Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Chembiochem ; 20(13): 1701-1711, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30856684

RESUMEN

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.


Asunto(s)
Liasas de Carbono-Oxígeno/química , Liasas de Carbono-Oxígeno/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Liasas de Carbono-Oxígeno/genética , Catálisis , Dominio Catalítico/genética , Pruebas de Enzimas/métodos , Hidroxiácidos/metabolismo , Cinética , Magnesio/metabolismo , Modelos Químicos , Mutagénesis Sitio-Dirigida , Filogenia , Unión Proteica , Alineación de Secuencia , Especificidad por Sustrato , Xanthomonas campestris/enzimología
2.
Antimicrob Agents Chemother ; 60(11): 6451-6459, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27527085

RESUMEN

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.


Asunto(s)
Oxidorreductasas de Alcohol/antagonistas & inhibidores , Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Farmacorresistencia Bacteriana Múltiple/genética , Mutación , Mycobacterium tuberculosis/efectos de los fármacos , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Sustitución de Aminoácidos , Antituberculosos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Cisteína/química , Cisteína/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Expresión Génica , Humanos , Macrófagos/microbiología , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/crecimiento & desarrollo , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crecimiento & desarrollo , Fenotipo , Piperazinas/química , Piperazinas/farmacología , Relación Estructura-Actividad , Tiazinas/química , Tiazinas/farmacología , Tuberculosis Resistente a Múltiples Medicamentos/microbiología
3.
Nat Chem Biol ; 10(2): 96-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24292073

RESUMEN

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.


Asunto(s)
Antituberculosos/química , Proteínas Bacterianas/química , NAD/química , Oligopéptidos/química , Oxidorreductasas/química , Sitios de Unión , Cristalografía por Rayos X , Modelos Moleculares , Especificidad por Sustrato
4.
Antimicrob Agents Chemother ; 59(8): 4446-52, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25987616

RESUMEN

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.


Asunto(s)
Oxidorreductasas de Alcohol/antagonistas & inhibidores , Proteínas Bacterianas/antagonistas & inhibidores , Mycobacterium tuberculosis/efectos de los fármacos , Piperazinas/farmacología , Piridinas/farmacología , Pirroles/farmacología , Compuestos de Espiro/farmacología , Tiazinas/farmacología , Animales , Antituberculosos/farmacología , Dominio Catalítico/efectos de los fármacos , Modelos Animales de Enfermedad , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana/métodos , Mycobacterium tuberculosis/metabolismo , Relación Estructura-Actividad , Tuberculosis/tratamiento farmacológico , Tuberculosis/metabolismo
5.
Bioorg Med Chem ; 23(24): 7694-710, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26643218

RESUMEN

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.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Antituberculosos/química , Antituberculosos/farmacología , Proteínas Bacterianas/metabolismo , Benzotiazoles/química , Benzotiazoles/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Oxidorreductasas de Alcohol/antagonistas & inhibidores , Proteínas Bacterianas/antagonistas & inhibidores , Diseño de Fármacos , Humanos , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología
6.
Mol Neurobiol ; 61(4): 2367-2389, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37874479

RESUMEN

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.


Asunto(s)
Epilepsia del Lóbulo Temporal , Animales , Humanos , Ratones , Regulación hacia Abajo , Gliosis , Hipocampo/metabolismo , Ácido Kaínico , Ratones Transgénicos , Proteasas Ubiquitina-Específicas/metabolismo
7.
Biochemistry ; 52(21): 3740-51, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-23672572

RESUMEN

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.


