Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters











Publication year range
1.
J Antibiot (Tokyo) ; 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090325

ABSTRACT

Boxazomycins A-C are potent broad-spectrum antibiotics isolated from Actinomycetes strain G495-1 in 1987. We now report that boxazomycin A inhibits bacterial growth by selectively inhibiting protein synthesis, its effect is bacteriostatic, and it is equally active against drug resistant bacterial strains. No cross-resistance to protein synthesis inhibitors was observed suggesting that its inhibition is distinct from clinical protein synthesis inhibitors. We also report in vivo efficacy in a Staphylococcus aureus murine infection model supported by corresponding pharmacokinetic studies.

2.
Bioorg Med Chem Lett ; 32: 127661, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33160023

ABSTRACT

We previously reported medicinal chemistry efforts that identified MK-5204, an orally efficacious ß-1,3-glucan synthesis inhibitor derived from the natural product enfumafungin. Further extensive optimization of the C2 triazole substituent identified 4-pyridyl as the preferred replacement for the carboxamide of MK-5204, leading to improvements in antifungal activity in the presence of serum, and increased oral exposure. Reoptimizing the aminoether at C3 in the presence of this newly discovered C2 substituent, confirmed that the (R) t-butyl, methyl aminoether of MK-5204 provided the best balance of these two key parameters, culminating in the discovery of ibrexafungerp, which is currently in phase III clinical trials. Ibrexafungerp displayed significantly improved oral efficacy in murine infection models, making it a superior candidate for clinical development as an oral treatment for Candida and Aspergillus infections.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus/drug effects , Candida albicans/drug effects , Glycosides/chemistry , Triterpenes/chemistry , beta-Glucans/metabolism , Administration, Oral , Animals , Antifungal Agents/chemical synthesis , Antifungal Agents/pharmacokinetics , Antifungal Agents/therapeutic use , Aspergillosis/drug therapy , Candidiasis/drug therapy , Disease Models, Animal , Glycosides/pharmacokinetics , Glycosides/pharmacology , Glycosides/therapeutic use , Half-Life , Mice , Structure-Activity Relationship , Triterpenes/pharmacokinetics , Triterpenes/pharmacology , Triterpenes/therapeutic use
3.
Bioorg Med Chem Lett ; 30(17): 127357, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32738971

ABSTRACT

Our previously reported efforts to produce an orally active ß-1,3-glucan synthesis inhibitor through the semi-synthetic modification of enfumafungin focused on replacing the C2 acetoxy moiety with an aminotetrazole and the C3 glycoside with a N,N-dimethylaminoether moiety. This work details further optimization of the C2 heterocyclic substituent, which identified 3-carboxamide-1,2,4-triazole as a replacement for the aminotetrazole with comparable antifungal activity. Alkylation of either the carboxamidetriazole at C2 or the aminoether at C3 failed to significantly improve oral efficacy. However, replacement of the isopropyl alpha amino substituent with a t-butyl, improved oral exposure while maintaining antifungal activity. These two structural modifications produced MK-5204, which demonstrated broad spectrum activity against Candida species and robust oral efficacy in a murine model of disseminated Candidiasis without the N-dealkylation liability observed for the previous lead.


Subject(s)
Antifungal Agents/chemistry , Triazoles/chemistry , beta-Glucans/metabolism , Administration, Oral , Animals , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Candida/drug effects , Candidiasis/drug therapy , Disease Models, Animal , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Glycosides/chemistry , Half-Life , Mice , Microbial Sensitivity Tests , Stereoisomerism , Structure-Activity Relationship , Triazoles/metabolism , Triazoles/pharmacology , Triazoles/therapeutic use , Triterpenes/chemistry , beta-Glucans/chemistry
4.
Article in English | MEDLINE | ID: mdl-30373794

