Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 19052, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37923786

ABSTRACT

The Streptococcus pyogenes cell envelope protease (SpyCEP) is vital to streptococcal pathogenesis and disease progression. Despite its strong association with invasive disease, little is known about enzymatic function beyond the ELR+ CXC chemokine substrate range. As a serine protease, SpyCEP has a catalytic triad consisting of aspartate (D151), histidine (H279), and serine (S617) residues which are all thought to be mandatory for full activity. We utilised a range of SpyCEP constructs to investigate the protein domains and catalytic residues necessary for enzyme function. We designed a high-throughput mass spectrometry assay to measure CXCL8 cleavage and applied this for the first time to study the enzyme kinetics of SpyCEP. Results revealed a remarkably low Michaelis-Menton constant (KM) of 82 nM and a turnover of 1.65 molecules per second. We found that an N-terminally-truncated SpyCEP C-terminal construct containing just the catalytic dyad of H279 and S617 was capable of cleaving CXCL8 with a similar KM of 55 nM, albeit with a reduced substrate turnover of 2.7 molecules per hour, representing a 2200-fold reduction in activity. We conclude that the SpyCEP C-terminus plays a key role in high affinity substrate recognition and binding, but that the N-terminus is required for full catalytic activity.


Subject(s)
Peptide Hydrolases , Streptococcus pyogenes , Streptococcus pyogenes/metabolism , Peptide Hydrolases/metabolism , Protein Domains
2.
J Med Chem ; 66(15): 10413-10431, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37506194

ABSTRACT

There is an urgent need for new treatments for Chagas disease, a parasitic infection which mostly impacts South and Central America. We previously reported on the discovery of GSK3494245/DDD01305143, a preclinical candidate for visceral leishmaniasis which acted through inhibition of the Leishmania proteasome. A related analogue, active against Trypanosoma cruzi, showed suboptimal efficacy in an animal model of Chagas disease, so alternative proteasome inhibitors were investigated. Screening a library of phenotypically active analogues against the T. cruzi proteasome identified an active, selective pyridazinone, the development of which is described herein. We obtained a cryo-EM co-structure of proteasome and a key inhibitor and used this to drive optimization of the compounds. Alongside this, optimization of the absorption, distribution, metabolism, and excretion (ADME) properties afforded a suitable compound for mouse efficacy studies. The outcome of these studies is discussed, alongside future plans to further understand the series and its potential to deliver a new treatment for Chagas disease.


Subject(s)
Chagas Disease , Leishmaniasis, Visceral , Trypanocidal Agents , Trypanosoma cruzi , Mice , Animals , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Proteasome Endopeptidase Complex , Chagas Disease/drug therapy , Chagas Disease/parasitology , Leishmaniasis, Visceral/drug therapy , Trypanocidal Agents/pharmacology , Trypanocidal Agents/therapeutic use , Trypanocidal Agents/chemistry
3.
Nat Commun ; 6: 10048, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26640131

ABSTRACT

New antibacterials are needed to tackle antibiotic-resistant bacteria. Type IIA topoisomerases (topo2As), the targets of fluoroquinolones, regulate DNA topology by creating transient double-strand DNA breaks. Here we report the first co-crystal structures of the antibacterial QPT-1 and the anticancer drug etoposide with Staphylococcus aureus DNA gyrase, showing binding at the same sites in the cleaved DNA as the fluoroquinolone moxifloxacin. Unlike moxifloxacin, QPT-1 and etoposide interact with conserved GyrB TOPRIM residues rationalizing why QPT-1 can overcome fluoroquinolone resistance. Our data show etoposide's antibacterial activity is due to DNA gyrase inhibition and suggests other anticancer agents act similarly. Analysis of multiple DNA gyrase co-crystal structures, including asymmetric cleavage complexes, led to a 'pair of swing-doors' hypothesis in which the movement of one DNA segment regulates cleavage and religation of the second DNA duplex. This mechanism can explain QPT-1's bacterial specificity. Structure-based strategies for developing topo2A antibacterials are suggested.


