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










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.
J Med Chem ; 56(15): 6190-9, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23819803

ABSTRACT

The antimalarial compound fosmidomycin targets DXR, the enzyme that catalyzes the first committed step in the MEP pathway, producing the essential isoprenoid precursors, isopentenyl diphosphate and dimethylallyl diphosphate. The MEP pathway is used by a number of pathogens, including Mycobacterium tuberculosis and apicomplexan parasites, and differs from the classical mevalonate pathway that is essential in humans. Using a structure-based approach, we designed a number of analogues of fosmidomycin, including a series that are substituted in both the Cα and the hydroxamate positions. The latter proved to be a stable framework for the design of inhibitors that extend from the polar and cramped (and so not easily druggable) substrate-binding site and can, for the first time, bridge the substrate and cofactor binding sites. A number of these compounds are more potent than fosmidomycin in terms of killing Plasmodium falciparum in an in vitro assay; the best has an IC50 of 40 nM.


Subject(s)
Aldose-Ketose Isomerases/antagonists & inhibitors , Antimalarials/chemical synthesis , Fosfomycin/analogs & derivatives , Aldose-Ketose Isomerases/chemistry , Antimalarials/chemistry , Antimalarials/pharmacology , Crystallography, X-Ray , Escherichia coli/enzymology , Fosfomycin/chemical synthesis , Fosfomycin/chemistry , Fosfomycin/pharmacology , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Inhibitory Concentration 50 , Models, Molecular , Mycobacterium tuberculosis/enzymology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Protein Binding , Protein Conformation , Structure-Activity Relationship
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.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 2): 134-43, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22281742

ABSTRACT

A number of pathogens, including the causative agents of tuberculosis and malaria, synthesize the essential isoprenoid precursor isopentenyl diphosphate via the 2-C-methyl-D-erythritol 4-phosphate (MEP) pathway rather than the classical mevalonate pathway that is found in humans. As part of a structure-based drug-discovery program against tuberculosis, DXR, the enzyme that carries out the second step in the MEP pathway, has been investigated. This enzyme is the target for the antibiotic fosmidomycin and its active acetyl derivative FR-900098. The structure of DXR from Mycobacterium tuberculosis in complex with FR-900098, manganese and the NADPH cofactor has been solved and refined. This is a new crystal form that diffracts to a higher resolution than any other DXR complex reported to date. Comparisons with other ternary complexes show that the conformation is that of the enzyme in an active state: the active-site flap is well defined and the cofactor-binding domain has a conformation that brings the NADPH into the active site in a manner suitable for catalysis. The substrate-binding site is highly conserved in a number of pathogens that use this pathway, so any new inhibitor that is designed for the M. tuberculosis enzyme is likely to exhibit broad-spectrum activity.


Subject(s)
Aldose-Ketose Isomerases/chemistry , Aldose-Ketose Isomerases/metabolism , Anti-Bacterial Agents/pharmacology , Fosfomycin/analogs & derivatives , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Mycobacterium tuberculosis/enzymology , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Erythritol/analogs & derivatives , Erythritol/metabolism , Fosfomycin/chemistry , Fosfomycin/pharmacology , Manganese/chemistry , Manganese/metabolism , Models, Molecular , Molecular Sequence Data , Mycobacterium tuberculosis/chemistry , NADP/chemistry , NADP/metabolism , Protein Binding , Sequence Alignment , Sugar Phosphates/metabolism
5.
J Org Chem ; 76(21): 8986-98, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21936546

ABSTRACT

Cinnamaldehyde derivatives were synthesized in good to excellent yields in one step by a mild and selective, base-free palladium(II)-catalyzed oxidative Heck reaction starting from acrolein and various arylboronic acids. Prepared α,ß-unsaturated aldehydes were used for synthesis of novel α-aryl substituted fosmidomycin analogues, which were evaluated for their inhibition of Mycobacterium tuberculosis 1-deoxy-D-xylulose 5-phosphate reductoisomerase. IC(50) values between 0.8 and 27.3 µM were measured. The best compound showed activity comparable to that of the most potent previously reported α-aryl substituted fosmidomycin-class inhibitor.


