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










Publication year range
2.
Nat Commun ; 11(1): 3387, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32636417

ABSTRACT

Biosynthesis of glycosylphosphatidylinositol (GPI) is required for anchoring proteins to the plasma membrane, and is essential for the integrity of the fungal cell wall. Here, we use a reporter gene-based screen in Saccharomyces cerevisiae for the discovery of antifungal inhibitors of GPI-anchoring of proteins, and identify the oligocyclopropyl-containing natural product jawsamycin (FR-900848) as a potent hit. The compound targets the catalytic subunit Spt14 (also referred to as Gpi3) of the fungal UDP-glycosyltransferase, the first step in GPI biosynthesis, with good selectivity over the human functional homolog PIG-A. Jawsamycin displays antifungal activity in vitro against several pathogenic fungi including Mucorales, and in vivo in a mouse model of invasive pulmonary mucormycosis due to Rhyzopus delemar infection. Our results provide a starting point for the development of Spt14 inhibitors for treatment of invasive fungal infections.


Subject(s)
Antifungal Agents/pharmacology , Glycosyltransferases/antagonists & inhibitors , Polyketides/pharmacology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Animals , Cell Proliferation , Disease Models, Animal , Fermentation , Genes, Reporter , Glycosylphosphatidylinositols/biosynthesis , HCT116 Cells , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , K562 Cells , Lung/microbiology , Male , Mice , Mice, Inbred ICR , Mucorales , Multigene Family , Rhizopus , Saccharomyces cerevisiae
3.
J Biol Chem ; 293(52): 19982-19995, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30327433

ABSTRACT

Actinobacteria possess a great wealth of pathways for production of bioactive compounds. Following advances in genome mining, dozens of natural product (NP) gene clusters are routinely found in each actinobacterial genome; however, the modus operandi of this large arsenal is poorly understood. During investigations of the secondary metabolome of Streptomyces rapamycinicus, the producer of rapamycin, we observed accumulation of two compounds never before reported from this organism. Structural elucidation revealed actinoplanic acid A and its demethyl analogue. Actinoplanic acids (APLs) are potent inhibitors of Ras farnesyltransferase and therefore represent bioactive compounds of medicinal interest. Supported with the unique structure of these polyketides and using genome mining, we identified a gene cluster responsible for their biosynthesis in S. rapamycinicus Based on experimental evidence and genetic organization of the cluster, we propose a stepwise biosynthesis of APL, the first bacterial example of a pathway incorporating the rare tricarballylic moiety into an NP. Although phylogenetically distant, the pathway shares some of the biosynthetic principles with the mycotoxins fumonisins. Namely, the core polyketide is acylated with the tricarballylate by an atypical nonribosomal peptide synthetase-catalyzed ester formation. Finally, motivated by the conserved colocalization of the rapamycin and APL pathway clusters in S. rapamycinicus and all other rapamycin-producing actinobacteria, we confirmed a strong synergism of these compounds in antifungal assays. Mining for such evolutionarily conserved coharboring of pathways would likely reveal further examples of NP sets, attacking multiple targets on the same foe. These could then serve as a guide for development of new combination therapies.


Subject(s)
Biosynthetic Pathways , Lactones/metabolism , Multigene Family , Polyketides/metabolism , Sirolimus/metabolism , Streptomyces/metabolism , Methylation , Secondary Metabolism , Streptomyces/genetics
4.
PLoS Genet ; 12(11): e1006374, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27855158

ABSTRACT

Invasive infections by fungal pathogens cause more deaths than malaria worldwide. We found the ergoline compound NGx04 in an antifungal screen, with selectivity over mammalian cells. High-resolution chemogenomics identified the lipid transfer protein Sec14p as the target of NGx04 and compound-resistant mutations in Sec14p define compound-target interactions in the substrate binding pocket of the protein. Beyond its essential lipid transfer function in a variety of pathogenic fungi, Sec14p is also involved in secretion of virulence determinants essential for the pathogenicity of fungi such as Cryptococcus neoformans, making Sec14p an attractive antifungal target. Consistent with this dual function, we demonstrate that NGx04 inhibits the growth of two clinical isolates of C. neoformans and that NGx04-related compounds have equal and even higher potency against C. neoformans. Furthermore NGx04 analogues showed fungicidal activity against a fluconazole resistant C. neoformans strain. In summary, we present genetic evidence that NGx04 inhibits fungal Sec14p and initial data supporting NGx04 as a novel antifungal starting point.