Asunto(s)
Glicoproteínas/metabolismo , Simulación de Dinámica Molecular , Neuraminidasa/metabolismo , Trypanosoma cruzi/enzimología , Animales , Conformación Proteica , Especificidad por Sustrato
8.
Mol Microbiol ; 77(5): 1172-85, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20624223

RESUMEN

Tuberculosis is still a leading cause of death in developing countries, for which there is an urgent need for new pharmacological agents. The synthesis of the novel antimycobacterial drug class of benzothiazinones (BTZs) and the identification of their cellular target as DprE1 (Rv3790), a component of the decaprenylphosphoryl-ß-d-ribose 2'-epimerase complex, have been reported recently. Here, we describe the identification and characterization of a novel resistance mechanism to BTZ in Mycobacterium smegmatis. The overexpression of the nitroreductase NfnB leads to the inactivation of the drug by reduction of a critical nitro-group to an amino-group. The direct involvement of NfnB in the inactivation of the lead compound BTZ043 was demonstrated by enzymology, microbiological assays and gene knockout experiments. We also report the crystal structure of NfnB in complex with the essential cofactor flavin mononucleotide, and show that a common amino acid stretch between NfnB and DprE1 is likely to be essential for the interaction with BTZ. We performed docking analysis of NfnB-BTZ in order to understand their interaction and the mechanism of nitroreduction. Although Mycobacterium tuberculosis seems to lack nitroreductases able to inactivate these drugs, our findings are valuable for the design of new BTZ molecules, which may be more effective in vivo.


Asunto(s)
Antituberculosos/farmacología , Farmacorresistencia Bacteriana , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium smegmatis/enzimología , Nitrorreductasas/química , Nitrorreductasas/metabolismo , Tiazinas/farmacología , Antituberculosos/metabolismo , Cristalografía por Rayos X , Técnicas de Inactivación de Genes , Pruebas de Sensibilidad Microbiana , Nitrorreductasas/genética , Oxidación-Reducción , Estructura Terciaria de Proteína , Tiazinas/metabolismo
9.
Biophys J ; 98(9): L38-40, 2010 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-20441732

RESUMEN

Molecular dynamics investigations into active site plasticity of Trypanosoma cruzi trans-sialidase, a protein implicated in Chagas disease, suggest that movement of the Trp(312) loop plays an important role in the enzyme's sialic acid transfer mechanism. The observed Trp(312) flexibility equates to a molecular shovel action, which leads to the expulsion of the donor aglycone leaving group from the catalytic site. These computational simulations provide detailed structural insights into sialyl transfer by the trans-sialidase and may aid the design of inhibitors effective against this neglected tropical disease.


Asunto(s)
Glicoproteínas/química , Glicoproteínas/metabolismo , Neuraminidasa/química , Neuraminidasa/metabolismo , Trypanosoma cruzi/enzimología , Triptófano/metabolismo , Biocatálisis , Dominio Catalítico , Glicosilación , Simulación de Dinámica Molecular , Ácido N-Acetilneuramínico/metabolismo
10.
Biochemistry ; 49(43): 9292-305, 2010 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-20853905

RESUMEN

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.


Asunto(s)
Acinetobacter baumannii/enzimología , Inhibidores Enzimáticos/química , Imidazoles/metabolismo , Oxazoles/metabolismo , Péptido Sintasas/antagonistas & inhibidores , Proteínas Bacterianas , Humanos , Cinética , Redes y Vías Metabólicas , Unión Proteica
11.
Biochemistry ; 49(8): 1689-701, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20099905