Subject(s)
Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , DNA Gyrase/chemistry , Etoposide/chemistry , Fluoroquinolones/chemistry , Staphylococcus aureus/enzymology , Topoisomerase II Inhibitors/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , DNA Gyrase/genetics , DNA Gyrase/metabolism , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Drug Resistance, Bacterial , Etoposide/pharmacology , Fluoroquinolones/pharmacology , Models, Molecular , Molecular Structure , Moxifloxacin , Staphylococcus aureus/chemistry , Staphylococcus aureus/drug effects , Topoisomerase II Inhibitors/pharmacology
4.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 10): 1242-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26457513

ABSTRACT

Fluoroquinolone drugs such as moxifloxacin kill bacteria by stabilizing the normally transient double-stranded DNA breaks created by bacterial type IIA topoisomerases. Previous crystal structures of Staphylococcus aureus DNA gyrase with asymmetric DNAs have had static disorder (with the DNA duplex observed in two orientations related by the pseudo-twofold axis of the complex). Here, 20-base-pair DNA homoduplexes were used to obtain crystals of covalent DNA-cleavage complexes of S. aureus DNA gyrase. Crystals with QPT-1, moxifloxacin or etoposide diffracted to between 2.45 and 3.15 Šresolution. A G/T mismatch introduced at the ends of the DNA duplexes facilitated the crystallization of slightly asymmetric complexes of the inherently flexible DNA-cleavage complexes.


Subject(s)
DNA Cleavage , DNA Gyrase/chemistry , Etoposide/chemistry , Fluoroquinolones/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Spiro Compounds/chemistry , Staphylococcus aureus/enzymology , Base Sequence , Crystallization , Crystallography, X-Ray , Molecular Sequence Data , Moxifloxacin
5.
PLoS One ; 7(7): e39847, 2012.
Article in English | MEDLINE | ID: mdl-22792191

ABSTRACT

SIRT6 is involved in inflammation, aging and metabolism potentially by modulating the functions of both NFκB and HIF1α. Since it is possible to make small molecule activators and inhibitors of Sirtuins we wished to establish biochemical and cellular assays both to assist in drug discovery efforts and to validate whether SIRT6 represents a valid drug target for these indications. We confirmed in cellular assays that SIRT6 can deacetylate acetylated-histone H3 lysine 9 (H3K9Ac), however this deacetylase activity is unusually low in biochemical assays. In an effort to develop alternative assay formats we observed that SIRT6 overexpression had no influence on TNFα induced nuclear translocation of NFκB, nor did it have an effect on nuclear mobility of RelA/p65. In an effort to identify a gene expression profile that could be used to identify a SIRT6 readout we conducted genome-wide expression studies. We observed that overexpression of SIRT6 had little influence on NFκB-dependent genes, but overexpression of the catalytically inactive mutant affected gene expression in developmental pathways.


Subject(s)
Gene Expression , Mutation , NF-kappa B/metabolism , Sirtuins/genetics , Sirtuins/metabolism , Acetylation , Active Transport, Cell Nucleus , Cell Line , Cell Nucleus/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Histones/metabolism , Humans , Kinetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology
6.
J Biomol Screen ; 17(5): 641-50, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22337655

ABSTRACT

Microsomal prostaglandin E synthase-1 (mPGES-1) represents an attractive target for the treatment of rheumatoid arthritis and pain, being upregulated in response to inflammatory stimuli. Biochemical assays for prostaglandin E synthase activity are complicated by the instability of the substrate (PGH(2)) and the challenge of detection of the product (PGE(2)). A coupled fluorescent assay is described for mPGES-1 where PGH(2) is generated in situ using the action of cyclooxygenase 2 (Cox-2) on arachidonic acid. PGE(2) is detected by coupling through 15-prostaglandin dehydrogenase (15-PGDH) and diaphorase. The overall coupled reaction was miniaturized to 1536-well plates and validated for high-throughput screening. For compound progression, a novel high-throughput mass spectrometry assay was developed using the RapidFire platform. The assay employs the same in situ substrate generation step as the fluorescent assay, after which both PGE(2) and a reduced form of the unreacted substrate were detected by mass spectrometry. Pharmacology and assay quality were comparable between both assays, but the mass spectrometry assay was shown to be less susceptible to interference and false positives. Exploiting the throughput of the fluorescent assay and the label-free, direct detection of the RapidFire has proved to be a powerful lead discovery strategy for this challenging target.


Subject(s)
Enzyme Inhibitors/pharmacology , Fluorescent Dyes/chemistry , High-Throughput Screening Assays/methods , Intramolecular Oxidoreductases/antagonists & inhibitors , Mass Spectrometry/methods , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Drug Discovery/methods , Fluorescent Dyes/metabolism , Humans , Inhibitory Concentration 50 , Intramolecular Oxidoreductases/metabolism , Prostaglandin-E Synthases
7.
Nature ; 466(7309): 935-40, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20686482

