Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 61(4): 2367-2389, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37874479

ABSTRACT

Structural epilepsies display complex immune activation signatures. However, it is unclear which neuroinflammatory pathways drive pathobiology. Transcriptome studies of brain resections from mesial temporal lobe epilepsy (mTLE) patients revealed a dysregulation of transforming growth factor ß, interferon α/ß, and nuclear factor erythroid 2-related factor 2 pathways. Since these pathways are regulated by ubiquitin-specific proteases (USP), in particular USP15, we hypothesized that USP15 blockade may provide therapeutic relief in treatment-resistant epilepsies. For validation, transgenic mice which either constitutively or inducibly lack Usp15 gene expression underwent intrahippocampal kainate injections to induce mTLE. We show that the severity of status epilepticus is unaltered in mice constitutively lacking Usp15 compared to wild types. Cell death, reactive gliosis, and changes in the inflammatory transcriptome were pronounced at 4 days after kainate injection. However, these brain inflammation signatures did not differ between genotypes. Likewise, induced deletion of Usp15 in chronic epilepsy did not affect seizure generation, cell death, gliosis, or the transcriptome. Concordantly, siRNA-mediated knockdown of Usp15 in a microglial cell line did not impact inflammatory responses in the form of cytokine release. Our data show that a lack of USP15 is insufficient to modulate the expression of relevant neuroinflammatory pathways in an mTLE mouse model and do not support targeting USP15 as a therapeutic approach for pharmacoresistant epilepsy.


Subject(s)
Epilepsy, Temporal Lobe , Animals , Humans , Mice , Down-Regulation , Gliosis , Hippocampus/metabolism , Kainic Acid , Mice, Transgenic , Ubiquitin-Specific Proteases/metabolism
2.
Chembiochem ; 20(13): 1701-1711, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30856684

ABSTRACT

Enzyme-catalyzed ß-lactone formation from ß-hydroxy acids is a crucial step in bacterial biosynthesis of ß-lactone natural products and membrane hydrocarbons. We developed a novel, continuous assay for ß-lactone synthetase activity using synthetic ß-hydroxy acid substrates with alkene or alkyne moieties. ß-Lactone formation is followed by rapid decarboxylation to form a conjugated triene chromophore for real-time evaluation by UV/Vis spectroscopy. The assay was used to determine steady-state kinetics of a long-chain ß-lactone synthetase, OleC, from the plant pathogen Xanthomonas campestris. Site-directed mutagenesis was used to test the involvement of conserved active site residues in Mg2+ and ATP binding. A previous report suggested OleC adenylated the substrate hydroxy group. Here we present several lines of evidence, including hydroxylamine trapping of the AMP intermediate, to demonstrate the substrate carboxyl group is adenylated prior to making the ß-lactone final product. A panel of nine substrate analogues were used to investigate the substrate specificity of X. campestris OleC by HPLC and GC-MS. Stereoisomers of 2-hexyl-3hydroxyoctanoic acid were synthesized and OleC preferred the (2R,3S) diastereomer consistent with the stereo-preference of upstream and downstream pathway enzymes. This biochemical knowledge was used to guide phylogenetic analysis of the ß-lactone synthetases to map their functional diversity within the acyl-CoA synthetase, NRPS adenylation domain, and luciferase superfamily.


Subject(s)
Carbon-Oxygen Lyases/chemistry , Carbon-Oxygen Lyases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Carbon-Oxygen Lyases/genetics , Catalysis , Catalytic Domain/genetics , Enzyme Assays/methods , Hydroxy Acids/metabolism , Kinetics , Magnesium/metabolism , Models, Chemical , Mutagenesis, Site-Directed , Phylogeny , Protein Binding , Sequence Alignment , Substrate Specificity , Xanthomonas campestris/enzymology
3.
ACS Chem Biol ; 13(11): 3184-3192, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30289689