RESUMEN

The dual-specificity protein kinase monopolar spindle 1 (Mps1) is a central component of the mitotic spindle assembly checkpoint (SAC), a sensing mechanism that prevents anaphase until all chromosomes are bioriented on the metaphase plate. Partial depletion of Mps1 protein levels sensitizes transformed, but not untransformed, human cells to therapeutic doses of the anticancer agent Taxol, making it an attractive novel therapeutic cancer target. We have previously determined the X-ray structure of the catalytic domain of human Mps1 in complex with the anthrapyrazolone kinase inhibitor SP600125. In order to validate distinct inhibitors that target this enzyme and improve our understanding of nucleotide binding site architecture, we now report a biophysical and structural evaluation of the Mps1 catalytic domain in the presence of ATP and the aspecific model kinase inhibitor staurosporine. Collective in silico, enzymatic, and fluorescent screens also identified several new lead quinazoline Mps1 inhibitors, including a low-affinity compound termed Compound 4 (Cpd 4), whose interaction with the Mps1 kinase domain was further characterized by X-ray crystallography. A novel biophysical analysis demonstrated that the intrinsic fluorescence of SP600125 changed markedly upon Mps1 binding, allowing spectrophotometric displacement analysis and determination of dissociation constants for ATP-competitive Mps1 inhibitors. By illuminating the structure of the Mps1 ATP-binding site our results provide novel biophysical insights into Mps1-ligand interactions that will be useful for the development of specific Mps1 inhibitors, including those employing a therapeutically validated quinazoline template.


Asunto(s)
Antracenos/química , Antracenos/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/química , Cristalografía por Rayos X/métodos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Dominio Catalítico , Proteínas de Ciclo Celular/metabolismo , Humanos , Estructura Molecular , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Secundaria de Proteína , Proteínas Tirosina Quinasas , Espectrometría de Fluorescencia , Espectrofotometría Ultravioleta
12.
Bioorg Med Chem Lett ; 19(3): 589-96, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19144516

RESUMEN

trans-Sialidase from Trypanosoma cruzi (TcTS) has emerged as a potential drug target for treatment of Chagas disease. Here, we report the results of virtual screening for the discovery of novel TcTS inhibitors, which targeted both the sialic acid and sialic acid acceptor sites of this enzyme. A library prepared from the Evotec database of commercially available compounds was screened using the molecular docking program GOLD, following the application of drug-likeness filters. Twenty-three compounds selected from the top-scoring ligands were purchased and assayed using a fluorimetric assay. Novel inhibitor scaffolds, with IC(50) values in the submillimolar range were discovered. The 3-benzothiazol-2-yl-4-phenyl-but-3-enoic acid scaffold was studied in more detail, and TcTS inhibition was confirmed by an alternative sialic acid transfer assay. Attempts to obtain crystal structures of these compounds with TcTS proved unsuccessful but provided evidence of ligand binding at the active site.


Asunto(s)
Química Farmacéutica/métodos , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Glicoproteínas/antagonistas & inhibidores , Neuraminidasa/antagonistas & inhibidores , Animales , Sitios de Unión , Dominio Catalítico , Química Farmacéutica/instrumentación , Cristalización , Cristalografía por Rayos X/métodos , Inhibidores Enzimáticos/química , Glicoproteínas/química , Concentración 50 Inhibidora , Cinética , Ligandos , Modelos Químicos , Ácido N-Acetilneuramínico/química , Neuraminidasa/química , Trypanosoma cruzi
13.
Biochemistry ; 47(45): 11735-49, 2008 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-18928302

RESUMEN

The design and synthesis of a fluorescent probe Fl-Sal-AMS 6 based on the tight-binding inhibitor 5'- O-[ N-(salicyl)sulfamoyl]adenosine (Sal-AMS) is described for the aryl acid adenylating enzymes (AAAEs) known as MbtA, YbtE, EntE, VibE, DhbE, and BasE involved in siderophore biosynthesis from Mycobacterium tuberculosis, Yersinia pestis, Escherichia coli, Vibrio cholerae, Bacillus subtilis, and Acinetobacter baumannii, respectively. The probe was successfully used to develop a fluorescence polarization assay for these six AAAEs, and equilibrium dissociation constants were determined in direct binding experiments. Fl-Sal-AMS was effective for AAAEs that utilize salicylic acid or 2,3-dihydroxybenzoic acid as native substrates, with dissociation constants ranging from 9-369 nM, but was ineffective for AsbC, the AAAE from Bacillus anthracis, which activates 3,4-dihydroxybenzoic acid. Competitive binding experiments using a series of ligands including substrates, reaction products, and inhibitors provided the first comparative structure-activity relationships for AAAEs. The fluorescence polarization assay was then 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) against BasE, an AAAE from Acinetobacter baumannii involved in production of the siderophore acinetobactin. Several small molecule inhibitors with new chemotypes were identified, and compound 23 containing a pyrazolo[5,4- a]pyridine scaffold emerged as the most promising ligand with a K D of 78 nM, which was independently confirmed by isothermal calorimetry, and inhibition was also verified in an ATP-[ (32)P]-pyrophosphate exchange steady-state kinetic assay.


