Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Nat Commun ; 12(1): 143, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420031

ABSTRACT

Coenzyme A (CoA) is a fundamental co-factor for all life, involved in numerous metabolic pathways and cellular processes, and its biosynthetic pathway has raised substantial interest as a drug target against multiple pathogens including Mycobacterium tuberculosis. The biosynthesis of CoA is performed in five steps, with the second and third steps being catalysed in the vast majority of prokaryotes, including M. tuberculosis, by a single bifunctional protein, CoaBC. Depletion of CoaBC was found to be bactericidal in M. tuberculosis. Here we report the first structure of a full-length CoaBC, from the model organism Mycobacterium smegmatis, describe how it is organised as a dodecamer and regulated by CoA thioesters. A high-throughput biochemical screen focusing on CoaB identified two inhibitors with different chemical scaffolds. Hit expansion led to the discovery of potent and selective inhibitors of M. tuberculosis CoaB, which we show to bind to a cryptic allosteric site within CoaB.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Carboxy-Lyases/antagonists & inhibitors , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/drug effects , Peptide Synthases/antagonists & inhibitors , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Antitubercular Agents/therapeutic use , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Carboxy-Lyases/ultrastructure , Coenzyme A/biosynthesis , Crystallography, X-Ray , Enzyme Assays , Gene Knockdown Techniques , High-Throughput Screening Assays , Humans , Microbial Sensitivity Tests , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Peptide Synthases/genetics , Peptide Synthases/metabolism , Peptide Synthases/ultrastructure , Tuberculosis/drug therapy , Tuberculosis/microbiology
2.
J Med Chem ; 64(1): 719-740, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33395287

ABSTRACT

Phenotypic screening of a Medicines for Malaria Venture compound library against Mycobacterium tuberculosis (Mtb) identified a cluster of pan-active 2-pyrazolylpyrimidinones. The biology triage of these actives using various tool strains of Mtb suggested a novel mechanism of action. The compounds were bactericidal against replicating Mtb and retained potency against clinical isolates of Mtb. Although selected MmpL3 mutant strains of Mtb showed resistance to these compounds, there was no shift in the minimum inhibitory concentration (MIC) against a mmpL3 hypomorph, suggesting mutations in MmpL3 as a possible resistance mechanism for the compounds but not necessarily as the target. RNA transcriptional profiling and the checkerboard board 2D-MIC assay in the presence of varying concentrations of ferrous salt indicated perturbation of the Fe-homeostasis by the compounds. Structure-activity relationship studies identified potent compounds with good physicochemical properties and in vitro microsomal metabolic stability with moderate selectivity over cytotoxicity against mammalian cell lines.


Subject(s)
Antitubercular Agents/chemistry , Pyrimidinones/chemistry , Animals , Antitubercular Agents/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Half-Life , Humans , Iron/metabolism , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Microsomes/metabolism , Mutation , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/isolation & purification , Pyrazoles/chemistry , Pyrimidinones/metabolism , Pyrimidinones/pharmacology , Rats , Structure-Activity Relationship
3.
Article in English | MEDLINE | ID: mdl-31383669

ABSTRACT

Mechanisms of magnesium homeostasis in Mycobacterium tuberculosis are poorly understood. Here, we describe the characterization of a pyrimidinetrione amide scaffold that disrupts magnesium homeostasis in the pathogen by direct binding to the CorA Mg2+/Co2+ transporter. Mutations in domains of CorA that are predicted to regulate the pore opening in response to Mg2+ ions conferred resistance to this scaffold. The pyrimidinetrione amides were cidal against the pathogen under both actively replicating and nonreplicating conditions in vitro and were efficacious against the organism during macrophage infection. However, the compound lacked efficacy in infected mice, possibly due to limited exposure. Our results indicate that inhibition of Mg2+ homeostasis by CorA is an attractive target for tuberculosis drug discovery and encourage identification of improved CorA inhibitors.


Subject(s)
Bacterial Proteins/metabolism , Cation Transport Proteins/metabolism , Magnesium/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Cation Transport Proteins/genetics , Homeostasis/drug effects , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Structure-Activity Relationship
4.
ACS Infect Dis ; 4(6): 954-969, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29522317

ABSTRACT

Mycobacterium tuberculosis ( MTb) possesses two nonproton pumping type II NADH dehydrogenase (NDH-2) enzymes which are predicted to be jointly essential for respiratory metabolism. Furthermore, the structure of a closely related bacterial NDH-2 has been reported recently, allowing for the structure-based design of small-molecule inhibitors. Herein, we disclose MTb whole-cell structure-activity relationships (SARs) for a series of 2-mercapto-quinazolinones which target the ndh encoded NDH-2 with nanomolar potencies. The compounds were inactivated by glutathione-dependent adduct formation as well as quinazolinone oxidation in microsomes. Pharmacokinetic studies demonstrated modest bioavailability and compound exposures. Resistance to the compounds in MTb was conferred by promoter mutations in the alternative nonessential NDH-2 encoded by ndhA in MTb. Bioenergetic analyses revealed a decrease in oxygen consumption rates in response to inhibitor in cells in which membrane potential was uncoupled from ATP production, while inverted membrane vesicles showed mercapto-quinazolinone-dependent inhibition of ATP production when NADH was the electron donor to the respiratory chain. Enzyme kinetic studies further demonstrated noncompetitive inhibition, suggesting binding of this scaffold to an allosteric site. In summary, while the initial MTb SAR showed limited improvement in potency, these results, combined with structural information on the bacterial protein, will aid in the future discovery of new and improved NDH-2 inhibitors.