ABSTRACT

Tedizolid phosphate, the prodrug of the active antibiotic tedizolid, is an oxazolidinone for the treatment of acute bacterial skin and skin structure infections. Studies in a mouse thigh infection model demonstrated that tedizolid has improved potency and pharmacokinetics/pharmacodynamics (PK/PD) compared with those of linezolid. Subsequent studies showed that the efficacy of tedizolid was enhanced in immunocompetent (IC) mice compared with neutropenic (immunosuppressed [IS]) mice, with stasis at clinically relevant doses being achieved only in the presence of granulocytes. The tedizolid label therefore contains a warning about its use in neutropenic patients. This study reevaluated the PK/PD of tedizolid and linezolid in the mouse thigh infection model in IC and IS mice using a methicillin-resistant Staphylococcus aureus (MRSA) strain (ATCC 33591) and a methicillin-susceptible S. aureus (MSSA) strain (ATCC 29213). The antistaphylococcal effect of doses ranging from 1 to 150 mg/kg of body weight tedizolid (once daily) or linezolid (twice daily) was determined at 24, 48, and 72 h after initiating treatment. In IC mice, stasis was achieved in the absence of antibiotics, and both tedizolid and linezolid reduced the burden further beyond a static effect. In IS mice, tedizolid achieved stasis against MRSA ATCC 33591 and MSSA ATCC 29213 at 72 h at a human clinical dose of 200 mg, severalfold lower than that in earlier studies. Linezolid achieved a static effect against MRSA ATCC 33591 in IS mice at a dose lower than that used clinically. This study demonstrates that, with time, both tedizolid and linezolid at clinically relevant exposures achieve stasis in neutropenic mice with an MRSA or MSSA thigh infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Linezolid/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Neutropenia/metabolism , Organophosphates/pharmacology , Oxazoles/pharmacology , Staphylococcal Skin Infections/drug therapy , Animals , Anti-Bacterial Agents/pharmacokinetics , Disease Models, Animal , Linezolid/pharmacokinetics , Mice , Microbial Sensitivity Tests , Organophosphates/pharmacokinetics , Oxazoles/pharmacokinetics , Staphylococcal Skin Infections/microbiology
5.
Cell Chem Biol ; 24(5): 576-588.e6, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28434876

ABSTRACT

Riboswitches are bacterial-specific, broadly conserved, non-coding RNA structural elements that control gene expression of numerous metabolic pathways and transport functions essential for cell growth. As such, riboswitch inhibitors represent a new class of potential antibacterial agents. Recently, we identified ribocil-C, a highly selective inhibitor of the flavin mononucleotide (FMN) riboswitch that controls expression of de novo riboflavin (RF, vitamin B2) biosynthesis in Escherichia coli. Here, we provide a mechanistic characterization of the antibacterial effects of ribocil-C as well as of roseoflavin (RoF), an antimetabolite analog of RF, among medically significant Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA) and Enterococcus faecalis. We provide genetic, biophysical, computational, biochemical, and pharmacological evidence that ribocil-C and RoF specifically inhibit dual FMN riboswitches, separately controlling RF biosynthesis and uptake processes essential for MRSA growth and pathogenesis. Such a dual-targeting mechanism is specifically required to develop broad-spectrum Gram-positive antibacterial agents targeting RF metabolism.


Subject(s)
Flavin Mononucleotide/genetics , Homeostasis/drug effects , Pyrimidines/pharmacology , Riboflavin/analogs & derivatives , Riboflavin/metabolism , Riboswitch/drug effects , Staphylococcus aureus/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Base Sequence , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism , Methicillin-Resistant Staphylococcus aureus/physiology , Mice , Models, Molecular , Molecular Targeted Therapy , Protein Conformation , Riboflavin/pharmacology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Staphylococcus aureus/physiology
6.
J Med Chem ; 60(9): 3851-3865, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28322556

ABSTRACT

We describe our optimization efforts to improve the physicochemical properties, solubility, and off-target profile of 1, an inhibitor of TarO, an early stage enzyme in the biosynthetic pathway for wall teichoic acid (WTA) synthesis. Compound 1 displayed a TarO IC50 of 125 nM in an enzyme assay and possessed very high lipophilicity (clogP = 7.1) with no measurable solubility in PBS buffer. Structure-activity relationship (SAR) studies resulted in a series of compounds with improved lipophilic ligand efficiency (LLE) consistent with the reduction of clogP. From these efforts, analog 9 was selected for our initial in vivo study, which in combination with subefficacious dose of imipenem (IPM) robustly lowered the bacterial burden in a neutropenic Staphylococci murine infection model. Concurrent with our in vivo optimization effort using 9, we further improved LLE as exemplified by a much more druglike analog 26.