Subject(s)
Carrier Proteins/chemistry , Cryptococcosis/drug therapy , Cryptococcus neoformans/drug effects , Ergolines/pharmacology , Lipid Metabolism/drug effects , Antifungal Agents/pharmacology , Carrier Proteins/genetics , Cryptococcosis/microbiology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , Ergolines/chemistry , Humans , Microbial Sensitivity Tests , Protein Conformation , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics
5.
Methods Mol Biol ; 1439: 171-9, 2016.
Article in English | MEDLINE | ID: mdl-27316995

ABSTRACT

Due to the advancements in modern medicine that have resulted in an increased number of immunocompromised individuals, the incidences and the associated mortality of invasive aspergillosis have continued to rise over the past three decades despite appropriate treatment. As a result, invasive aspergillosis has emerged as a leading cause of infection-related mortality in immunocompromised individuals. Utilizing the resazurin to resorufin conversion fluorescence readout to monitor cell viability, herein, we outline a high-throughput screening method amenable to profiling a large pharmaceutical library against the clinically relevant but less frequently screened fungal pathogen Aspergillus fumigatus. This enables the user to conduct high-throughput screening using a disease-relevant fungal growth assay and identify novel antifungal chemotypes as drug leads.


Subject(s)
Antifungal Agents/pharmacology , Aspergillosis/drug therapy , Aspergillus fumigatus/drug effects , Growth Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Microbial Sensitivity Tests/methods , Aspergillosis/microbiology , Aspergillus fumigatus/growth & development , Fluorescent Dyes/analysis , Fluorescent Dyes/metabolism , Humans , Oxazines/analysis , Oxazines/metabolism , Xanthenes/analysis , Xanthenes/metabolism
6.
J Biomol Screen ; 21(3): 306-15, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26459507

ABSTRACT

Phenotypic screens are effective starting points to identify compounds with desirable activities. To find novel antifungals, we conducted a phenotypic screen in Saccharomyces cerevisiae and identified two discrete scaffolds with good growth inhibitory characteristics. Lack of broad-spectrum activity against pathogenic fungi called for directed chemical compound optimization requiring knowledge of the molecular target. Chemogenomic profiling identified effects on geranylgeranyltransferase I (GGTase I), an essential enzyme that prenylates proteins involved in cell signaling, such as Cdc42p and Rho1p. Selection of resistant mutants against both compounds confirmed the target hypothesis and enabled mapping of the compound binding site to the substrate binding pocket. Differential resistance-conferring mutations and selective substrate competition demonstrate distinct binding modes for the two chemotypes. Exchange of the S. cerevisiae GGTase I subunits with those of Candida albicans resulted in an absence of growth inhibition for both compounds, thus confirming the identified target as well as the narrow antifungal spectrum of activity. This prenylation pathway is reported to be nonessential in pathogenic species and challenges the therapeutic value of these leads while demonstrating the importance of an integrated target identification platform following a phenotypic screen.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antifungal Agents/pharmacology , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Microbial Sensitivity Tests , Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Antifungal Agents/chemistry , Drug Resistance, Fungal , Enzyme Inhibitors/chemistry , Gene Expression Profiling , Metabolomics/methods , Models, Molecular , Molecular Conformation , Mutation , Protein Binding , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics
7.
Nat Chem Biol ; 11(12): 958-66, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26479441

ABSTRACT

High-throughput screening (HTS) is an integral part of early drug discovery. Herein, we focused on those small molecules in a screening collection that have never shown biological activity despite having been exhaustively tested in HTS assays. These compounds are referred to as 'dark chemical matter' (DCM). We quantified DCM, validated it in quality control experiments, described its physicochemical properties and mapped it into chemical space. Through analysis of prospective reporter-gene assay, gene expression and yeast chemogenomics experiments, we evaluated the potential of DCM to show biological activity in future screens. We demonstrated that, despite the apparent lack of activity, occasionally these compounds can result in potent hits with unique activity and clean safety profiles, which makes them valuable starting points for lead optimization efforts. Among the identified DCM hits was a new antifungal chemotype with strong activity against the pathogen Cryptococcus neoformans but little activity at targets relevant to human safety.