ABSTRACT

Benzothiazinones (BTZ) are highly potent bactericidal inhibitors of mycobacteria and the lead compound, BTZ043, and the optimized drug candidate, PBTZ169, have potential for the treatment of tuberculosis. Here, we exploited the tractability of the BTZ scaffold by attaching a range of fluorophores to the 2-substituent of the BTZ ring via short linkers. We show by means of fluorescence imaging that the most advanced derivative, JN108, is capable of efficiently labeling its target, the essential flavoenzyme DprE1, both in cell-free extracts and after purification as well as in growing cells of different actinobacterial species. DprE1 displays a polar localization in Mycobacterium tuberculosis, M. marinum, M. smegmatis, and Nocardia farcinica but not in Corynebacterium glutamicum. Finally, mutation of the cysteine residue in DprE1 in these species, to which BTZ covalently binds, abolishes completely the interaction with JN108, thereby highlighting the specificity of this fluorescent probe.


Subject(s)
Affinity Labels/pharmacology , Alcohol Oxidoreductases/antagonists & inhibitors , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Thiazines/pharmacology , Actinomycetales/drug effects , Actinomycetales/enzymology , Affinity Labels/chemical synthesis , Alcohol Oxidoreductases/genetics , Antitubercular Agents/chemical synthesis , Bacterial Proteins/genetics , Cell Membrane/metabolism , Drug Design , Enzyme Inhibitors/chemical synthesis , Fluoresceins/chemical synthesis , Fluoresceins/pharmacology , Fluorescence , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/pharmacology , Hep G2 Cells , Humans , Microbial Sensitivity Tests , Microscopy, Fluorescence/methods , Mutation , Thiazines/chemical synthesis
4.
Antimicrob Agents Chemother ; 60(11): 6451-6459, 2016 11.
Article in English | MEDLINE | ID: mdl-27527085

ABSTRACT

Benzothiazinones (BTZs) are a class of compounds found to be extremely potent against both drug-susceptible and drug-resistant Mycobacterium tuberculosis strains. The potency of BTZs is explained by their specificity for their target decaprenylphosphoryl-d-ribose oxidase (DprE1), in particular by covalent binding of the activated form of the compound to the critical cysteine 387 residue of the enzyme. To probe the role of C387, we used promiscuous site-directed mutagenesis to introduce other codons at this position into dprE1 of M. tuberculosis The resultant viable BTZ-resistant mutants were characterized in vitro, ex vivo, and biochemically to gain insight into the effects of these mutations on DprE1 function and on M. tuberculosis Five different mutations (C387G, C387A, C387S, C387N, and C387T) conferred various levels of resistance to BTZ and exhibited different phenotypes. The C387G and C387N mutations resulted in a lower growth rate of the mycobacterium on solid medium, which could be attributed to the significant decrease in the catalytic efficiency of the DprE1 enzyme. All five mutations rendered the mycobacterium less cytotoxic to macrophages. Finally, differences in the potencies of covalent and noncovalent DprE1 inhibitors in the presence of C387 mutations were revealed by enzymatic assays. As expected from the mechanism of action, the covalent inhibitor PBTZ169 only partially inhibited the mutant DprE1 enzymes compared to the near-complete inhibition with a noncovalent DprE1 inhibitor, Ty38c. This study emphasizes the importance of the C387 residue for DprE1 activity and for the killing action of covalent inhibitors such as BTZs and other recently identified nitroaromatic inhibitors.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Resistance, Multiple, Bacterial/genetics , Mutation , Mycobacterium tuberculosis/drug effects , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Amino Acid Substitution , Antitubercular Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Cysteine/chemistry , Cysteine/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , Macrophages/microbiology , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/growth & development , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Phenotype , Piperazines/chemistry , Piperazines/pharmacology , Structure-Activity Relationship , Thiazines/chemistry , Thiazines/pharmacology , Tuberculosis, Multidrug-Resistant/microbiology
5.
ChemMedChem ; 11(3): 331-9, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26751718