Subject(s)
Lipids/chemistry , Small Molecule Libraries , Animals , Female , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Solubility , Structure-Activity Relationship
7.
Anal Biochem ; 518: 9-15, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27815077

ABSTRACT

The emergence of methicillin-resistant Staphylococcus aureus (MRSA) has created an urgent need for new therapeutic agents capable of combating this threat. We have previously reported on the discovery of novel inhibitors targeting enzymes involved in the biosynthesis of wall teichoic acid (WTA) and demonstrated that these agents can restore ß-lactam efficacy against MRSA. In those previous reports pathway engagement of inhibitors was demonstrated by reduction in WTA levels measured by polyacrylamide gel electrophoresis. To enable a more rigorous analysis of these inhibitors we sought to develop a quantitative method for measuring whole-cell reductions in WTA. Herein we describe a robust methodology for hydrolyzing polymeric WTA to the monomeric component ribitol-N-acetylglucosamine coupled with measurement by LC-MS/MS. Critical elements of the protocol were found to include the time and temperature of hydrofluoric acid-mediated hydrolysis of polymeric WTA and optimization of these parameters is fully described. Most significantly, the assay enabled accurate and reproducible measurement of depletion EC50s for tunicamycin and representatives from the novel class of TarO inhibitors, the tarocins. The method described can readily be adapted to quantifying levels of WTA in tissue homogenates from a murine model of infection, highlighting the applicability for both in vitro and in vivo characterizations.


Subject(s)
Mass Spectrometry/methods , Methicillin-Resistant Staphylococcus aureus/metabolism , Teichoic Acids/metabolism , Chromatography, Liquid/methods , Methicillin-Resistant Staphylococcus aureus/chemistry , Teichoic Acids/chemistry , Tunicamycin/pharmacology
8.
J Pharmacol Exp Ther ; 359(1): 194-206, 2016 10.
Article in English | MEDLINE | ID: mdl-27432892

ABSTRACT

The renal outer medullary potassium (ROMK) channel, located at the apical surface of epithelial cells in the thick ascending loop of Henle and cortical collecting duct, contributes to salt reabsorption and potassium secretion, and represents a target for the development of new mechanism of action diuretics. This idea is supported by the phenotype of antenatal Bartter's syndrome type II associated with loss-of-function mutations in the human ROMK channel, as well as, by cardiovascular studies of heterozygous carriers of channel mutations associated with type II Bartter's syndrome. Although the pharmacology of ROMK channels is still being developed, channel inhibitors have been identified and shown to cause natriuresis and diuresis, in the absence of any significant kaliuresis, on acute oral dosing to rats or dogs. Improvements in potency and selectivity have led to the discovery of MK-7145 [5,5'-((1R,1'R)-piperazine-1,4-diylbis(1-hydroxyethane-2,1-diyl))bis(4-methylisobenzofuran-1(3H)-one)], a potential clinical development candidate. In spontaneously hypertensive rats, oral dosing of MK-7145 causes dose-dependent lowering of blood pressure that is maintained during the entire treatment period, and that displays additive/synergistic effects when administered in combination with hydrochlorothiazide or candesartan, respectively. Acute or chronic oral administration of MK-7145 to normotensive dogs led to dose-dependent diuresis and natriuresis, without any significant urinary potassium losses or changes in plasma electrolyte levels. Elevations in bicarbonate and aldosterone were found after 6 days of dosing. These data indicate that pharmacological inhibition of ROMK has potential as a new mechanism for the treatment of hypertension and/or congestive heart failure. In addition, Bartter's syndrome type II features are manifested on exposure to ROMK inhibitors.