ABSTRACT

Despite the success of genomics in identifying new essential bacterial genes, there is a lack of sustainable leads in antibacterial drug discovery to address increasing multidrug resistance. Type IIA topoisomerases cleave and religate DNA to regulate DNA topology and are a major class of antibacterial and anticancer drug targets, yet there is no well developed structural basis for understanding drug action. Here we report the 2.1 A crystal structure of a potent, new class, broad-spectrum antibacterial agent in complex with Staphylococcus aureus DNA gyrase and DNA, showing a new mode of inhibition that circumvents fluoroquinolone resistance in this clinically important drug target. The inhibitor 'bridges' the DNA and a transient non-catalytic pocket on the two-fold axis at the GyrA dimer interface, and is close to the active sites and fluoroquinolone binding sites. In the inhibitor complex the active site seems poised to cleave the DNA, with a single metal ion observed between the TOPRIM (topoisomerase/primase) domain and the scissile phosphate. This work provides new insights into the mechanism of topoisomerase action and a platform for structure-based drug design of a new class of antibacterial agents against a clinically proven, but conformationally flexible, enzyme class.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , DNA Gyrase/chemistry , Quinolines/chemistry , Quinolines/pharmacology , Staphylococcus aureus/enzymology , Topoisomerase II Inhibitors , Anti-Bacterial Agents/metabolism , Apoenzymes/chemistry , Apoenzymes/metabolism , Arginine/metabolism , Aspartic Acid/metabolism , Binding Sites , Catalytic Domain , Ciprofloxacin/chemistry , Ciprofloxacin/metabolism , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , DNA Cleavage , DNA Gyrase/metabolism , DNA, Superhelical/chemistry , DNA, Superhelical/metabolism , Drug Design , Drug Resistance , Escherichia coli/enzymology , Manganese/metabolism , Models, Molecular , Protein Conformation , Quinolines/metabolism , Quinolones/chemistry , Quinolones/metabolism , Structure-Activity Relationship
8.
Nat Struct Mol Biol ; 17(9): 1152-3, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20802486

ABSTRACT

Quinolone antibacterials have been used to treat bacterial infections for over 40 years. A crystal structure of moxifloxacin in complex with Acinetobacter baumannii topoisomerase IV now shows the wedge-shaped quinolone stacking between base pairs at the DNA cleavage site and binding conserved residues in the DNA cleavage domain through chelation of a noncatalytic magnesium ion. This provides a molecular basis for the quinolone inhibition mechanism, resistance mutations and invariant quinolone antibacterial structural features.


Subject(s)
Acinetobacter baumannii/enzymology , DNA Topoisomerase IV/chemistry , Enzyme Inhibitors/chemistry , Quinolones/chemistry , DNA Topoisomerase IV/pharmacology , Enzyme Inhibitors/pharmacology , Models, Molecular , Protein Structure, Quaternary , Protein Structure, Tertiary , Quinolones/pharmacology
9.
Bioorg Med Chem Lett ; 15(9): 2305-9, 2005 May 02.
Article in English | MEDLINE | ID: mdl-15837314

ABSTRACT

High throughput screening of Staphylococcus aureus phenylalanyl tRNA synthetase (FRS) identified ethanolamine 1 as a sub-micromolar hit. Optimisation studies led to the enantiospecific lead 64, a single-figure nanomolar inhibitor. The inhibitor series shows selectivity with respect to the mammalian enzyme and the potential for broad spectrum bacterial FRS inhibition.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Ethanolamines/chemical synthesis , Ethanolamines/pharmacology , Phenylalanine-tRNA Ligase/antagonists & inhibitors , Staphylococcus aureus/enzymology , Animals , Anti-Bacterial Agents/pharmacology , Drug Design , Kinetics , Mammals , Microbial Sensitivity Tests , Models, Molecular , Sensitivity and Specificity , Staphylococcus aureus/drug effects , Structure-Activity Relationship
10.
Eur J Pharmacol ; 494(2-3): 91-9, 2004 Jun 28.
Article in English | MEDLINE | ID: mdl-15212962

ABSTRACT

The guinea pig 5-hydroxytryptamine(5A) (gp5-ht(5A)) receptor was cloned from guinea pig brain using degenerate polymerase chain reaction (PCR) and shows 88%, 85% and 84% amino acid sequence identity versus the human, rat and mouse 5-ht(5A) receptors, respectively. The receptor was transiently expressed in human embryonic kidney (HEK) 293 cells. [(3)H]-Lysergic acid diethylamide (LSD) bound saturably to gp5-ht(5A)/HEK293 membranes with a K(d) of 2.3+/-0.1 nM and B(max) of 15.7+/-3.4 pmol/mg protein. The receptor binding profile, determined by competition with [(3)H]LSD, correlated well with that for the human 5-ht(5A) receptor. 5-HT stimulated [(35)S]GTPgammaS binding to gp5-ht(5A)/HEK293 membranes (pEC(50) 8.1+/-0.2), and the response was surmountably antagonised by methiothepin and ritanserin, giving apparent pK(B) values of 8.0 and 7.2, respectively. The 5-HT response was absent using membranes prepared from gp5-ht(5A)/HEK293 cells pretreated with pertussis toxin (PTX). These data suggest that the gp5-ht(5A) receptor couples to G(i)-proteins in this expression system and shows a similar pharmacological profile to that for the human 5-ht(5A) receptor.