Subject(s)
Antifungal Agents/pharmacology , Cryptococcus neoformans/drug effects , Drug Discovery , High-Throughput Screening Assays , Antifungal Agents/chemistry , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
8.
Nat Commun ; 6: 8613, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26456460

ABSTRACT

FR171456 is a natural product with cholesterol-lowering properties in animal models, but its molecular target is unknown, which hinders further drug development. Here we show that FR171456 specifically targets the sterol-4-alpha-carboxylate-3-dehydrogenase (Saccharomyces cerevisiae--Erg26p, Homo sapiens--NSDHL (NAD(P) dependent steroid dehydrogenase-like)), an essential enzyme in the ergosterol/cholesterol biosynthesis pathway. FR171456 significantly alters the levels of cholesterol pathway intermediates in human and yeast cells. Genome-wide yeast haploinsufficiency profiling experiments highlight the erg26/ERG26 strain, and multiple mutations in ERG26 confer resistance to FR171456 in growth and enzyme assays. Some of these ERG26 mutations likely alter Erg26 binding to FR171456, based on a model of Erg26. Finally, we show that FR171456 inhibits an artificial Hepatitis C viral replicon, and has broad antifungal activity, suggesting potential additional utility as an anti-infective. The discovery of the target and binding site of FR171456 within the target will aid further development of this compound.


Subject(s)
3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Antifungal Agents/chemistry , Cholesterol/analogs & derivatives , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/genetics , 3-Hydroxysteroid Dehydrogenases/genetics , Candida albicans , Cholesterol/chemistry , Drug Resistance, Fungal/genetics , Ergosterol/biosynthesis , Mutation , Saccharomyces cerevisiae Proteins/genetics
9.
J Mol Microbiol Biotechnol ; 23(6): 396-400, 2013.
Article in English | MEDLINE | ID: mdl-23942459

ABSTRACT

Biofilm growth represents one of the most challenging problems associated with Candida infections, largely due to the natural resistance of biofilm to the common antifungal drugs. As elevated expression of heat shock proteins (HSP) promotes Candida yeast-hyphae switch, which is an essential step in biofilm formation, we investigated the expression of hsp genes during Candida albicans biofilm development. By measuring mRNA levels using qRT-PCR, we found that all three hsp genes that we monitored are overexpressed in the initial stage of C. albicans biofilm formation. To corroborate this finding, we examined the effect of 17-DMAG, a specific Hsp90 inhibitor, on the formation of C. albicans biofilm. Our results indicate the requirement of HSP during the early phase of Candida biofilm development.


Subject(s)
Biofilms/growth & development , Candida albicans/physiology , Heat-Shock Proteins/metabolism , Candida albicans/genetics , Candida albicans/growth & development , Candida albicans/metabolism , Gene Expression Profiling , Heat-Shock Proteins/genetics , Real-Time Polymerase Chain Reaction
10.
Antimicrob Agents Chemother ; 57(5): 2272-80, 2013 May.
Article in English | MEDLINE | ID: mdl-23478965

ABSTRACT

High-throughput phenotypic screening against the yeast Saccharomyces cerevisiae revealed a series of triazolopyrimidine-sulfonamide compounds with broad-spectrum antifungal activity, no significant cytotoxicity, and low protein binding. To elucidate the target of this series, we have applied a chemogenomic profiling approach using the S. cerevisiae deletion collection. All compounds of the series yielded highly similar profiles that suggested acetolactate synthase (Ilv2p, which catalyzes the first common step in branched-chain amino acid biosynthesis) as a possible target. The high correlation with profiles of known Ilv2p inhibitors like chlorimuron-ethyl provided further evidence for a similar mechanism of action. Genome-wide mutagenesis in S. cerevisiae identified 13 resistant clones with 3 different mutations in the catalytic subunit of acetolactate synthase that also conferred cross-resistance to established Ilv2p inhibitors. Mapping of the mutations into the published Ilv2p crystal structure outlined the chlorimuron-ethyl binding cavity, and it was possible to dock the triazolopyrimidine-sulfonamide compound into this pocket in silico. However, fungal growth inhibition could be bypassed through supplementation with exogenous branched-chain amino acids or by the addition of serum to the medium in all of the fungal organisms tested except for Aspergillus fumigatus. Thus, these data support the identification of the triazolopyrimidine-sulfonamide compounds as inhibitors of acetolactate synthase but suggest that targeting may be compromised due to the possibility of nutrient bypass in vivo.