ABSTRACT

Nitroarenes are less preferred in drug discovery due to their potential to be mutagenic. However, several nitroarenes were shown to be promising antitubercular agents with specific modes of action, namely, nitroimidazoles and benzothiazinones. The nitro group in these compounds is activated through different mechanisms, both enzymatic and non-enzymatic, in mycobacteria prior to binding to the target of interest. From a whole-cell screening program, we identified a novel lead nitrobenzothiazole (BT) series that acts by inhibition of decaprenylphosphoryl-ß-d-ribose 2'-epimerase (DprE1) of Mycobacterium tuberculosis (Mtb). The lead was found to be mutagenic to start with. Our efforts to mitigate mutagenicity resulted in the identification of 6-methyl-7-nitro-5-(trifluoromethyl)-1,3-benzothiazoles (cBTs), a novel class of antitubercular agents that are non-mutagenic and exhibit an improved safety profile. The methyl group ortho to the nitro group decreases the electron affinity of the series, and is hence responsible for the non-mutagenic nature of these compounds. Additionally, the co-crystal structure of cBT in complex with Mtb DprE1 established the mode of binding. This investigation led to a new non-mutagenic antitubercular agent and demonstrates that the mutagenic nature of nitroarenes can be solved by modulation of stereoelectronic properties.


Subject(s)
Antitubercular Agents/pharmacology , Benzothiazoles/pharmacology , Mutagens/chemistry , Mycobacterium tuberculosis/drug effects , Nitro Compounds/pharmacology , Antitubercular Agents/adverse effects , Antitubercular Agents/chemistry , Benzothiazoles/adverse effects , Benzothiazoles/chemistry , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Nitro Compounds/adverse effects , Nitro Compounds/chemistry , Stereoisomerism , Structure-Activity Relationship
6.
Bioorg Med Chem ; 23(24): 7694-710, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26643218

ABSTRACT

We report the discovery of benzothiazoles, a novel anti-mycobacterial series, identified from a whole cell based screening campaign. Benzothiazoles exert their bactericidal activity against Mycobacterium tuberculosis (Mtb) through potent inhibition of decaprenylphosphoryl-ß-d-ribose 2'-oxidase (DprE1), the key enzyme involved in arabinogalactan synthesis. Specific target linkage and mode of binding were established using co-crystallization and protein mass spectrometry studies. Most importantly, the current study provides insights on the utilization of systematic medicinal chemistry approaches to mitigate safety liabilities while improving potency during progression from an initial genotoxic hit, the benzothiazole N-oxides (BTOs) to the lead-like AMES negative, crowded benzothiazoles (cBTs). These findings offer opportunities for development of safe clinical candidates against tuberculosis. The design strategy adopted could find potential application in discovery of safe drugs in other therapy areas too.


Subject(s)
Alcohol Oxidoreductases/metabolism , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Alcohol Oxidoreductases/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Drug Design , Humans , Molecular Docking Simulation , Structure-Activity Relationship , Tuberculosis/drug therapy , Tuberculosis/microbiology
7.
Antimicrob Agents Chemother ; 59(8): 4446-52, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25987616

ABSTRACT

8-Nitro-benzothiazinones (BTZs), such as BTZ043 and PBTZ169, inhibit decaprenylphosphoryl-ß-d-ribose 2'-oxidase (DprE1) and display nanomolar bactericidal activity against Mycobacterium tuberculosis in vitro. Structure-activity relationship (SAR) studies revealed the 8-nitro group of the BTZ scaffold to be crucial for the mechanism of action, which involves formation of a semimercaptal bond with Cys387 in the active site of DprE1. To date, substitution of the 8-nitro group has led to extensive loss of antimycobacterial activity. Here, we report the synthesis and characterization of the pyrrole-benzothiazinones PyrBTZ01 and PyrBTZ02, non-nitro-benzothiazinones that retain significant antimycobacterial activity, with MICs of 0.16 µg/ml against M. tuberculosis. These compounds inhibit DprE1 with 50% inhibitory concentration (IC50) values of <8 µM and present favorable in vitro absorption-distribution-metabolism-excretion/toxicity (ADME/T) and in vivo pharmacokinetic profiles. The most promising compound, PyrBTZ01, did not show efficacy in a mouse model of acute tuberculosis, suggesting that BTZ-mediated killing through DprE1 inhibition requires a combination of both covalent bond formation and compound potency.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Piperazines/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Spiro Compounds/pharmacology , Thiazines/pharmacology , Animals , Antitubercular Agents/pharmacology , Catalytic Domain/drug effects , Disease Models, Animal , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests/methods , Mycobacterium tuberculosis/metabolism , Structure-Activity Relationship , Tuberculosis/drug therapy , Tuberculosis/metabolism
8.
ACS Chem Biol ; 10(7): 1631-6, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-25906160