Subject(s)
Acrolein/analogs & derivatives , Aldose-Ketose Isomerases/antagonists & inhibitors , Aldose-Ketose Isomerases/chemical synthesis , Antitubercular Agents/chemistry , Antitubercular Agents/chemical synthesis , Fosfomycin/analogs & derivatives , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/chemical synthesis , Mycobacterium tuberculosis/chemistry , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/chemical synthesis , Acrolein/chemical synthesis , Acrolein/chemistry , Antitubercular Agents/pharmacology , Catalysis , Fosfomycin/chemical synthesis , Fosfomycin/chemistry , Fosfomycin/pharmacology , Humans , Inhibitory Concentration 50 , Models, Molecular , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Oxidation-Reduction , Palladium/chemistry , Protein Binding
6.
Bioorg Med Chem Lett ; 21(18): 5403-7, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21824775
7.
J Med Chem ; 54(14): 4964-76, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21678907

ABSTRACT

The natural antibiotic fosmidomycin acts via inhibition of 1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR), an essential enzyme in the non-mevalonate pathway of isoprenoid biosynthesis. Fosmidomycin is active on Mycobacterium tuberculosis DXR (MtDXR), but it lacks antibacterial activity probably because of poor uptake. α-Aryl substituted fosmidomycin analogues have more favorable physicochemical properties and are also more active in inhibiting malaria parasite growth. We have solved crystal structures of MtDXR in complex with 3,4-dichlorophenyl substituted fosmidomycin analogues; these show important differences compared to our previously described forsmidomycin-DXR complex. Our best inhibitor has an IC(50) = 0.15 µM on MtDXR but still lacked activity in a mycobacterial growth assay (MIC > 32 µg/mL). The combined results, however, provide insights into how DXR accommodates the new inhibitors and serve as an excellent starting point for the design of other novel and more potent inhibitors, particularly against pathogens where uptake is less of a problem, such as the malaria parasite.


Subject(s)
Aldose-Ketose Isomerases/antagonists & inhibitors , Antitubercular Agents/chemical synthesis , Fosfomycin/analogs & derivatives , Multienzyme Complexes/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Oxidoreductases/antagonists & inhibitors , Aldose-Ketose Isomerases/chemistry , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Catalytic Domain , Crystallography, X-Ray , Drug Design , Fosfomycin/chemical synthesis , Fosfomycin/chemistry , Fosfomycin/pharmacology , Models, Molecular , Multienzyme Complexes/chemistry , Mycobacterium tuberculosis/drug effects , Oxidoreductases/chemistry , Protein Binding , Protein Conformation , Structure-Activity Relationship
8.
Acta Crystallogr D Biol Crystallogr ; 67(Pt 5): 403-14, 2011 May.
Article in English | MEDLINE | ID: mdl-21543842

ABSTRACT

A number of pathogens, including the causative agents of tuberculosis and malaria, synthesize isopentenyl diphosphate via the 2-C-methyl-D-erythritol 4-phosphate (MEP) pathway rather than the classical mevalonate pathway found in humans. As part of a structure-based drug-discovery program against tuberculosis, IspD, the enzyme that carries out the third step in the MEP pathway, was targeted. Constructs of both the Mycobacterium smegmatis and the Mycobacterium tuberculosis enzymes that were suitable for structural and inhibitor-screening studies were engineered. Two crystal structures of the M. smegmatis enzyme were produced, one in complex with CTP and the other in complex with CMP. In addition, the M. tuberculosis enzyme was crystallized in complex with CTP. Here, the structure determination and crystallographic refinement of these crystal forms and the enzymatic characterization of the M. tuberculosis enzyme construct are reported. A comparison with known IspD structures allowed the definition of the structurally conserved core of the enzyme. It indicates potential flexibility in the enzyme and in particular in areas close to the active site. These well behaved constructs provide tools for future target-based screening of potential inhibitors. The conserved nature of the extended active site suggests that any new inhibitor will potentially exhibit broad-spectrum activity.