Subject(s)
Acetolactate Synthase/antagonists & inhibitors , Antifungal Agents/pharmacology , Pyrimidines/pharmacology , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/drug effects , Sulfonamides/pharmacology , Sulfonylurea Compounds/pharmacology , Acetolactate Synthase/chemistry , Acetolactate Synthase/genetics , Acetolactate Synthase/metabolism , Amino Acids, Branched-Chain/metabolism , Amino Acids, Branched-Chain/pharmacology , Antifungal Agents/chemistry , Catalytic Domain/drug effects , High-Throughput Screening Assays , Humans , Microbial Sensitivity Tests , Molecular Docking Simulation , Mutation , Protein Binding , Pyrimidines/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Serum/chemistry , Serum/metabolism , Sulfonamides/chemistry , Sulfonylurea Compounds/chemistry
11.
Antimicrob Agents Chemother ; 56(8): 4233-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22615293

ABSTRACT

Systemic life-threatening fungal infections represent a significant unmet medical need. Cell-based, phenotypic screening can be an effective means of discovering potential novel antifungal compounds, but it does not address target identification, normally required for compound optimization by medicinal chemistry. Here, we demonstrate a combination of screening, genetic, and biochemical approaches to identify and characterize novel antifungal compounds. We isolated a set of novel non-azole antifungal compounds for which no target or mechanism of action is known, using a screen for inhibition of Saccharomyces cerevisiae proliferation. Haploinsufficiency profiling of these compounds in S. cerevisiae suggests that they target Erg11p, a cytochrome P450 family member, which is the target of azoles. Consistent with this, metabolic profiling in S. cerevisiae revealed a buildup of the metabolic intermediates prior to Erg11p activity, following compound treatment. Further, human cytochrome P450 is also inhibited in in vitro assays by these compounds. We modeled the Erg11p protein based on the human CYP51 crystal structure, and in silico docking of these compounds suggests that they interact with the heme center in a manner similar to that of azoles. Consistent with these docking observations, Candida strains carrying azole-resistant alleles of ERG11 are also resistant to the compounds in this study. Thus, we have identified non-azole Erg11p inhibitors, using a systematic approach for ligand and target characterization.


Subject(s)
Antifungal Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/drug effects , Antifungal Agents/chemistry , Azoles/pharmacology , Cytochrome P-450 Enzyme System , Drug Resistance, Fungal/genetics , High-Throughput Screening Assays , Microbial Sensitivity Tests , Models, Molecular , Protein Structure, Quaternary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
12.
J Biomol Screen ; 16(6): 637-46, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21593486

ABSTRACT

A simple, optical density-based assay for inhibitors of the mevalonate-dependent pathway for isoprenoid biosynthesis was developed. The assay uses pathway-sensitized Staphylococcus aureus strains and is fully compatible with high-density screening in a 1536-well format. S. aureus strains were constructed in which genes required for mevalonate-dependent isopentenyl pyrophosphate (IPP) synthesis were regulated by an isopropyl-ß-D-thiogalactopyranoside (IPTG)-inducible promoter. Inhibitors of the target enzymes displayed greater antibacterial potency in media containing low concentrations of IPTG, and therefore less induction of mevalonate pathway genes, than in media with high IPTG conditions. This differential growth phenotype was exploited to bias the cell-based screening hits toward specific inhibitors of mevalonate-dependent IPP biosynthesis. Screens were run against strains engineered for regulation of the enzymes HMG-CoA synthase (MvaS) and mevalonate kinase (mvaK1), mevalonate diphosphate decarboxylase (mvaD), and phosphomevalonate kinase (mvaK2). The latter three enzymes are regulated as an operon. These assays resulted in the discovery of potent antibacterial hits that were progressed to an active hit-to-lead program. The example presented here demonstrates that a cell sensitization strategy can be successfully applied to a 1.3-million compound high-throughput screen in a high-density 1536-well format.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Mevalonic Acid/metabolism , Staphylococcus aureus/drug effects , Biosynthetic Pathways/drug effects , Biosynthetic Pathways/genetics , Hydroxymethylglutaryl-CoA Synthase/antagonists & inhibitors , Hydroxymethylglutaryl-CoA Synthase/genetics , Hydroxymethylglutaryl-CoA Synthase/metabolism , Mevalonic Acid/chemistry , Microbial Sensitivity Tests , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
13.
J Bacteriol ; 193(13): 3304-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21551303

