Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Antimicrob Agents Chemother ; 58(6): 3312-26, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24687493

ABSTRACT

Pantothenate kinase (PanK) catalyzes the phosphorylation of pantothenate, the first committed and rate-limiting step toward coenzyme A (CoA) biosynthesis. In our earlier reports, we had established that the type I isoform encoded by the coaA gene is an essential pantothenate kinase in Mycobacterium tuberculosis, and this vital information was then exploited to screen large libraries for identification of mechanistically different classes of PanK inhibitors. The present report summarizes the synthesis and expansion efforts to understand the structure-activity relationships leading to the optimization of enzyme inhibition along with antimycobacterial activity. Additionally, we report the progression of two distinct classes of inhibitors, the triazoles, which are ATP competitors, and the biaryl acetic acids, with a mixed mode of inhibition. Cocrystallization studies provided evidence of these inhibitors binding to the enzyme. This was further substantiated with the biaryl acids having MIC against the wild-type M. tuberculosis strain and the subsequent establishment of a target link with an upshift in MIC in a strain overexpressing PanK. On the other hand, the ATP competitors had cellular activity only in a M. tuberculosis knockdown strain with reduced PanK expression levels. Additionally, in vitro and in vivo survival kinetic studies performed with a M. tuberculosis PanK (MtPanK) knockdown strain indicated that the target levels have to be significantly reduced to bring in growth inhibition. The dual approaches employed here thus established the poor vulnerability of PanK in M. tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Blotting, Western , Gene Knockdown Techniques , Humans , Microbial Sensitivity Tests , Mycobacterium bovis/genetics , Mycobacterium tuberculosis/genetics , Phenotype , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Conformation , Quinolones/pharmacology , Structure-Activity Relationship , Triazoles/pharmacology
2.
Bioorg Med Chem Lett ; 24(3): 870-9, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24405701

ABSTRACT

Scaffold hopping from the thiazolopyridine ureas led to thiazolopyridone ureas with potent antitubercular activity acting through inhibition of DNA GyrB ATPase activity. Structural diversity was introduced, by extension of substituents from the thiazolopyridone N-4 position, to access hydrophobic interactions in the ribose pocket of the ATP binding region of GyrB. Further optimization of hydrogen bond interactions with arginines in site-2 of GyrB active site pocket led to potent inhibition of the enzyme (IC50 2 nM) along with potent cellular activity (MIC=0.1 µM) against Mycobacterium tuberculosis (Mtb). Efficacy was demonstrated in an acute mouse model of tuberculosis on oral administration.


Subject(s)
Mycobacterium tuberculosis/drug effects , Pyridones/chemical synthesis , Thiazoles/chemical synthesis , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/pharmacology , Urea/chemical synthesis , Urea/pharmacology , Administration, Oral , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Disease Models, Animal , Inhibitory Concentration 50 , Mice , Microbial Sensitivity Tests , Molecular Structure , Pyridones/chemistry , Pyridones/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology , Topoisomerase II Inhibitors/chemistry , Urea/chemistry
3.
J Biol Chem ; 288(25): 18260-70, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23661699

ABSTRACT

Mycobacterium tuberculosis, the bacterial causative agent of tuberculosis, currently affects millions of people. The emergence of drug-resistant strains makes development of new antibiotics targeting the bacterium a global health priority. Pantothenate kinase, a key enzyme in the universal biosynthesis of the essential cofactor CoA, was targeted in this study to find new tuberculosis drugs. The biochemical characterizations of two new classes of compounds that inhibit pantothenate kinase from M. tuberculosis are described, along with crystal structures of their enzyme-inhibitor complexes. These represent the first crystal structures of this enzyme with engineered inhibitors. Both classes of compounds bind in the active site of the enzyme, overlapping with the binding sites of the natural substrate and product, pantothenate and phosphopantothenate, respectively. One class of compounds also interferes with binding of the cofactor ATP. The complexes were crystallized in two crystal forms, one of which is in a new space group for this enzyme and diffracts to the highest resolution reported for any pantothenate kinase structure. These two crystal forms allowed, for the first time, modeling of the cofactor-binding loop in both open and closed conformations. The structures also show a binding mode of ATP different from that previously reported for the M. tuberculosis enzyme but similar to that in the pantothenate kinases of other organisms.