Subject(s)
Bacterial Proteins/chemistry , Erythritol/analogs & derivatives , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , Sugar Phosphates/metabolism , Tuberculosis/enzymology , Amino Acid Sequence , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Drug Design , Enzyme Inhibitors/pharmacology , Erythritol/metabolism , Hemiterpenes/metabolism , Humans , Molecular Sequence Data , Mycobacterium smegmatis/chemistry , Mycobacterium tuberculosis/chemistry , Organophosphorus Compounds/metabolism , Sequence Alignment , Tuberculosis/drug therapy
9.
Acta Crystallogr D Biol Crystallogr ; 62(Pt 7): 807-13, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16790937

ABSTRACT

1-deoxy-D-xylulose 5-phosphate reductoisomerase catalyzes the NADPH-dependent rearrangement and reduction of 1-deoxy-D-xylulose 5-phosphate to form 2-C-methyl-D-erythritol 4-phosphate, as the second step of the deoxyxylulose 5-phosphate/methylerythritol 4-phosphate pathway found in many bacteria and plants. The end product, isopentenyl diphosphate, is the precursor of various isoprenoids vital to all living organisms. The pathway is not found in humans; the mevalonate pathway is instead used for the formation of isopentenyl diphosphate. This difference, combined with its essentiality, makes the reductoisomerase an excellent drug target in a number of pathogenic organisms. The structure of 1-deoxy-D-xylulose 5-phosphate reductoisomerase from Mycobacterium tuberculosis (Rv2870c) was solved by molecular replacement and refined to a resolution of 1.9 A. The enzyme exhibited an estimated kcat of 5.3 s-1 and Km and kcat/Km values of 7.2 microM and 7.4x10(5) M-1 s-1 for NADPH and 340 microM and 1.6x10(4) M-1 s-1 for 1-deoxy-D-xylulose 5-phosphate. In the structure, a sulfate is bound at the expected site of the phosphate moiety of the sugar substrate. The M. tuberculosis enzyme displays a similar fold to the previously published structures from Escherichia coli and Zymomonas mobilis. Comparisons offer suggestions for the design of specific drugs. Furthermore, the new structure represents an intermediate conformation between the open apo form and the closed holo form observed previously, giving insights into the conformational changes associated with catalysis.


Subject(s)
Aldose-Ketose Isomerases/chemistry , Crystallography, X-Ray/methods , Multienzyme Complexes/chemistry , Mycobacterium tuberculosis/enzymology , Oxidoreductases/chemistry , Aldose-Ketose Isomerases/genetics , Aldose-Ketose Isomerases/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Escherichia coli/enzymology , Escherichia coli/metabolism , Models, Molecular , Molecular Sequence Data , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mycobacterium tuberculosis/genetics , Oxidoreductases/genetics , Oxidoreductases/metabolism , Pentosephosphates/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Sulfates/chemistry , Sulfates/metabolism
10.
J Biol Chem ; 280(19): 18782-9, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15753099

ABSTRACT

Carbonic anhydrases catalyze the reversible hydration of carbon dioxide to form bicarbonate. This activity is universally required for fatty acid biosynthesis as well as for the production of a number of small molecules, pH homeostasis, and other functions. At least three different carbonic anhydrase families are known to exist, of which the alpha-class found in humans has been studied in most detail. In the present work, we describe the structures of two of the three beta-class carbonic anhydrases that have been identified in Mycobacterium tuberculosis, i.e. Rv1284 and Rv3588c. Both structures were solved by molecular replacement and then refined to resolutions of 2.0 and 1.75 A, respectively. The active site of Rv1284 is small and almost completely shielded from solvent, whereas that of Rv3588c is larger and quite open to solution. Differences in coordination of the active site metal are also observed. In Rv3588c, an aspartic acid side chain displaces a water molecule and coordinates directly to the zinc ion, thereby closing the zinc coordination sphere and breaking the salt link to a nearby arginine that is a feature of Rv1284. The two carbonic anhydrases thus exhibit both of the metal coordination geometries that have previously been observed for structures in this family. Activity studies demonstrate that Rv3588c is a completely functional carbonic anhydrase. The apparent lack of activity of Rv1284 in the present assay system is likely exacerbated by the observed depletion of zinc in the preparation.


Subject(s)
Carbonic Anhydrases/chemistry , Carbonic Anhydrases/physiology , Mycobacterium tuberculosis/enzymology , Amino Acid Sequence , Aspartic Acid/chemistry , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Dimerization , Hydrogen-Ion Concentration , Ions , Ligands , Metals/chemistry , Models, Molecular , Molecular Sequence Data , Multigene Family , Protein Conformation , Protein Structure, Tertiary , Salts/pharmacology , Serine/chemistry , Solvents/chemistry , Structure-Activity Relationship , Time Factors , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...