ABSTRACT

The flavo-enzyme DprE1 catalyzes a key epimerization step in the decaprenyl-phosphoryl d-arabinose (DPA) pathway, which is essential for mycobacterial cell wall biogenesis and targeted by several new tuberculosis drug candidates. Here, using differential radiolabeling with DPA precursors and high-resolution fluorescence microscopy, we disclose the unexpected extracytoplasmic localization of DprE1 and periplasmic synthesis of DPA. Collectively, this explains the vulnerability of DprE1 and the remarkable potency of the best inhibitors.


Subject(s)
Alcohol Oxidoreductases/analysis , Alcohol Oxidoreductases/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/analysis , Bacterial Proteins/metabolism , Cell Wall/metabolism , Mycobacterium tuberculosis/cytology , Mycobacterium tuberculosis/enzymology , Tuberculosis/microbiology , Cell Wall/drug effects , Humans , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy
9.
ACS Chem Biol ; 10(3): 705-14, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25427196

ABSTRACT

Phenotypic screening of a quinoxaline library against replicating Mycobacterium tuberculosis led to the identification of lead compound Ty38c (3-((4-methoxybenzyl)amino)-6-(trifluoromethyl)quinoxaline-2-carboxylic acid). With an MIC99 and MBC of 3.1 µM, Ty38c is bactericidal and active against intracellular bacteria. To investigate its mechanism of action, we isolated mutants resistant to Ty38c and sequenced their genomes. Mutations were found in rv3405c, coding for the transcriptional repressor of the divergently expressed rv3406 gene. Biochemical studies clearly showed that Rv3406 decarboxylates Ty38c into its inactive keto metabolite. The actual target was then identified by isolating Ty38c-resistant mutants of an M. tuberculosis strain lacking rv3406. Here, mutations were found in dprE1, encoding the decaprenylphosphoryl-d-ribose oxidase DprE1, essential for biogenesis of the mycobacterial cell wall. Genetics, biochemical validation, and X-ray crystallography revealed Ty38c to be a noncovalent, noncompetitive DprE1 inhibitor. Structure-activity relationship studies generated a family of DprE1 inhibitors with a range of IC50's and bactericidal activity. Co-crystal structures of DprE1 in complex with eight different quinoxaline analogs provided a high-resolution interaction map of the active site of this extremely vulnerable target in M. tuberculosis.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/drug effects , Quinoxalines/pharmacology , Small Molecule Libraries/pharmacology , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Antitubercular Agents/chemical synthesis , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cell Wall/drug effects , Cell Wall/enzymology , Crystallography, X-Ray , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Gene Expression , Hydrogen Bonding , Microbial Sensitivity Tests , Models, Molecular , Mutation , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/growth & development , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Quinoxalines/chemical synthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship
10.
J Med Chem ; 57(12): 5419-34, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24871036

ABSTRACT

4-Aminoquinolone piperidine amides (AQs) were identified as a novel scaffold starting from a whole cell screen, with potent cidality on Mycobacterium tuberculosis (Mtb). Evaluation of the minimum inhibitory concentrations, followed by whole genome sequencing of mutants raised against AQs, identified decaprenylphosphoryl-ß-d-ribose 2'-epimerase (DprE1) as the primary target responsible for the antitubercular activity. Mass spectrometry and enzyme kinetic studies indicated that AQs are noncovalent, reversible inhibitors of DprE1 with slow on rates and long residence times of ∼100 min on the enzyme. In general, AQs have excellent leadlike properties and good in vitro secondary pharmacology profile. Although the scaffold started off as a single active compound with moderate potency from the whole cell screen, structure-activity relationship optimization of the scaffold led to compounds with potent DprE1 inhibition (IC50 < 10 nM) along with potent cellular activity (MIC = 60 nM) against Mtb.