Subject(s)
Bacterial Proteins/chemistry , Enzyme Inhibitors/chemistry , Mycobacterium tuberculosis/enzymology , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biocatalysis/drug effects , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Pantothenic Acid/analogs & derivatives , Pantothenic Acid/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Substrate Specificity
4.
J Biomol Screen ; 17(3): 293-302, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22086722

ABSTRACT

The authors describe the discovery of anti-mycobacterial compounds through identifying mechanistically diverse inhibitors of the essential Mycobacterium tuberculosis (Mtb) enzyme, pantothenate kinase (CoaA). Target-driven drug discovery technologies often work with purified enzymes, and inhibitors thus discovered may not optimally inhibit the form of the target enzyme predominant in the bacterial cell or may not be available at the desired concentration. Therefore, in addition to addressing entry or efflux issues, inhibitors with diverse mechanisms of inhibition (MoI) could be prioritized before hit-to-lead optimization. The authors describe a high-throughput assay based on protein thermal melting to screen large numbers of compounds for hits with diverse MoI. Following high-throughput screening for Mtb CoaA enzyme inhibitors, a concentration-dependent increase in protein thermal stability was used to identify true binders, and the degree of enhancement or reduction in thermal stability in the presence of substrate was used to classify inhibitors as competitive or non/uncompetitive. The thermal shift-based MoI assay could be adapted to screen hundreds of compounds in a single experiment as compared to traditional biochemical approaches for MoI determination. This MoI was confirmed through mechanistic studies that estimated K(ie) and K(ies) for representative compounds and through nuclear magnetic resonance-based ligand displacement assays.


Subject(s)
Enzyme Inhibitors/chemistry , High-Throughput Screening Assays/methods , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Biological Assay , Drug Design , Drug Evaluation, Preclinical , Enzyme Inhibitors/analysis , Enzyme Inhibitors/metabolism
5.
J Med Chem ; 50(5): 962-78, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17284021

ABSTRACT

Highly rigid and geometrically well-defined rods composed of ethynylene-substituted aromatic spacers [oligo(p-phenyleneethynylene), OPE] were incorporated as acyl moieties on diacylglycerol lactones (DAG-lactones) and investigated for their ability to bind to protein kinase C (PKC) and translocate PKC alpha and delta isoforms to plasma and internal membranes. The kinetics of PKC translocation were correlated with biological responses, viz. ERK phosphorylation, induction of IL-6 secretion, inhibition of cell proliferation, and induction of cellular attachment, that display very different time courses. Because OPE rods assemble through noncovalent forces and form stable films, they may influence the microdomain environment around the DAG-lactone membrane-binding site. A comparison of two DAG-lactones (1 and 10), one with two PE units (1) and the other with an equivalent flexible acyl chain (10) of matching lipophilicity, clearly demonstrated the effect of the rigid OPE chain in substantially prolonging the translocated state of both PKC alpha and delta.


Subject(s)
Cell Membrane/metabolism , Diglycerides/chemical synthesis , Lactones/chemical synthesis , Protein Kinase C-alpha/metabolism , Protein Kinase C/metabolism , Animals , Binding Sites , Cell Adhesion , Cell Line , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Diglycerides/chemistry , Diglycerides/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-6/metabolism , Isoenzymes/metabolism , Kinetics , Lactones/chemistry , Lactones/pharmacology , Ligands , Molecular Conformation , Phosphorylation , Protein Binding , Protein Kinase C beta , Protein Transport , Structure-Activity Relationship
6.
Org Lett ; 8(7): 1335-7, 2006 Mar 30.
Article in English | MEDLINE | ID: mdl-16562885

ABSTRACT

[reaction: see text] Commercial 1,2:5,6-di-O-isopropylidene-alpha-d-allofuranose was converted to a protected bicyclic octosyl acid thioglycoside donor by a 10-step sequence that features an intramolecular ester enolate alkylation. Glycosylation of N-benzoyladenine and methyl uridine-5-carboxylate followed by deprotection gave the respective nucleosides "octosyl adenine" and octosyl acid A.


Subject(s)
Nucleosides/chemical synthesis , Adenine/chemistry , Glycosylation , Molecular Structure
7.
Bioorg Med Chem ; 13(8): 2717-22, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15781383

ABSTRACT

Flavone-8-acetic acid (FAA) is a potent immunomodulatory small molecule that is uniquely characterized as being active on mouse but not human cells. Although FAA is a potent inducer of murine cytokine, chemokine and interferon gene expression, its mode of action remains unknown. In this report, we describe the synthesis of a new flavone acetic acid (FAA) analogue, (2-[2-(4-azidophenyl)-4-oxochromen-8-yl-]acetic acid (compound 2). We demonstrate that compound 2 is equally active as the parent FAA in inducing chemokine gene expression and that the azide functional group is capable of reacting with a reporter molecule, such as the FLAG peptide-phosphine, under mild conditions. This reaction will be useful for detecting the drug-bound protein active complex utilizing an anti-FLAG antibody.


Subject(s)
Azides/chemistry , Flavonoids/chemical synthesis , Flavonoids/pharmacology , Animals , Binding Sites/drug effects , Cell Line , Chemokines/genetics , Drug Design , Flavonoids/chemistry , Gene Expression/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Molecular Structure , Oligopeptides , Peptides/chemistry , Phosphines/chemistry , RNA/drug effects , RNA/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...