Subject(s)
Bartter Syndrome/physiopathology , Benzofurans/pharmacology , Blood Pressure/drug effects , Phenotype , Piperazines/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Animals , Bartter Syndrome/drug therapy , Benzimidazoles/pharmacology , Benzofurans/therapeutic use , Biphenyl Compounds , Dogs , Dose-Response Relationship, Drug , Drug Synergism , Female , HEK293 Cells , Humans , Hydrochlorothiazide/pharmacology , Male , Piperazines/therapeutic use , Potassium Channel Blockers/therapeutic use , Rats , Tetrazoles/pharmacology
9.
Antimicrob Agents Chemother ; 60(8): 4830-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27246784

ABSTRACT

Oxabicyclooctane-linked novel bacterial topoisomerase inhibitors (NBTIs) represent a new class of recently described antibacterial agents with broad-spectrum activity. NBTIs dually inhibit the clinically validated bacterial targets DNA gyrase and topoisomerase IV and have been shown to bind distinctly from known classes of antibacterial agents directed against these targets. Herein we report the molecular, cellular, and in vivo characterization of AM-8722 as a representative N-alkylated-1,5-naphthyridone left-hand-side-substituted NBTI. Consistent with its mode of action, macromolecular labeling studies revealed a specific effect of AM-8722 to dose dependently inhibit bacterial DNA synthesis. AM-8722 displayed greater intrinsic enzymatic potency than levofloxacin versus both DNA gyrase and topoisomerase IV from Staphylococcus aureus and Escherichia coli and displayed selectivity against human topoisomerase II. AM-8722 was rapidly bactericidal and exhibited whole-cell activity versus a range of Gram-negative and Gram-positive organisms, with no whole-cell potency shift due to the presence of DNA or human serum. Frequency-of-resistance studies demonstrated an acceptable rate of resistance emergence in vitro at concentrations 16- to 32-fold the MIC. AM-8722 displayed acceptable pharmacokinetic properties and was shown to be efficacious in mouse models of bacterial septicemia. Overall, AM-8722 is a selective and potent NBTI that displays broad-spectrum antimicrobial activity in vitro and in vivo.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cyclooctanes/pharmacology , DNA Gyrase/metabolism , DNA Topoisomerase IV/antagonists & inhibitors , DNA Topoisomerases, Type II/metabolism , Topoisomerase II Inhibitors/pharmacology , Animals , Cell Line , DNA, Bacterial/genetics , Dogs , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Infections/drug therapy , Humans , Mice , Microbial Sensitivity Tests , Rats , Rats, Wistar , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics
10.
PLoS Pathog ; 12(5): e1005585, 2016 05.
Article in English | MEDLINE | ID: mdl-27144276

ABSTRACT

Here we describe a chemical biology strategy performed in Staphylococcus aureus and Staphylococcus epidermidis to identify MnaA, a 2-epimerase that we demonstrate interconverts UDP-GlcNAc and UDP-ManNAc to modulate substrate levels of TarO and TarA wall teichoic acid (WTA) biosynthesis enzymes. Genetic inactivation of mnaA results in complete loss of WTA and dramatic in vitro ß-lactam hypersensitivity in methicillin-resistant S. aureus (MRSA) and S. epidermidis (MRSE). Likewise, the ß-lactam antibiotic imipenem exhibits restored bactericidal activity against mnaA mutants in vitro and concomitant efficacy against 2-epimerase defective strains in a mouse thigh model of MRSA and MRSE infection. Interestingly, whereas MnaA serves as the sole 2-epimerase required for WTA biosynthesis in S. epidermidis, MnaA and Cap5P provide compensatory WTA functional roles in S. aureus. We also demonstrate that MnaA and other enzymes of WTA biosynthesis are required for biofilm formation in MRSA and MRSE. We further determine the 1.9Å crystal structure of S. aureus MnaA and identify critical residues for enzymatic dimerization, stability, and substrate binding. Finally, the natural product antibiotic tunicamycin is shown to physically bind MnaA and Cap5P and inhibit 2-epimerase activity, demonstrating that it inhibits a previously unanticipated step in WTA biosynthesis. In summary, MnaA serves as a new Staphylococcal antibiotic target with cognate inhibitors predicted to possess dual therapeutic benefit: as combination agents to restore ß-lactam efficacy against MRSA and MRSE and as non-bioactive prophylactic agents to prevent Staphylococcal biofilm formation.