Subject(s)
Amides/chemistry , Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Oxidoreductases/antagonists & inhibitors , Piperidines/chemistry , Quinolones/chemistry , Alcohol Oxidoreductases , Amides/pharmacokinetics , Amides/pharmacology , Animals , Antitubercular Agents/pharmacokinetics , Antitubercular Agents/pharmacology , Catalytic Domain , Cell Line, Tumor , Drug Resistance, Bacterial , Genome, Bacterial , Humans , Kinetics , Microbial Sensitivity Tests , Molecular Docking Simulation , Mutation , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Piperidines/pharmacokinetics , Piperidines/pharmacology , Protein Binding , Quinolones/pharmacokinetics , Quinolones/pharmacology , Rats, Wistar , Stereoisomerism , Structure-Activity Relationship
11.
Carbohydr Res ; 391: 61-5, 2014 Jun 04.
Article in English | MEDLINE | ID: mdl-24785389

ABSTRACT

Epimerisation between ribofuranose and arabinofuranose sugars is crucial in several biosynthetic pathways, but is typically challenging to monitor. Here, we have screened for fluorescent boronic acids that can be used as molecular probes for the specific detection of ribofuranose over arabinofuranose sugars in solution. We show excellent specificity of the fluorescent response of 3-biphenylboronic acid to ribofuranose at physiological pH. This provides a tool for in situ monitoring of carbohydrate modifying enzymes and provides a viable alternative to traditional radiolabelled assays.


Subject(s)
Molecular Probes/analysis , Molecular Probes/chemistry , Pentoses/analysis , Pentoses/chemistry , Arabinose/analogs & derivatives , Arabinose/analysis , Arabinose/chemistry , Boronic Acids/analysis , Boronic Acids/chemistry , Fluorescence , Hydrogen-Ion Concentration , Molecular Structure
12.
EMBO Mol Med ; 6(3): 372-83, 2014 03.
Article in English | MEDLINE | ID: mdl-24500695

ABSTRACT

The benzothiazinone lead compound, BTZ043, kills Mycobacterium tuberculosis by inhibiting the essential flavo-enzyme DprE1, decaprenylphosphoryl-beta-D-ribose 2-epimerase. Here, we synthesized a new series of piperazine-containing benzothiazinones (PBTZ) and show that, like BTZ043, the preclinical candidate PBTZ169 binds covalently to DprE1. The crystal structure of the DprE1-PBTZ169 complex reveals formation of a semimercaptal adduct with Cys387 in the active site and explains the irreversible inactivation of the enzyme. Compared to BTZ043, PBTZ169 has improved potency, safety and efficacy in zebrafish and mouse models of tuberculosis (TB). When combined with other TB drugs, PBTZ169 showed additive activity against M. tuberculosis in vitro except with bedaquiline (BDQ) where synergy was observed. A new regimen comprising PBTZ169, BDQ and pyrazinamide was found to be more efficacious than the standard three drug treatment in a murine model of chronic disease. PBTZ169 is thus an attractive drug candidate to treat TB in humans.