ABSTRACT

Coenzyme A (CoA) plays a central and essential role in all living organisms. The pathway leading to CoA biosynthesis has been considered an attractive target for developing new antimicrobial agents with novel mechanisms of action. By using an arabinose-regulated expression system, the essentiality of coaBC, a single gene encoding a bifunctional protein catalyzing two consecutive steps in the CoA pathway converting 4'-phosphopantothenate to 4'-phosphopantetheine, was confirmed in Escherichia coli. Utilizing this regulated coaBC strain, it was further demonstrated that E. coli can effectively metabolize pantethine to bypass the requirement for coaBC. Interestingly, pantethine cannot be used by Pseudomonas aeruginosa to obviate coaBC. Through reciprocal complementation studies in combination with biochemical characterization, it was demonstrated that the differential characteristics of pantethine utilization in these two microorganisms are due to the different substrate specificities associated with endogenous pantothenate kinase, the first enzyme in the CoA biosynthetic pathway encoded by coaA in E. coli and coaX in P. aeruginosa.


Subject(s)
Carboxy-Lyases/deficiency , Escherichia coli/enzymology , Escherichia coli/metabolism , Pantetheine/analogs & derivatives , Peptide Synthases/deficiency , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/metabolism , Biosynthetic Pathways/genetics , Escherichia coli Proteins , Gene Deletion , Genes, Essential , Genetic Complementation Test , Multienzyme Complexes/deficiency , Pantetheine/metabolism
14.
Biochemistry ; 49(25): 5366-76, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20476728

ABSTRACT

Undecaprenyl pyrophosphate synthase (UPPS) catalyzes the consecutive condensation of eight molecules of isopentenyl pyrophosphate (IPP) with farnesyl pyrophosphate (FPP) to generate the C(55) undecaprenyl pyrophosphate (UPP). It has been demonstrated that tetramic acids (TAs) are selective and potent inhibitors of UPPS, but the mode of inhibition was unclear. In this work, we used a fluorescent FPP probe to study possible TA binding at the FPP binding site. A photosensitive TA analogue was designed and synthesized for the study of the site of interaction of TA with UPPS using photo-cross-linking and mass spectrometry. The interaction of substrates with UPPS and with the UPPS.TA complex was investigated by protein fluorescence spectroscopy. Our results suggested that tetramic acid binds to UPPS at an allosteric site adjacent to the FPP binding site. TA binds to free UPPS enzyme but not to substrate-bound UPPS. Unlike Escherichia coli UPPS which follows an ordered substrate binding mechanism, Streptococcus pneumoniae UPPS appears to follow a random-sequential substrate binding mechanism. Only one substrate, FPP or IPP, is able to bind to the UPPS.TA complex, but the quaternary complex, UPPS.TA.FPP.IPP, cannot be formed. We propose that binding of TA to UPPS significantly alters the conformation of UPPS needed for proper substrate binding. As the result, substrate turnover is prevented, leading to the inhibition of UPPS catalytic activity. These probe compounds and biophysical assays also allowed us to quickly study the mode of inhibition of other UPPS inhibitors identified from a high-throughput screening and inhibitors produced from a medicinal chemistry program.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pyrrolidinones/pharmacology , Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/isolation & purification , Alkyl and Aryl Transferases/metabolism , Allosteric Regulation , Amino Acid Sequence , Biophysics , Escherichia coli/enzymology , Fluorescent Dyes , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Pyrrolidinones/antagonists & inhibitors , Spectrometry, Mass, Electrospray Ionization , Streptococcus pneumoniae/enzymology
15.
Microbiology (Reading) ; 156(Pt 5): 1372-1383, 2010 May.
Article in English | MEDLINE | ID: mdl-20167623

