Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
ACS Infect Dis ; 6(12): 3163-3173, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33164482

ABSTRACT

The rise of bacterial antibiotic resistance coupled with a diminished antibiotic drug pipeline underlines the importance of developing rational strategies to discover new antimicrobials. Microbially derived natural products are the basis for most of the antibiotic arsenal available to modern medicine. Here, we demonstrate a resistance-based approach to identify producers of elfamycins, an under-explored class of natural product antibiotics that target the essential translation factor EF-Tu. Antibiotic producers carry self-resistance genes to avoid suicide. These genes are often found within the same biosynthetic gene cluster (BGC) responsible for making the antibiotic, and we exploited this trait to identify members of the kirromycin class of elfamycin producers. Genome mining of Streptomyces spp. led to the identification of three isolates that harbor kirromycin-resistant EF-Tu (EF-TuKirR) within predicted natural product BGCs. Activity-guided purification on extracts of one of the Streptomyces isolates, which was not known to produce an elfamycin, identified it as a producer of phenelfamycin B, a linear polyketide. Phenelfamycin B demonstrates impressive antibacterial activity (MIC ∼ 1 µg/mL) against multidrug-resistant Neisseria gonorrhoeae, a clinically important Gram negative pathogen. The antigonococcal activity of phenelfamycin was shown to be the result of inhibition of protein biosynthesis by binding to EF-Tu. These results indicate that a resistance-based approach of identifying elfamycin producers is translatable to other antibiotic classes that can identify new and overlooked antibiotics necessary to address the antibiotic crisis.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Streptomyces , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Humans , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factor Tu/metabolism , Protein Biosynthesis/drug effects , Streptomyces/genetics
3.
J Med Chem ; 50(4): 755-64, 2007 Feb 22.
Article in English | MEDLINE | ID: mdl-17300162

ABSTRACT

The biosynthesis of methionine is an attractive antibiotic target given its importance in protein and DNA metabolism and its absence in mammals. We have performed a high-throughput screen of the methionine biosynthesis enzyme cystathionine beta-lyase (CBL) against a library of 50 000 small molecules and have identified several compounds that inhibit CBL enzyme activity in vitro. These hit molecules were of two classes: those that blocked CBL activity with mixed steady-state inhibition and those that covalently interacted with the enzyme at the active site pyridoxal phosphate cofactor with slow-binding inhibition kinetics. We determined the crystal structure of one of the slow-binding inhibitors in complex with CBL and used this structure as a guide in the synthesis of a small, focused library of analogues, some of which had improved enzyme inhibition properties. These studies provide the first lead molecules for antimicrobial agents that target cystathionine beta-lyase in methionine biosynthesis.


Subject(s)
Anti-Infective Agents/chemical synthesis , Bacteria/enzymology , Benzamides/chemical synthesis , Hydrazines/chemical synthesis , Lyases/antagonists & inhibitors , Lyases/chemistry , Models, Molecular , Quantitative Structure-Activity Relationship , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Benzamides/chemistry , Benzamides/pharmacology , Candida albicans/drug effects , Crystallography, X-Ray , Escherichia coli/enzymology , Hydrazines/chemistry , Hydrazines/pharmacology , Lyases/genetics , Microbial Sensitivity Tests , Salmonella typhi/enzymology
4.
Chem Biol ; 13(4): 437-41, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16632256

ABSTRACT

The lipopolysaccharide (LPS)-rich outer membrane of gram-negative bacteria provides a protective barrier that insulates these organisms from the action of numerous antibiotics. Breach of the LPS layer can therefore provide access to the cell interior to otherwise impermeant toxic molecules and can expose vulnerable binding sites for immune system components such as complement. Inhibition of LPS biosynthesis, leading to a truncated LPS molecule, is an alternative strategy for antibacterial drug development in which this vital cellular structure is weakened. A significant challenge for in vitro screens of small molecules for inhibition of LPS biosynthesis is the difficulty in accessing the complex carbohydrate substrates. We have optimized an assay of the enzymes required for LPS heptose biosynthesis that simultaneously surveys five enzyme activities by using commercially available substrates and report its use in a small-molecule screen that identifies an inhibitor of heptose synthesis.


Subject(s)
Adenosine Diphosphate Sugars/biosynthesis , Enzyme Inhibitors/pharmacology , Glycosyltransferases/antagonists & inhibitors , Lipopolysaccharides/biosynthesis , Cell Membrane/drug effects , Cell Membrane/metabolism , Drug Evaluation, Preclinical , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/metabolism , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/enzymology , Gram-Negative Bacteria/metabolism , Kinetics , Microbial Sensitivity Tests , Multienzyme Complexes/antagonists & inhibitors , Nucleotidyltransferases/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Recombinant Proteins/antagonists & inhibitors
5.
Chem Biol ; 13(2): 171-81, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16492565