Subject(s)
Bacterial Proteins/metabolism , Racemases and Epimerases/metabolism , Staphylococcus aureus/metabolism , Staphylococcus epidermidis/metabolism , Teichoic Acids/biosynthesis , Animals , Bacterial Proteins/chemistry , Biofilms/growth & development , Cell Wall/metabolism , Crystallography, X-Ray , Disease Models, Animal , Methicillin-Resistant Staphylococcus aureus , Mice , Microbial Sensitivity Tests , Microscopy, Fluorescence , Nuclear Magnetic Resonance, Biomolecular , Racemases and Epimerases/chemistry , Staphylococcal Infections/metabolism
11.
Sci Transl Med ; 8(329): 329ra32, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26962156

ABSTRACT

The widespread emergence of methicillin-resistant Staphylococcus aureus (MRSA) has dramatically eroded the efficacy of current ß-lactam antibiotics and created an urgent need for new treatment options. We report an S. aureus phenotypic screening strategy involving chemical suppression of the growth inhibitory consequences of depleting late-stage wall teichoic acid biosynthesis. This enabled us to identify early-stage pathway-specific inhibitors of wall teichoic acid biosynthesis predicted to be chemically synergistic with ß-lactams. We demonstrated by genetic and biochemical means that each of the new chemical series discovered, herein named tarocin A and tarocin B, inhibited the first step in wall teichoic acid biosynthesis (TarO). Tarocins do not have intrinsic bioactivity but rather demonstrated potent bactericidal synergy in combination with broad-spectrum ß-lactam antibiotics against diverse clinical isolates of methicillin-resistant staphylococci as well as robust efficacy in a murine infection model of MRSA. Tarocins and other inhibitors of wall teichoic acid biosynthesis may provide a rational strategy to develop Gram-positive bactericidal ß-lactam combination agents active against methicillin-resistant staphylococci.


Subject(s)
Bacterial Proteins/metabolism , Biosynthetic Pathways/drug effects , Cell Wall/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Teichoic Acids/biosynthesis , beta-Lactams/pharmacology , Animals , Cell Wall/drug effects , Dicloxacillin/pharmacology , Dicloxacillin/therapeutic use , Female , Mice, Inbred BALB C , Microbial Sensitivity Tests , Models, Molecular , Phenotype , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Treatment Outcome
12.
Bioorg Med Chem Lett ; 25(24): 5813-8, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26542966

ABSTRACT

The clinical success of the echinocandins, which can only be administered parentally, has validated ß-1,3-glucan synthase (GS) as an antifungal target. Semi-synthetic modification of enfumafungin, a triterpene glycoside natural product, was performed with the aim of producing a new class of orally active GS inhibitors. Replacement of the C2 acetoxy moiety with various heterocycles did not improve GS or antifungal potency. However, replacement of the C3 glycoside with an aminoether moiety dramatically improved oral pharmacokinetic (PK) properties while maintaining GS and antifungal potency. Installing an aminotetrazole at C2 in conjunction with an N-alkylated aminoether at C3 produced derivatives with significantly improved GS and antifungal potency that exhibited robust oral efficacy in a murine model of disseminated candidiasis.


Subject(s)
Antifungal Agents/chemistry , Glycosides/chemistry , Triterpenes/chemistry , beta-Glucans/chemistry , Administration, Oral , Animals , Antifungal Agents/pharmacokinetics , Antifungal Agents/therapeutic use , Aspergillus fumigatus/drug effects , Candida albicans/drug effects , Candidiasis/drug therapy , Candidiasis/veterinary , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Half-Life , Mice , Microbial Sensitivity Tests , Structure-Activity Relationship , Terpenes/chemistry , beta-Glucans/pharmacokinetics , beta-Glucans/therapeutic use
13.
Chem Biol ; 22(10): 1362-73, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26456734

ABSTRACT

Resistance to existing classes of antibiotics drives the need for discovery of novel compounds with unique mechanisms of action. Nargenicin A1, a natural product with limited antibacterial spectrum, was rediscovered in a whole-cell antisense assay. Macromolecular labeling in both Staphylococcus aureus and an Escherichia coli tolC efflux mutant revealed selective inhibition of DNA replication not due to gyrase or topoisomerase IV inhibition. S. aureus nargenicin-resistant mutants were selected at a frequency of ∼1 × 10(-9), and whole-genome resequencing found a single base-pair change in the dnaE gene, a homolog of the E. coli holoenzyme α subunit. A DnaE single-enzyme assay was exquisitely sensitive to inhibition by nargenicin, and other in vitro characterization studies corroborated DnaE as the target. Medicinal chemistry efforts may expand the spectrum of this novel mechanism antibiotic.