Subject(s)
Antitubercular Agents/therapeutic use , Spiro Compounds/therapeutic use , Thiazines/therapeutic use , Tuberculosis/drug therapy , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/metabolism , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Disease Models, Animal , Embryo, Nonmammalian/drug effects , Hep G2 Cells , Humans , Lung/metabolism , Mice , Molecular Dynamics Simulation , Mycobacterium tuberculosis/drug effects , Piperazines/chemistry , Piperazines/pharmacology , Piperazines/therapeutic use , Spiro Compounds/chemistry , Spiro Compounds/pharmacokinetics , Spiro Compounds/pharmacology , Spleen/metabolism , Thiazines/chemistry , Thiazines/pharmacokinetics , Thiazines/pharmacology , Zebrafish/growth & development
13.
Curr Pharm Des ; 20(27): 4379-403, 2014.
Article in English | MEDLINE | ID: mdl-24245764

ABSTRACT

Several groups working in the field of the development of new antituberculosis drugs have recently reported active compounds targeting mycobacterial enzyme DprE1. Along with its counterpart, DprE2, it catalyses a unique epimerization reaction resulting in the synthesis of decaprenylphosphoryl arabinose, the single donor of arabinosyl residues for the build-up of arabinans, fundamental components of the mycobacterial cell wall. This review presents the historical background leading to the discovery of DprE1, focusing on the biochemical and structural characterization of this important emerging target and introducing the molecules acting on DprE1 including the development of the most successful series--the benzothiazinones, currently in late pre-clinical development, which turned to be suicide inhibitors of DprE1.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Discovery/methods , Mycobacterium tuberculosis/drug effects , Thiazines/pharmacology , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Antitubercular Agents/chemistry , Antitubercular Agents/therapeutic use , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cell Wall/drug effects , Cell Wall/metabolism , Humans , Models, Molecular , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Polysaccharides/biosynthesis , Protein Conformation , Structure-Activity Relationship , Thiazines/chemistry , Thiazines/therapeutic use , Tuberculosis, Multidrug-Resistant/drug therapy , Tuberculosis, Multidrug-Resistant/microbiology
14.
Nat Chem Biol ; 10(2): 96-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24292073

ABSTRACT

Pyridomycin, a natural product with potent antituberculosis activity, inhibits a major drug target, the InhA enoyl reductase. Here, we unveil the co-crystal structure and unique ability of pyridomycin to block both the NADH cofactor- and lipid substrate-binding pockets of InhA. This is to our knowledge a first-of-a-kind binding mode that discloses a new means of InhA inhibition. Proof-of-principle studies show how structure-assisted drug design can improve the activity of new pyridomycin derivatives.


Subject(s)
Antitubercular Agents/chemistry , Bacterial Proteins/chemistry , NAD/chemistry , Oligopeptides/chemistry , Oxidoreductases/chemistry , Binding Sites , Crystallography, X-Ray , Models, Molecular , Substrate Specificity
15.
Biochemistry ; 52(21): 3740-51, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23672572

ABSTRACT

Trypanosoma cruzitrans-sialidase (TcTS), which catalyzes the transfer or hydrolysis of terminal sialic acid residues, is crucial to the development and proliferation of the T. cruzi parasite and thus has emerged as a potential drug target for the treatment of Chagas disease. We here probe the origin of the observed preference for the transfer reaction over hydrolysis where the substrate for TcTS is the natural sialyl donor (represented in this work by sialyllactose). Thus, acceptor lactose preferentially attacks the sialyl-enyzme intermediate rather than water. We compare this with the weaker preference for such transfer shown by a synthetic donor substrate, 4-methylumbelliferyl α-d-acetylneuraminide. For this reason, we conducted molecular dynamics simulations of TcTS following its sialylation by the substrate to examine the behavior of the asialyl leaving group by the protein. These simulations indicate that, where lactose is released, this leaving group samples well-defined interactions in the acceptor site, some of which are mediated by localized water molecules; also, the extent of the opening of the acceptor site to solvent is reduced as compared with those of unliganded forms of TcTS. However, where there is release of 4-methylumbelliferone, this leaving group explores a range of transient poses; surrounding active site water is also more disordered. The acceptor site explores more open conformations, similar to the case in which the 4-methylumbelliferone is absent. Thus, the predicted solvent accessibility of sialylated TcTS is increased when 4-methylumbelliferyl α-d-acetylneuraminide is the substrate compared to sialyllactose; this in turn is likely to contribute to a greater propensity for hydrolysis of the covalent intermediate. These computational simulations, which suggest that protein flexibility has a role in the transferase/sialidase activity of TcTS, have the potential to aid in the design of anti-Chagas inhibitors effective against this neglected tropical disease.