ABSTRACT

Transcriptional profiling data accumulated in recent years for the clinically relevant pathogen Staphylococcus aureus have established a cell wall stress stimulon, which comprises a coordinately regulated set of genes that are upregulated in response to blockage of cell wall biogenesis. In particular, the expression of cwrA (SA2343, N315 notation), which encodes a putative 63 amino acid polypeptide of unknown biological function, increases over 100-fold in response to cell wall inhibition. Herein, we seek to understand the biological role that this gene plays in S. aureus. cwrA was found to be robustly induced by all cell wall-targeting antibiotics tested - vancomycin, oxacillin, penicillin G, phosphomycin, imipenem, hymeglusin and bacitracin - but not by antibiotics with other mechanisms of action, including ciprofloxacin, erythromycin, chloramphenicol, triclosan, rifampicin, novobiocin and carbonyl cyanide 3-chlorophenylhydrazone. Although a DeltacwrA S. aureus strain had no appreciable shift in MICs for cell wall-targeting antibiotics, the knockout was shown to have reduced cell wall integrity in a variety of other assays. Additionally, the gene was shown to be important for virulence in a mouse sepsis model of infection.


Subject(s)
Bacterial Proteins/physiology , Cell Wall/physiology , Staphylococcus aureus/genetics , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacteriolysis , Cell Wall/drug effects , Cell Wall/ultrastructure , Gene Expression Profiling , Gene Knockout Techniques , Genes, Reporter , Lysostaphin/pharmacology , Mice , Microbial Sensitivity Tests , Sepsis/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Staphylococcus aureus/ultrastructure , Virulence
16.
J Bacteriol ; 192(7): 1946-55, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20097861

ABSTRACT

Bacterial persister cells constitute a small portion of a culture which is tolerant to killing by lethal doses of bactericidal antibiotics. These phenotypic variants are formed in numerous bacterial species, including those with clinical relevance like the opportunistic pathogen Pseudomonas aeruginosa. Although persisters are believed to contribute to difficulties in the treatment of many infectious diseases, the underlying mechanisms affecting persister formation are not well understood. Here we show that even though P. aeruginosa cultures have a significantly smaller fraction of multidrug-tolerant persister cells than cultures of Escherichia coli or Staphylococcus aureus, they can increase persister numbers in response to quorum-sensing-related signaling molecules. The phenazine pyocyanin (and the closely related molecule paraquat) and the acyl-homoserine lactone 3-OC12-HSL significantly increased the persister numbers in logarithmic P. aeruginosa PAO1 or PA14 cultures but not in E. coli or S. aureus cultures.


Subject(s)
Drug Resistance, Multiple, Bacterial , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Quorum Sensing , Signal Transduction , Stress, Physiological , Acyl-Butyrolactones/metabolism , Anti-Bacterial Agents , Bacterial Proteins/genetics , Carbenicillin/pharmacology , Ciprofloxacin/pharmacology , Colony Count, Microbial , Gene Deletion , Genetic Complementation Test , Humans , Microbial Viability/drug effects , Paraquat/metabolism , Pyocyanine/metabolism
17.
Biochemistry ; 48(5): 1025-35, 2009 Feb 10.
Article in English | MEDLINE | ID: mdl-19138128

ABSTRACT

Siderophores are key virulence factors that allow bacteria to grow in iron-restricted environments. The Gram-positive pathogen Staphylococcus aureus is known to produce four siderophores for which genetic and/or structural data are unknown. Here we characterize the gene cluster responsible for producing the prevalent siderophore staphyloferrin A. In addition to expressing the cluster in the heterologous host Escherichia coli, which confers the ability to synthesize the siderophore, we reconstituted staphyloferrin A biosynthesis in vitro by expressing and purifying two key enzymes in the pathway. As with other polycarboxylate siderophores, staphyloferrin A is biosynthesized using the recently described nonribosomal peptide synthetase independent siderophore (NIS) biosynthetic pathway. Two NIS synthetases condense two molecules of citric acid to d-ornithine in a stepwise ordered process with SfnaD using the delta-amine as a nucleophile to form the first amide followed by SfnaB utilizing the alpha-amine to complete staphyloferrin A synthesis.