Subject(s)
DNA Polymerase III/genetics , Drug Discovery , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , DNA Replication/drug effects , DNA-Directed DNA Polymerase/metabolism , Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , Inhibitory Concentration 50 , Lactones/chemistry , Lactones/metabolism , Lactones/pharmacology , Mutation , Nucleic Acid Synthesis Inhibitors/chemistry , Nucleic Acid Synthesis Inhibitors/pharmacology , Staphylococcus aureus/drug effects
14.
Nature ; 526(7575): 672-7, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26416753

ABSTRACT

Riboswitches are non-coding RNA structures located in messenger RNAs that bind endogenous ligands, such as a specific metabolite or ion, to regulate gene expression. As such, riboswitches serve as a novel, yet largely unexploited, class of emerging drug targets. Demonstrating this potential, however, has proven difficult and is restricted to structurally similar antimetabolites and semi-synthetic analogues of their cognate ligand, thus greatly restricting the chemical space and selectivity sought for such inhibitors. Here we report the discovery and characterization of ribocil, a highly selective chemical modulator of bacterial riboflavin riboswitches, which was identified in a phenotypic screen and acts as a structurally distinct synthetic mimic of the natural ligand, flavin mononucleotide, to repress riboswitch-mediated ribB gene expression and inhibit bacterial cell growth. Our findings indicate that non-coding RNA structural elements may be more broadly targeted by synthetic small molecules than previously expected.


Subject(s)
Pyrimidines/chemistry , Pyrimidines/pharmacology , RNA, Bacterial/chemistry , RNA, Bacterial/drug effects , Riboswitch/drug effects , Animals , Aptamers, Nucleotide/chemistry , Bacteria/cytology , Bacteria/drug effects , Bacteria/growth & development , Base Sequence , Crystallography, X-Ray , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Female , Flavin Mononucleotide/metabolism , Gene Expression Regulation, Bacterial/drug effects , Heat-Shock Proteins/genetics , Intramolecular Transferases/genetics , Ligands , Mice , Mice, Inbred DBA , Models, Molecular , Molecular Sequence Data , Pyrimidines/isolation & purification , Pyrimidines/therapeutic use , RNA, Bacterial/genetics , Reproducibility of Results , Riboflavin/biosynthesis , Riboswitch/genetics , Substrate Specificity
15.
Bioorg Med Chem Lett ; 25(12): 2473-8, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25978963

ABSTRACT

Novel bacterial topoisomerase inhibitors (NBTIs) are a new class of broad-spectrum antibacterial agents targeting bacterial Gyrase A and ParC and have potential utility in combating antibiotic resistance. (R)-Hydroxy-1,5-naphthyridinone left-hand side (LHS) oxabicyclooctane linked pyridoxazinone right-hand side (RHS) containing NBTIs showed a potent Gram-positive antibacterial profile. SAR around the RHS moiety, including substitutions around pyridooxazinone, pyridodioxane, and phenyl propenoids has been described. A fluoro substituted pyridoxazinone showed an MIC against Staphylococcus aureus of 0.5 µg/mL with reduced functional hERG activity (IC50 333 µM) and good in vivo efficacy [ED90 12 mg/kg, intravenous (iv) and 15 mg/kg, oral (p.o.)]. A pyridodioxane-containing NBTI showed a S. aureus MIC of 0.5 µg/mL, significantly improved hERG IC50 764 µM and strong efficacy of 11 mg/kg (iv) and 5 mg/kg (p.o.). A phenyl propenoid series of compounds showed potent antibacterial activity, but also showed potent hERG binding activity. Many of the compounds in the hydroxy-tricyclic series showed strong activity against Acinetobacter baumannii, but reduced activity against Escherichia coli and Pseudomonas aeruginosa. Bicyclic heterocycles appeared to be the best RHS moiety for the hydroxy-tricyclic oxabicyclooctane linked NBTIs.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Naphthyridines/chemistry , Topoisomerase Inhibitors/chemistry , Topoisomerase Inhibitors/pharmacology , Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/chemical synthesis , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , DNA Gyrase/chemistry , DNA Gyrase/metabolism , Escherichia coli/drug effects , Microbial Sensitivity Tests , Oxazoles/chemistry , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects , Structure-Activity Relationship , Topoisomerase Inhibitors/chemical synthesis
16.
J Antibiot (Tokyo) ; 67(7): 527-31, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24690911