Subject(s)
Glycoproteins/metabolism , Molecular Dynamics Simulation , Neuraminidase/metabolism , Trypanosoma cruzi/enzymology , Animals , Protein Conformation , Substrate Specificity
16.
J Med Chem ; 56(6): 2385-405, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23437866

ABSTRACT

Siderophores are small-molecule iron chelators produced by bacteria and other microorganisms for survival under iron limiting conditions such as found in a mammalian host. Siderophore biosynthesis is essential for the virulence of many important Gram-negative pathogens including Acinetobacter baumannii, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Escherichia coli. We performed high-throughput screening against BasE, which is involved in siderophore biosynthesis in A. baumannii, and identified 6-phenyl-1-(pyridin-4-ylmethyl)-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid 15. Herein we report the synthesis, biochemical, and microbiological evaluation of a systematic series of analogues of the HTS hit 15. Analogue 67 is the most potent analogue with a KD of 2 nM against BasE. Structural characterization of the inhibitors with BasE reveals that they bind in a unique orientation in the active site, occupying all three substrate binding sites, and thus can be considered as multisubstrate inhibitors. These results provide a foundation for future studies aimed at increasing both enzyme potency and antibacterial activity.


Subject(s)
Acinetobacter baumannii/enzymology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Siderophores/biosynthesis , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/metabolism , Microbial Sensitivity Tests , Models, Molecular , Protein Conformation , Structure-Activity Relationship
17.
EMBO Mol Med ; 4(10): 1032-42, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22987724

ABSTRACT

Tuberculosis, a global threat to public health, is becoming untreatable due to widespread drug resistance to frontline drugs such as the InhA-inhibitor isoniazid. Historically, by inhibiting highly vulnerable targets, natural products have been an important source of antibiotics including potent anti-tuberculosis agents. Here, we describe pyridomycin, a compound produced by Dactylosporangium fulvum with specific cidal activity against mycobacteria. By selecting pyridomycin-resistant mutants of Mycobacterium tuberculosis, whole-genome sequencing and genetic validation, we identified the NADH-dependent enoyl- (Acyl-Carrier-Protein) reductase InhA as the principal target and demonstrate that pyridomycin inhibits mycolic acid synthesis in M. tuberculosis. Furthermore, biochemical and structural studies show that pyridomycin inhibits InhA directly as a competitive inhibitor of the NADH-binding site, thereby identifying a new, druggable pocket in InhA. Importantly, the most frequently encountered isoniazid-resistant clinical isolates remain fully susceptible to pyridomycin, thus opening new avenues for drug development. →See accompanying article http://dx.doi.org/10.1002/emmm.201201811.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Biological Products/pharmacology , Micromonosporaceae/chemistry , Mycobacterium tuberculosis/drug effects , Oligopeptides/pharmacology , Oxidoreductases/antagonists & inhibitors , Antitubercular Agents/isolation & purification , Biological Products/isolation & purification , Biosynthetic Pathways/drug effects , Drug Resistance, Bacterial , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Mutation , Mycolic Acids/metabolism , Oligopeptides/isolation & purification , Selection, Genetic
18.
Sci Transl Med ; 4(150): 150ra121, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-22956199

ABSTRACT

The benzothiazinone BTZ043 is a tuberculosis drug candidate with nanomolar whole-cell activity. BTZ043 targets the DprE1 catalytic component of the essential enzyme decaprenylphosphoryl-ß-D-ribofuranose-2'-epimerase, thus blocking biosynthesis of arabinans, vital components of mycobacterial cell walls. Crystal structures of DprE1, in its native form and in a complex with BTZ043, reveal formation of a semimercaptal adduct between the drug and an active-site cysteine, as well as contacts to a neighboring catalytic lysine residue. Kinetic studies confirm that BTZ043 is a mechanism-based, covalent inhibitor. This explains the exquisite potency of BTZ043, which, when fluorescently labeled, localizes DprE1 at the poles of growing bacteria. Menaquinone can reoxidize the flavin adenine dinucleotide cofactor in DprE1 and may be the natural electron acceptor for this reaction in the mycobacterium. Our structural and kinetic analysis provides both insight into a critical epimerization reaction and a platform for structure-based design of improved inhibitors.