Subject(s)
Bacterial Proteins/genetics , Multigene Family , Ornithine/analogs & derivatives , Peptide Synthases/genetics , Siderophores/genetics , Staphylococcus aureus/genetics , Bacterial Proteins/biosynthesis , Bacterial Proteins/chemistry , Citrates/chemistry , Computational Biology/methods , Escherichia coli/enzymology , Escherichia coli/genetics , Genetic Code , Ornithine/chemistry , Ornithine/genetics , Peptide Synthases/chemistry , Siderophores/biosynthesis , Siderophores/chemistry , Staphylococcus aureus/chemistry
18.
J Bacteriol ; 191(3): 851-61, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19028897

ABSTRACT

Isoprenoids are a class of ubiquitous organic molecules synthesized from the five-carbon starter unit isopentenyl pyrophosphate (IPP). Comprising more than 30,000 known natural products, isoprenoids serve various important biological functions in many organisms. In bacteria, undecaprenyl pyrophosphate is absolutely required for the formation of cell wall peptidoglycan and other cell surface structures, while ubiquinones and menaquinones, both containing an essential prenyl moiety, are key electron carriers in respiratory energy generation. There is scant knowledge on the nature and regulation of bacterial isoprenoid pathways. In order to explore the cellular responses to perturbations in the mevalonate pathway, responsible for producing the isoprenoid precursor IPP in many gram-positive bacteria and eukaryotes, we constructed three strains of Staphylococcus aureus in which each of the mevalonate pathway genes is regulated by an IPTG (isopropyl-beta-D-thiogalactopyranoside)-inducible promoter. We used DNA microarrays to profile the transcriptional effects of downregulating the components of the mevalonate pathway in S. aureus and demonstrate that decreased expression of the mevalonate pathway leads to widespread downregulation of primary metabolism genes, an upregulation in virulence factors and cell wall biosynthetic determinants, and surprisingly little compensatory expression in other isoprenoid biosynthetic genes. We subsequently correlate these transcriptional changes with downstream metabolic consequences.


Subject(s)
Mevalonic Acid/metabolism , Signal Transduction/physiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Glycolysis/genetics , Hemiterpenes/metabolism , Microbial Viability/drug effects , Oligonucleotide Array Sequence Analysis , Organophosphorus Compounds/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Transcription, Genetic
19.
Bioorg Med Chem Lett ; 18(6): 1840-4, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18295483

ABSTRACT

Based on a pharmacophore hypothesis substituted tetramic and tetronic acid 3-carboxamides as well as dihydropyridin-2-one-3-carboxamides were investigated as inhibitors of undecaprenyl pyrophosphate synthase (UPPS) for use as novel antimicrobial agents. Synthesis and structure-activity relationship patterns for this class of compounds are discussed. Selectivity data and antibacterial activities for selected compounds are provided.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Amides/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Furans/pharmacology , Pyrrolidinones/pharmacology , Streptococcus pneumoniae/drug effects , Alkyl and Aryl Transferases/metabolism , Amides/chemical synthesis , Cyclization , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli/enzymology , Furans/chemical synthesis , Microbial Sensitivity Tests , Molecular Structure , Polyisoprenyl Phosphates/metabolism , Protein Conformation , Pyrrolidinones/chemical synthesis , Pyrrolidinones/chemistry , Sesquiterpenes/metabolism , Streptococcus pneumoniae/growth & development , Structure-Activity Relationship
20.
Expert Opin Ther Targets ; 6(4): 507-16, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12223065

ABSTRACT

There is an urgent need to develop novel classes of antibiotics to counter the inexorable rise of resistant bacterial pathogens. Modern antibacterial drug discovery is focused on the identification and validation of novel protein targets that may have a suitable therapeutic index. In combination with assays for function, the advent of microbial genomics has been invaluable in identifying novel antibacterial drug targets. The major challenge in this field is the implementation of methods that validate protein targets leading to the discovery of new chemical entities. Ligand-directed drug discovery has the distinct advantage of having a concurrent analysis of both the importance of a target in the disease process and its amenability to functional modulation by small molecules. VITA is a process that enables a target-based paradigm by using peptide ligands for direct in vitro and in vivo validation of antibacterial targets and the implementation of high-throughput assays to identify novel inhibitory molecules. This process can establish sufficient levels of confidence indicating that the target is relevant to the disease process and inhibition of the target will lead to effective disease treatment.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Design , Drug Evaluation, Preclinical/methods , Peptides/antagonists & inhibitors , Animals , Anti-Bacterial Agents/classification , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Drug Delivery Systems , Escherichia coli/drug effects , Escherichia coli/genetics , Genome, Bacterial , Genomics , Humans , Ligands , Mice , Peptide Library , Peptides/isolation & purification , Protein Binding , Structure-Activity Relationship , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...