ABSTRACT

The ever-increasing bacterial resistance to clinical antibiotics is making many drugs ineffective and creating significant treatment gaps. This can be only circumvented by the discovery of antibiotics with new mechanisms of action. We report here the identification of a new tetramic acid, ascosetin, from an Ascomycete using the Staphylococcus aureus fitness test screening method. The structure was elucidated by spectroscopic methods including 2D NMR and HRMS. Relative stereochemistry was determined by ROESY and absolute configuration was deduced by comparative CD spectroscopy. Ascosetin inhibited bacterial growth with 2-16 µg ml(-1) MIC values against Gram-positive strains including methicillin-resistant S. aureus. It also inhibited the growth of Haemophilus influenzae with a MIC value of 8 µg ml(-1). It inhibited DNA, RNA, protein and lipid synthesis with similar IC50 values, suggesting a lack of specificity; however, it produced neither bacterial membrane nor red blood cell lysis. It showed selectivity for bacterial growth inhibition compared with fungal but not mammalian cells. The isolation, structure and biological activity of ascosetin have been detailed here.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacology , Anti-Bacterial Agents/isolation & purification , Ascomycota/drug effects , Haemophilus influenzae/drug effects , Magnetic Resonance Spectroscopy , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Molecular Conformation , Pyrrolidinones/isolation & purification , Staphylococcus aureus/drug effects
17.
ACS Chem Biol ; 8(11): 2442-51, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23957438

ABSTRACT

Modern medicine is founded on the discovery of penicillin and subsequent small molecules that inhibit bacterial peptidoglycan (PG) and cell wall synthesis. However, the discovery of new chemically and mechanistically distinct classes of PG inhibitors has become exceedingly rare, prompting speculation that intracellular enzymes involved in PG precursor synthesis are not 'druggable' targets. Here, we describe a ß-lactam potentiation screen to identify small molecules that augment the activity of ß-lactams against methicillin-resistant Staphylococcus aureus (MRSA) and mechanistically characterize a compound resulting from this screen, which we have named murgocil. We provide extensive genetic, biochemical, and structural modeling data demonstrating both in vitro and in whole cells that murgocil specifically inhibits the intracellular membrane-associated glycosyltransferase, MurG, which synthesizes the lipid II PG substrate that penicillin binding proteins (PBPs) polymerize and cross-link into the cell wall. Further, we demonstrate that the chemical synergy and cidality achieved between murgocil and the ß-lactam imipenem is mediated through MurG dependent localization of PBP2 to the division septum. Collectively, these data validate our approach to rationally identify new target-specific bioactive ß-lactam potentiation agents and demonstrate that murgocil now serves as a highly selective and potent chemical probe to assist our understanding of PG biosynthesis and cell wall biogenesis across Staphylococcal species.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/antagonists & inhibitors , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Peptidoglycan Glycosyltransferase/metabolism , Pyrazoles/pharmacology , Staphylococcus aureus/drug effects , Sterols/pharmacology , Computer Simulation , Drug Resistance, Bacterial , Enzyme Inhibitors/pharmacology , Humans , Microscopy, Fluorescence , Models, Molecular , Pyrazoles/chemistry , Staphylococcus aureus/enzymology , Sterols/chemistry
18.
Chem Biol ; 20(2): 272-84, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23438756

