencimidazoles/síntesis química , Bencimidazoles/química , Bencimidazoles/metabolismo , Bencimidazoles/farmacología , Dominio Catalítico , Enoil-ACP Reductasa (NADH)/química , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Francisella tularensis/efectos de los fármacos , Concentración 50 Inhibidora , Cinética , Modelos Moleculares , NAD/metabolismo , Unión Proteica , Relación Estructura-Actividad
16.
J Med Chem ; 55(1): 268-79, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-22098466

RESUMEN

Enoyl-acyl carrier protein (ACP) reductase, FabI, is a key enzyme in the bacterial fatty acid biosynthesis pathway (FAS II). FabI is an NADH-dependent oxidoreductase that acts to reduce enoyl-ACP substrates in a final step of the pathway. The absence of this enzyme in humans makes it an attractive target for the development of new antibacterial agents. FabI is known to be unresponsive to structure-based design efforts due to a high degree of induced fit and a mobile flexible loop encompassing the active site. Here we discuss the development, validation, and careful application of a ligand-based virtual screen used for the identification of novel inhibitors of the Francisella tularensis FabI target. In this study, four known classes of FabI inhibitors were used as templates for virtual screens that involved molecular shape and electrostatic matching. The program ROCS was used to search a high-throughput screening library for compounds that matched any of the four molecular shape queries. Matching compounds were further refined using the program EON, which compares and scores compounds by matching electrostatic properties. Using these techniques, 50 compounds were selected, ordered, and tested. The tested compounds possessed novel chemical scaffolds when compared to the input query compounds. Several hits with low micromolar activity were identified and follow-up scaffold-based searches resulted in the identification of a lead series with submicromolar enzyme inhibition, high ligand efficiency, and a novel scaffold. Additionally, one of the most active compounds showed promising whole-cell antibacterial activity against several Gram-positive and Gram-negative species, including the target pathogen. The results of a preliminary structure-activity relationship analysis are presented.


Asunto(s)
Antibacterianos/síntesis química , Bencimidazoles/síntesis química , Compuestos de Bencilo/síntesis química , Enoil-ACP Reductasa (NADPH Específica B)/antagonistas & inhibidores , Francisella tularensis/enzimología , Modelos Moleculares , Antibacterianos/química , Antibacterianos/farmacología , Bencimidazoles/química , Bencimidazoles/farmacología , Compuestos de Bencilo/química , Compuestos de Bencilo/farmacología , Bases de Datos Factuales , Francisella tularensis/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Conformación Molecular , Electricidad Estática , Relación Estructura-Actividad
17.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 66(Pt 11): 1432-5, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21045288

RESUMEN

Dihydroorotase (EC 3.5.2.3) catalyzes the reversible cyclization of N-carbamoyl-L-aspartate to L-dihydroorotate in the third step of the pyrimidine-biosynthesis pathway in Bacillus anthracis. A comparison is made between the structures of dihydroorotase from four different organisms, including B. anthracis dihydroorotase, and reveals substantial variations in the active site, dimer interface and overall tertiary structure. These differences demonstrate the utility of exploring multiple structures of a molecular target as expressed from different organisms and how these differences can be exploited for structure-based drug discovery.


Asunto(s)
Bacillus anthracis/enzimología , Dihidroorotasa/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia , Homología Estructural de Proteína
18.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 66(Pt 11): 1436-40, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21045289

RESUMEN

Enoyl-acyl carrier protein reductase (FabI) catalyzes the last rate-limiting step in the elongation cycle of the fatty-acid biosynthesis pathway and has been validated as a potential antimicrobial drug target in Francisella tularensis. The development of new antibiotic therapies is important both to combat potential drug-resistant bioweapons and to address the broader societal problem of increasing antibiotic resistance among many pathogenic bacteria. The crystal structure of FabI from F. tularensis (FtuFabI) in complex with the inhibitor triclosan and the cofactor NAD(+) has been solved to a resolution of 2.1 Å. Triclosan is known to effectively inhibit FabI from different organisms. Precise characterization of the mode of triclosan binding is required to develop highly specific inhibitors. Comparison of our structure with the previously determined FtuFabI structure (PDB code 2jjy) which is bound to only NAD(+) reveals the conformation of the substrate-binding loop, electron density for which was missing in the earlier structure, and demonstrates a shift in the conformation of the NAD(+) cofactor. This shift in the position of the phosphate groups allows more room in the active site for substrate or inhibitor to bind and be better accommodated. This information will be crucial for virtual screening studies to identify novel scaffolds for development into new active inhibitors.


Asunto(s)
Enoil-ACP Reductasa (NADH)/química , Inhibidores Enzimáticos/química , Francisella tularensis/enzimología , NAD/química , Triclosán/química , Cristalografía por Rayos X , Enoil-ACP Reductasa (NADH)/metabolismo , Inhibidores Enzimáticos/metabolismo , Modelos Moleculares , NAD/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Homología Estructural de Proteína , Triclosán/metabolismo
19.
J Biol Chem ; 285(19): 14572-84, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20228407

RESUMEN

We have solved the crystal structure of a segment of nonerythroid alpha-spectrin (alphaII) consisting of the first 147 residues to a resolution of 2.3 A. We find that the structure of this segment is generally similar to a corresponding segment from erythroid alpha-spectrin (alphaI) but exhibits unique differences with functional significance. Specific features include the following: (i) an irregular and frayed first helix (Helix C'); (ii) a helical conformation in the junction region connecting Helix C' with the first structural domain (D1); (iii) a long A(1)B(1) loop in D1; and (iv) specific inter-helix hydrogen bonds/salt bridges that stabilize D1. Our findings suggest that the hydrogen bond networks contribute to structural domain stability, and thus rigidity, in alphaII, and the lack of such hydrogen bond networks in alphaI leads to flexibility in alphaI. We have previously shown the junction region connecting Helix C' to D1 to be unstructured in alphaI (Park, S., Caffrey, M. S., Johnson, M. E., and Fung, L. W. (2003) J. Biol. Chem. 278, 21837-21844) and now find it to be helical in alphaII, an important difference for alpha-spectrin association with beta-spectrin in forming tetramers. Homology modeling and molecular dynamics simulation studies of the structure of the tetramerization site, a triple helical bundle of partial domain helices, show that mutations in alpha-spectrin will affect Helix C' structural flexibility and/or the junction region conformation and may alter the equilibrium between spectrin dimers and tetramers in cells. Mutations leading to reduced levels of functional tetramers in cells may potentially lead to abnormal neuronal functions.


Asunto(s)
Encéfalo/metabolismo , Células Eritroides/metabolismo , Multimerización de Proteína , Proteínas Recombinantes/química , Espectrina/química , Cristalización , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Mutación/genética , Estructura Secundaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrina/genética , Espectrina/metabolismo
20.
Biochemistry ; 48(29): 7045-55, 2009 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-19552402

RESUMEN

Glutamate racemase (RacE) is a bacterial enzyme that converts l-glutamate to d-glutamate, an essential precursor for peptidoglycan synthesis. In prior work, we have shown that both isoforms cocrystallize with d-glutamate as dimers, and the enzyme is in a closed conformation with limited access to the active site [May, M., et al. (2007) J. Mol. Biol. 371, 1219-1237]. The active site of RacE2 is especially restricted. We utilize several computational and experimental approaches to understand the overall conformational dynamics involved during catalysis when the ligand enters and the product exits the active site. Our steered molecular dynamics simulations and normal-mode analysis results indicate that the monomeric form of the enzyme is more flexible than the native dimeric form. These results suggest that the monomeric enzyme might be more active than the dimeric form. We thus generated site-specific mutations that disrupt dimerization and find that the mutants exhibit significantly higher catalytic rates in the d-Glu to l-Glu reaction direction than the native enzyme. Low-resolution models restored from solution X-ray scattering studies correlate well with the first six normal modes of the dimeric form of the enzyme, obtained from NMA. Thus, along with the local active site residues, global domain motions appear to be implicated in the catalytically relevant structural dynamics of this enzyme and suggest that increased flexibility may accelerate catalysis. This is a novel observation that residues distant from the catalytic site restrain catalytic activity through formation of the dimer structure.


Asunto(s)
Isomerasas de Aminoácido/metabolismo , Isomerasas de Aminoácido/química , Isomerasas de Aminoácido/genética , Biocatálisis , Cromatografía en Gel , Dimerización , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Dispersión de Radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...