Subject(s)
Receptors, Serotonin/drug effects , Receptors, Serotonin/genetics , Serotonin/analogs & derivatives , Amino Acid Sequence , Animals , Brain Chemistry/genetics , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cloning, Molecular , Exons/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guinea Pigs , Humans , Lysergic Acid Diethylamide/metabolism , Methiothepin/pharmacology , Mice , Molecular Sequence Data , Radioligand Assay , Rats , Receptors, Serotonin/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/metabolism , Species Specificity
11.
J Biotechnol ; 109(1-2): 201-11, 2004 Apr 08.
Article in English | MEDLINE | ID: mdl-15063628

ABSTRACT

Building on the basic design concepts of Randers-Eichhorn [Biotechnol. Bioeng. 55 (1997) 921], an on-line, real-time robust, steam sterilisable optical sensor for monitoring green fluorescent protein (GFP) has been developed. A general cloning vector for fusion expression proteins was constructed, allowing expression of both GFP and the target protein as a fusion. Cultivations were carried out at the 20l scale with the signal from the sensor being relayed directly to the control system of the bioreactors. The production of GFP was then measured on-line, the signal was interfaced directly with other controlling parameters, thereby allowing the microbial process to be controlled directly based on recombinant protein expression. A positive expression correlation between on-line and off-line data was obtained. Protein accretion measured off-line was quantified using both LC-MS and plate reader assays. The potential of such a sensor for many aspects of process development is considerable and we have developed a working system which allows the optimisation of production conditions, for example, linking pH control directly to the fusion protein. Results are also presented that illustrate GFP does not alter the cultivation characteristics of the target protein when compared to the native construct. Whether GFP expressed as a fusion influences the solubility of the target protein is also discussed.


Subject(s)
Biosensing Techniques/instrumentation , Fermentation , Genetic Vectors/genetics , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/biosynthesis
12.
Mol Microbiol ; 49(5): 1191-200, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12940980

ABSTRACT

Two-component signal transduction (TCST) pathways are regulatory systems that are highly homologous throughout the bacterial kingdom. Their established role in virulence and absence in vertebrates has made TCST an attractive target for therapeutic intervention. However, such systems have yet to yield success in the development of novel antibiotics. CheY serves as a prototype for the analysis of response regulator function. The protein structure exhibits several conformations by both X-ray and nuclear magnetic resonance (NMR) analyses. Knowledge of which structures are relevant in vivo would be valuable in a rational drug design project. Our aim was to probe the in vivo conformation and ligand binding of CheY in Escherichia coli under resting conditions by in-cell NMR methods. CheY was selectively labelled with 15N by the control of growth and expression conditions. NMR spectra obtained in vivo demonstrated that the Mg2+ complex was the predominant form even though cells were resuspended in metal-free buffers and the intracellular free Mg2+ was low. In-cell NMR also confirmed the uptake and in vivo binding mode to CheY of small-molecular-weight compounds identified in vitro. This paper reports the first observation of the structure and interactions with a potential drug of a regulator protein in its native host in vivo using NMR spectroscopy.


Subject(s)
Bacterial Proteins , Escherichia coli/chemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Protein Conformation , Cations, Divalent/metabolism , Chemotaxis , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Ligands , Magnesium/metabolism , Methyl-Accepting Chemotaxis Proteins , Models, Molecular , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Signal Transduction
13.
Protein Sci ; 12(2): 349-60, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538898

ABSTRACT

Polypeptide deformylase (PDF) catalyzes the deformylation of polypeptide chains in bacteria. It is essential for bacterial cell viability and is a potential antibacterial drug target. Here, we report the crystal structures of polypeptide deformylase from four different species of bacteria: Streptococcus pneumoniae, Staphylococcus aureus, Haemophilus influenzae, and Escherichia coli. Comparison of these four structures reveals significant overall differences between the two Gram-negative species (E. coli and H. influenzae) and the two Gram-positive species (S. pneumoniae and S. aureus). Despite these differences and low overall sequence identity, the S1' pocket of PDF is well conserved among the four enzymes studied. We also describe the binding of nonpeptidic inhibitor molecules SB-485345, SB-543668, and SB-505684 to both S. pneumoniae and E. coli PDF. Comparison of these structures shows similar binding interactions with both Gram-negative and Gram-positive species. Understanding the similarities and subtle differences in active site structure between species will help to design broad-spectrum polypeptide deformylase inhibitor molecules.