Subject(s)
Mycobacterium tuberculosis/enzymology , NADH Dehydrogenase/chemistry , Quinazolinones/chemistry , Molecular Structure , NADH Dehydrogenase/antagonists & inhibitors , Quinazolinones/chemical synthesis , Quinazolinones/pharmacology , Structure-Activity Relationship
5.
J Med Chem ; 60(23): 9790-9806, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29125744

ABSTRACT

N-Myristoyltransferase (NMT) represents a promising drug target within the parasitic protozoa Trypanosoma brucei (T. brucei), the causative agent for human African trypanosomiasis (HAT) or sleeping sickness. We have previously validated T. brucei NMT as a promising druggable target for the treatment of HAT in both stages 1 and 2 of the disease. We report on the use of the previously reported DDD85646 (1) as a starting point for the design of a class of potent, brain penetrant inhibitors of T. brucei NMT.


Subject(s)
Acyltransferases/antagonists & inhibitors , Aminopyridines/chemistry , Aminopyridines/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/enzymology , Trypanosomiasis, African/drug therapy , Acyltransferases/metabolism , Aminopyridines/chemical synthesis , Aminopyridines/pharmacokinetics , Animals , Brain/metabolism , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Mice , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/pharmacokinetics , Trypanosomiasis, African/metabolism
7.
ACS Infect Dis ; 3(1): 18-33, 2017 01 13.
Article in English | MEDLINE | ID: mdl-27704782

ABSTRACT

A potent, noncytotoxic indazole sulfonamide was identified by high-throughput screening of >100,000 synthetic compounds for activity against Mycobacterium tuberculosis (Mtb). This noncytotoxic compound did not directly inhibit cell wall biogenesis but triggered a slow lysis of Mtb cells as measured by release of intracellular green fluorescent protein (GFP). Isolation of resistant mutants followed by whole-genome sequencing showed an unusual gene amplification of a 40 gene region spanning from Rv3371 to Rv3411c and in one case a potential promoter mutation upstream of guaB2 (Rv3411c) encoding inosine monophosphate dehydrogenase (IMPDH). Subsequent biochemical validation confirmed direct inhibition of IMPDH by an uncompetitive mode of inhibition, and growth inhibition could be rescued by supplementation with guanine, a bypass mechanism for the IMPDH pathway. Beads containing immobilized indazole sulfonamides specifically interacted with IMPDH in cell lysates. X-ray crystallography of the IMPDH-IMP-inhibitor complex revealed that the primary interactions of these compounds with IMPDH were direct pi-pi interactions with the IMP substrate. Advanced lead compounds in this series with acceptable pharmacokinetic properties failed to show efficacy in acute or chronic murine models of tuberculosis (TB). Time-kill experiments in vitro suggest that sustained exposure to drug concentrations above the minimum inhibitory concentration (MIC) for 24 h were required for a cidal effect, levels that have been difficult to achieve in vivo. Direct measurement of guanine levels in resected lung tissue from tuberculosis-infected animals and patients revealed 0.5-2 mM concentrations in caseum and normal lung tissue. The high lesional levels of guanine and the slow lytic, growth-rate-dependent effect of IMPDH inhibition pose challenges to developing drugs against this target for use in treating TB.


Subject(s)
Antitubercular Agents/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Sulfonamides/pharmacology , Animals , Drug Design , Drug Discovery , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Mutation , Protein Conformation , Rabbits , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Tuberculosis/drug therapy
8.
Antimicrob Agents Chemother ; 58(11): 6962-5, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25155596

ABSTRACT

We report here a series of five chemically diverse scaffolds that have in vitro activities on replicating and hypoxic nonreplicating bacilli by targeting the respiratory bc1 complex in Mycobacterium tuberculosis in a strain-dependent manner. Deletion of the cytochrome bd oxidase generated a hypersusceptible mutant in which resistance was acquired by a mutation in qcrB. These results highlight the promiscuity of the bc1 complex and the risk of targeting energy metabolism with new drugs.


Subject(s)
Antitubercular Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Electron Transport Complex IV/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Binding Sites , Electron Transport/drug effects , Electron Transport Complex IV/genetics , Energy Metabolism/drug effects , Gene Knockout Techniques , Microbial Sensitivity Tests , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Oxazines/chemistry , Protein Structure, Tertiary , Pyridines/pharmacology , Xanthenes/chemistry
9.
J Med Chem ; 55(1): 140-52, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22148754

ABSTRACT

N-Myristoyltransferase (NMT) represents a promising drug target for human African trypanosomiasis (HAT), which is caused by the parasitic protozoa Trypanosoma brucei. We report the optimization of a high throughput screening hit (1) to give a lead molecule DDD85646 (63), which has potent activity against the enzyme (IC(50) = 2 nM) and T. brucei (EC(50) = 2 nM) in culture. The compound has good oral pharmacokinetics and cures rodent models of peripheral HAT infection. This compound provides an excellent tool for validation of T. brucei NMT as a drug target for HAT as well as a valuable lead for further optimization.


Subject(s)
Acyltransferases/antagonists & inhibitors , Aminopyridines/chemical synthesis , Sulfonamides/chemical synthesis , Trypanocidal Agents/chemical synthesis , Administration, Oral , Aminopyridines/pharmacokinetics , Aminopyridines/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Crystallography, X-Ray , Databases, Factual , Humans , Models, Molecular , Molecular Conformation , Parasitic Sensitivity Tests , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Trypanocidal Agents/pharmacokinetics , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL