Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 2): 105-112, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29400320

ABSTRACT

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.


Subject(s)
Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/chemistry , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/genetics , Amino Acid Sequence , Cell Line , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/isolation & purification , Humans , Protein Structure, Secondary , X-Ray Diffraction/methods
2.
Bioorg Med Chem Lett ; 25(6): 1292-6, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25677657

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/chemistry , Benzimidazoles/chemistry , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Francisella tularensis/enzymology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Francisella tularensis/drug effects , Kinetics , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Staphylococcus aureus/drug effects , Structure-Activity Relationship
3.
Xenobiotica ; 44(5): 404-16, 2014 May.
Article in English | MEDLINE | ID: mdl-24171690

ABSTRACT

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.


Subject(s)
Benzimidazoles/metabolism , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Francisella tularensis/enzymology , Quantitative Structure-Activity Relationship , Animals , Anti-Bacterial Agents/chemistry , Benzimidazoles/chemistry , Blood Proteins/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/metabolism , Female , Mice , Mice, Inbred BALB C
4.
Protein Expr Purif ; 85(1): 100-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22820244

ABSTRACT

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.


Subject(s)
Cloning, Molecular , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Porphyromonas gingivalis/enzymology , Crystallization , Crystallography, X-Ray , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/chemistry , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/isolation & purification , Flavin Mononucleotide/metabolism , Kinetics , NADP/metabolism , Porphyromonas gingivalis/chemistry , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
5.
J Med Chem ; 55(12): 5933-41, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22642319

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Francisella tularensis/enzymology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Catalytic Domain , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Francisella tularensis/drug effects , Inhibitory Concentration 50 , Kinetics , Models, Molecular , NAD/metabolism , Protein Binding , Structure-Activity Relationship
6.
J Med Chem ; 55(1): 268-79, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22098466

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Benzimidazoles/chemical synthesis , Benzyl Compounds/chemical synthesis , Enoyl-(Acyl-Carrier Protein) Reductase (NADPH, B-Specific)/antagonists & inhibitors , Francisella tularensis/enzymology , Models, Molecular , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Benzyl Compounds/chemistry , Benzyl Compounds/pharmacology , Databases, Factual , Francisella tularensis/drug effects , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Microbial Sensitivity Tests , Molecular Conformation , Static Electricity , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...