Subject(s)
Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Microbial Viability/drug effects , Mycobacterium tuberculosis/drug effects , Thiazines/chemistry , Thiazines/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Crystallography, X-Ray , Cysteine/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Flavin-Adenine Dinucleotide/analogs & derivatives , Flavin-Adenine Dinucleotide/metabolism , Flavoproteins/chemistry , Fluorescent Dyes/metabolism , Kinetics , Lysine/chemistry , Microbial Sensitivity Tests , Models, Molecular , Mutagenesis, Site-Directed , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , Oxidation-Reduction/drug effects , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/chemistry , Protein Structure, Tertiary , Protein Transport/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
19.
ChemMedChem ; 5(12): 2079-87, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21053346

ABSTRACT

A simple steady-state kinetic high-throughput assay was developed for the salicylate synthase MbtI from Mycobacterium tuberculosis, which catalyzes the first committed step of mycobactin biosynthesis. The mycobactins are small-molecule iron chelators produced by M. tuberculosis, and their biosynthesis has been identified as a promising target for the development of new antitubercular agents. The assay was miniaturized to a 384-well plate format and high-throughput screening was performed at the National Screening Laboratory for the Regional Centers of Excellence in Biodefense and Emerging Infectious Diseases (NSRB). Three classes of compounds were identified comprising the benzisothiazolones (class I), diarylsulfones (class II), and benzimidazole-2-thiones (class III). Each of these compound series was further pursued to investigate their biochemical mechanism and structure-activity relationships. Benzimidazole-2-thione 4 emerged as the most promising inhibitor owing to its potent reversible inhibition.


Subject(s)
Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Lyases/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Lyases/metabolism , Sulfones/chemical synthesis , Sulfones/chemistry , Sulfones/pharmacology , Thiazoles/chemical synthesis , Thiazoles/chemistry , Thiazoles/pharmacology
20.
Biochemistry ; 49(43): 9292-305, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20853905

ABSTRACT

The human pathogen Acinetobacter baumannii produces a siderophore called acinetobactin that is derived from one molecule each of threonine, histidine, and 2,3-dihydroxybenzoic acid (DHB). The activity of several nonribosomal peptide synthetase (NRPS) enzymes is used to combine the building blocks into the final molecule. The acinetobactin synthesis pathway initiates with a self-standing adenylation enzyme, BasE, that activates the DHB molecule and covalently transfers it to the pantetheine cofactor of an aryl-carrier protein of BasF, a strategy that is shared with many siderophore-producing NRPS clusters. In this reaction, DHB reacts with ATP to form the aryl adenylate and pyrophosphate. In a second partial reaction, the DHB is transferred to the carrier protein. Inhibitors of BasE and related enzymes have been identified that prevent growth of bacteria on iron-limiting media. Recently, a new inhibitor of BasE has been identified via high-throughput screening using a fluorescence polarization displacement assay. We present here biochemical and structural studies to examine the binding mode of this inhibitor. The kinetics of the wild-type BasE enzyme is shown, and inhibition studies demonstrate that the new compound exhibits competitive inhibition against both ATP and 2,3-dihydroxybenzoate. Structural examination of BasE bound to this inhibitor illustrates a novel binding mode in which the phenyl moiety partially fills the enzyme pantetheine binding tunnel. Structures of rationally designed bisubstrate inhibitors are also presented.


Subject(s)
Acinetobacter baumannii/enzymology , Enzyme Inhibitors/chemistry , Imidazoles/metabolism , Oxazoles/metabolism , Peptide Synthases/antagonists & inhibitors , Bacterial Proteins , Humans , Kinetics , Metabolic Networks and Pathways , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...