Subject(s)
Amidohydrolases , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Enzyme Inhibitors/metabolism , Amino Acid Sequence , Aminopeptidases/metabolism , Bacterial Proteins/antagonists & inhibitors , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Escherichia coli/enzymology , Haemophilus influenzae/drug effects , Haemophilus influenzae/enzymology , Kinetics , Microbial Sensitivity Tests , Models, Molecular , Molecular Sequence Data , Peptides , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology
14.
Bioorg Med Chem Lett ; 12(21): 3171-4, 2002 Nov 04.
Article in English | MEDLINE | ID: mdl-12372526

ABSTRACT

The antimicrobial natural product chuangxinmycin has been found to be a potent and selective inhibitor of bacterial tryptophanyl tRNA synthetase (WRS). A number of analogues have been synthesised. The interaction with WRS appears to be highly constrained, as only sterically smaller analogues afforded significant inhibition. The only analogue to show inhibition comparable to chuangxinmycin also had antibacterial activity. WRS inhibition may contribute to the antibacterial action of chuangxinmycin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Staphylococcus aureus/enzymology , Tryptophan-tRNA Ligase/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Bacteria/drug effects , Enzyme Inhibitors/chemical synthesis , Hydrolysis , Indicators and Reagents , Indoles/chemical synthesis , Microbial Sensitivity Tests , Staphylococcus aureus/drug effects , Stereoisomerism , Structure-Activity Relationship
15.
Antimicrob Agents Chemother ; 46(10): 3118-24, 2002 10.
Article in English | MEDLINE | ID: mdl-12234833

ABSTRACT

Bacterial enoyl-acyl carrier protein (ACP) reductase (FabI) catalyzes the final step in each elongation cycle of bacterial fatty acid biosynthesis and is an attractive target for the development of new antibacterial agents. High-throughput screening of the Staphylococcus aureus FabI enzyme identified a novel, weak inhibitor with no detectable antibacterial activity against S. aureus. Iterative medicinal chemistry and X-ray crystal structure-based design led to the identification of compound 4 [(E)-N-methyl-N-(2-methyl-1H-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)acrylamide], which is 350-fold more potent than the original lead compound obtained by high-throughput screening in the FabI inhibition assay. Compound 4 has exquisite antistaphylococci activity, achieving MICs at which 90% of isolates are inhibited more than 500 times lower than those of nine currently available antibiotics against a panel of multidrug-resistant strains of S. aureus and Staphylococcus epidermidis. Furthermore, compound 4 exhibits excellent in vivo efficacy in an S. aureus infection model in rats. Biochemical and genetic approaches have confirmed that the mode of antibacterial action of compound 4 and related compounds is via inhibition of FabI. Compound 4 also exhibits weak FabK inhibitory activity, which may explain its antibacterial activity against Streptococcus pneumoniae and Enterococcus faecalis, which depend on FabK and both FabK and FabI, respectively, for their enoyl-ACP reductase function. These results show that compound 4 is representative of a new, totally synthetic series of antibacterial agents that has the potential to provide novel alternatives for the treatment of S. aureus infections that are resistant to our present armory of antibiotics.


Subject(s)
Anti-Bacterial Agents , Enzyme Inhibitors , Oxidoreductases/antagonists & inhibitors , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Multiple, Bacterial , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH) , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/enzymology , Humans , Male , Microbial Sensitivity Tests , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology , Structure-Activity Relationship
16.
J Med Chem ; 45(10): 1959-62, 2002 May 09.
Article in English | MEDLINE | ID: mdl-11985462

ABSTRACT

Potent nanomolar inhibitors of Staphylococcus aureus methionyl tRNA synthetase have been derived from a file compound high throughput screening hit. Optimized compounds show excellent antibacterial activity against staphylococcal and enterococcal pathogens, including strains resistant to clinical antibiotics. Compound 11 demonstrated in vivo efficacy in an S. aureus rat abscess infection model.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Enterococcus/drug effects , Enzyme Inhibitors/chemical synthesis , Methionine-tRNA Ligase/antagonists & inhibitors , Quinolones/chemical synthesis , Staphylococcus/drug effects , Abscess/drug therapy , Abscess/microbiology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Quinolones/chemistry , Quinolones/pharmacology , Rats , Rats, Sprague-Dawley , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...