ABSTRACT

Innovative strategies are needed to combat drug resistance associated with methicillin-resistant Staphylococcus aureus (MRSA). Here, we investigate the potential of wall teichoic acid (WTA) biosynthesis inhibitors as combination agents to restore ß-lactam efficacy against MRSA. Performing a whole-cell pathway-based screen, we identified a series of WTA inhibitors (WTAIs) targeting the WTA transporter protein, TarG. Whole-genome sequencing of WTAI-resistant isolates across two methicillin-resistant Staphylococci spp. revealed TarG as their common target, as well as a broad assortment of drug-resistant bypass mutants mapping to earlier steps of WTA biosynthesis. Extensive in vitro microbiological analysis and animal infection studies provide strong genetic and pharmacological evidence of the potential effectiveness of WTAIs as anti-MRSA ß-lactam combination agents. This work also highlights the emerging role of whole-genome sequencing in antibiotic mode-of-action and resistance studies.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Wall/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Teichoic Acids/biosynthesis , beta-Lactams/metabolism , Amino Acid Substitution , Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Evaluation, Preclinical , Drug Resistance, Bacterial , Genome, Bacterial , Methicillin-Resistant Staphylococcus aureus/genetics , Microbial Sensitivity Tests , Osmolar Concentration , Phenotype , Sequence Analysis, DNA , Teichoic Acids/chemistry , Temperature , beta-Lactams/chemistry
19.
Sci Transl Med ; 4(126): 126ra35, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22440737

ABSTRACT

Despite the need for new antibiotics to treat drug-resistant bacteria, current clinical combinations are largely restricted to ß-lactam antibiotics paired with ß-lactamase inhibitors. We have adapted a Staphylococcus aureus antisense knockdown strategy to genetically identify the cell division Z ring components-FtsA, FtsZ, and FtsW-as ß-lactam susceptibility determinants of methicillin-resistant S. aureus (MRSA). We demonstrate that the FtsZ-specific inhibitor PC190723 acts synergistically with ß-lactam antibiotics in vitro and in vivo and that this combination is efficacious in a murine model of MRSA infection. Fluorescence microscopy localization studies reveal that synergy between these agents is likely to be elicited by the concomitant delocalization of their cognate drug targets (FtsZ and PBP2) in MRSA treated with PC190723. A 2.0 Å crystal structure of S. aureus FtsZ in complex with PC190723 identifies the compound binding site, which corresponds to the predominant location of mutations conferring resistance to PC190723 (PC190723(R)). Although structural studies suggested that these drug resistance mutations may be difficult to combat through chemical modification of PC190723, combining PC190723 with the ß-lactam antibiotic imipenem markedly reduced the spontaneous frequency of PC190723(R) mutants. Multiple MRSA PC190723(R) FtsZ mutants also displayed attenuated virulence and restored susceptibility to ß-lactam antibiotics in vitro and in a mouse model of imipenem efficacy. Collectively, these data support a target-based approach to rationally develop synergistic combination agents that mitigate drug resistance and effectively treat MRSA infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , beta-Lactams/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Division/drug effects , Crystallography, X-Ray , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Drug Resistance, Bacterial/drug effects , Drug Synergism , Gene Regulatory Networks/genetics , Guanosine Diphosphate , Imipenem/pharmacology , Methicillin-Resistant Staphylococcus aureus/cytology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Microbial Sensitivity Tests , Mutation/genetics , Protein Structure, Secondary , Protein Transport/drug effects , Pyridines/chemistry , Pyridines/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Thiazoles/chemistry , Thiazoles/pharmacology , Virulence/drug effects , beta-Lactams/therapeutic use
20.
ACS Med Chem Lett ; 3(10): 814-7, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-24900384

ABSTRACT

Ilicicolin H is a polyketide-nonribosomal peptide synthase (NRPS)-natural product isolated from Gliocadium roseum, which exhibits potent and broad spectrum antifungal activity, with sub-µg/mL MICs against Candida spp., Aspergillus fumigatus, and Cryptococcus spp. It showed a novel mode of action, potent inhibition (IC50 = 2-3 ng/mL) of the mitochondrial cytochrome bc1 reductase, and over 1000-fold selectivity relative to rat liver cytochrome bc1 reductase. Ilicicolin H exhibited in vivo efficacy in murine models of Candida albicans and Cryptococcus neoformans infections, but efficacy may have been limited by high plasma protein binding. Systematic structural modification of ilicicolin H was undertaken to understand the structural requirement for the antifungal activity. The details of the biological activity of ilicicolin H and structural modification of some of the key parts of the molecule and resulting activity of the derivatives are discussed. These data suggest that the ß-keto group is critical for the antifungal activity.

SELECTION OF CITATIONS
SEARCH DETAIL