ABSTRACT

The unique glycopeptide antibiotic A47934, produced by Streptomyces toyocaensis, possesses a nonglycosylated heptapeptide core that is sulfated on the phenolic hydroxyl of the N-terminal 4-hydroxy-L-phenylglycine residue. Genetic and biochemical experiments confirmed that StaL is a sulfotransferase capable of sulfating the predicted crosslinked heptapeptide substrate to produce A47934 both in vivo and in vitro. Incubation of purified His(6)-StaL with various substrates in vitro revealed substrate specificity and yielded two sulfo-glycopeptide antibiotics: sulfo-teicoplanin aglycone and sulfo-teicoplanin. Quantification of the antibacterial activity of desulfo-A47934, A47934, teicoplanin, and sulfo-teicoplanin demonstrated that sulfation slightly increased the minimum inhibitory concentration. This unique modification by sulfation expands glycopeptide diversity with potential application for the development of new antibiotics.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Glycopeptides/chemistry , Sulfates/chemistry , Sulfotransferases/metabolism , Anti-Bacterial Agents/chemistry , Base Sequence , Blotting, Southern , Chromatography, High Pressure Liquid , DNA Primers , Streptomyces/enzymology , Streptomyces/metabolism
6.
Chem Biol ; 12(2): 229-35, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15734650

ABSTRACT

Streptogramin antibiotics are comprised of two distinct chemical components: the type A polyketides and the type B cyclic depsipeptides. Clinical resistance to the type B streptogramins can occur via enzymatic degradation catalyzed by the lyase Vgb or by target modification through the action of Erm ribosomal RNA methyltransferases. We have prepared through chemical and chemo-enzymatic approaches a series of chimeric antibiotics composed of elements of type B streptogramins and the membrane-active antibiotic tyrocidine that evade these resistance mechanisms. These new compounds show broad antibiotic activity against gram-positive bacteria including a number of important pathogens, and chimeras appear to function by a mechanism that is distinct from their parent antibiotics. These results allow for the development of a brand new class of antibiotics with the ability to evade type B streptogramin-resistance mechanisms.


Subject(s)
Anti-Bacterial Agents/chemistry , Streptogramins/chemistry , Tyrocidine/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Drug Resistance, Bacterial , Methyltransferases/metabolism , Microbial Sensitivity Tests , Molecular Structure , Stereoisomerism , Streptogramins/pharmacology , Tyrocidine/pharmacology
7.
J Biol Chem ; 279(50): 52346-52, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15452119

ABSTRACT

The tetracycline antibiotics block microbial translation and constitute an important group of antimicrobial agents that find broad clinical utility. Resistance to this class of antibiotics is primarily the result of active efflux or ribosomal protection; however, a novel mechanism of resistance has been reported to be oxygen-dependent destruction of the drugs catalyzed by the enzyme TetX. Paradoxically, the tetX genes have been identified on transposable elements found in anaerobic bacteria of the genus Bacteroides. Overexpression of recombinant TetX in Escherichia coli followed by protein purification revealed a stoichiometric complex with flavin adenine dinucleotide. Reconstitution of in vitro enzyme activity demonstrated a broad tetracycline antibiotic spectrum and a requirement for molecular oxygen and NADPH in antibiotic degradation. The tetracycline products of TetX activity were unstable at neutral pH, but mass spectral and NMR characterization under acidic conditions supported initial monohydroxylation at position 11a followed by intramolecular cyclization and non-enzymatic breakdown to other undefined products. TetX is therefore a FAD-dependent monooxygenase. The enzyme not only catalyzed efficient degradation of a broad range of tetracycline analogues but also conferred resistance to these antibiotics in vivo. This is the first molecular characterization of an antibiotic-inactivating monooxygenase, the origins of which may lie in environmental bacteria.


Subject(s)
Bacterial Proteins/metabolism , Bacteroides fragilis/drug effects , Bacteroides fragilis/enzymology , Oxygenases/metabolism , Bacterial Proteins/genetics , Bacteroides fragilis/genetics , Escherichia coli/genetics , Flavoproteins/genetics , Flavoproteins/metabolism , Kinetics , Nuclear Magnetic Resonance, Biomolecular , Oxygenases/genetics , Oxytetracycline/chemistry , Oxytetracycline/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tetracycline Resistance/genetics
8.
Anal Biochem ; 324(1): 100-5, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14654051

ABSTRACT

A rapid and stoichiometric method for the synthesis of analogues of coenzyme A is described. The method links the enzymes pantothenate kinase, phosphopantotheine adenylyltransferase, and dephosphocoenzyme A kinase in vitro to generate a variety of CoA analogues from chemically synthesized pantothenic acid derivatives. The Escherichia coli CoA biosynthetic enzymes were overexpressed as hexa-histidine-tagged proteins, providing an abundant source of pure active catalysts for the reaction. The synthesis of five novel CoA derivatives is reported and the method is shown to be robust and tolerant of a number of different pantothenic acid structures, which indicates that the procedure should be widely applicable.


Subject(s)
Coenzyme A/biosynthesis , Coenzyme A/chemical synthesis , Base Sequence , Catalysis , Chromatography, High Pressure Liquid , Cloning, Molecular , Coenzyme A/chemistry , Coenzyme A/genetics , Coenzyme A/metabolism , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Escherichia coli/metabolism , Molecular Sequence Data , Molecular Structure , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Pantothenic Acid/analogs & derivatives , Pantothenic Acid/chemistry , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Recombinant Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...