Asunto(s)
Proteínas Bacterianas/metabolismo , Polarización de Fluorescencia/métodos , Ligasas/metabolismo , Sideróforos/biosíntesis , Adenosina/análogos & derivados , Adenosina/química , Coenzima A Ligasas/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Proteínas de Escherichia coli/metabolismo , Hidroxibenzoatos/metabolismo , Cinética , Ligandos , Estructura Molecular , Ácido Salicílico/metabolismo , Sideróforos/química , Relación Estructura-Actividad , Especificidad por Sustrato
14.
Drug Discov Today ; 13(3-4): 110-7, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18275908

RESUMEN

Trypanosoma cruzi trans-sialidase is a potential target for Chagas disease chemotherapy. From the specific need of T. cruzi to obtain sialic acid through trans-sialidase-mediated transfers from host sources and the lack of alternative to this for the parasite, a good case can be made for T. cruzi trans-sialidase to serve as a potential drug target against Chagas disease. This review deals with both the particular aspects relevant to T. cruzi trans-sialidase as a target and generalises the situation for drug design in its broader aspects on the basis of some special problems in terms of rational drug design that T. cruzi trans-sialidase raises, particularly those of multiple gene copies and active site plasticity.


Asunto(s)
Diseño de Fármacos , Parasitología/métodos , Trypanosoma cruzi/efectos de los fármacos , Animales , Antiprotozoarios/química , Antiprotozoarios/farmacología , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/enzimología , Enfermedad de Chagas/patología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/metabolismo , Humanos , Estructura Molecular , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/metabolismo , Trypanosoma cruzi/enzimología
15.
ACS Chem Biol ; 13(11): 3184-3192, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30289689

RESUMEN

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.


Asunto(s)
Marcadores de Afinidad/farmacología , Oxidorreductasas de Alcohol/antagonistas & inhibidores , Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Tiazinas/farmacología , Actinomycetales/efectos de los fármacos , Actinomycetales/enzimología , Marcadores de Afinidad/síntesis química , Oxidorreductasas de Alcohol/genética , Antituberculosos/síntesis química , Proteínas Bacterianas/genética , Membrana Celular/metabolismo , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Fluoresceínas/síntesis química , Fluoresceínas/farmacología , Fluorescencia , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/farmacología , Células Hep G2 , Humanos , Pruebas de Sensibilidad Microbiana , Microscopía Fluorescente/métodos , Mutación , Tiazinas/síntesis química
16.
ChemMedChem ; 11(3): 331-9, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26751718

RESUMEN

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.


Asunto(s)
Antituberculosos/farmacología , Benzotiazoles/farmacología , Mutágenos/química , Mycobacterium tuberculosis/efectos de los fármacos , Nitrocompuestos/farmacología , Antituberculosos/efectos adversos , Antituberculosos/química , Benzotiazoles/efectos adversos , Benzotiazoles/química , Relación Dosis-Respuesta a Droga , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Nitrocompuestos/efectos adversos , Nitrocompuestos/química , Estereoisomerismo , Relación Estructura-Actividad
17.
J Med Chem ; 48(15): 4861-70, 2005 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-16033266

RESUMEN

Human leukocyte elastase (HLE) is a serine protease that very efficiently degrades various tissue matrix proteins such as elastin. The imbalance between HLE and its endogenous inhibitors leads to excessive elastin proteolysis and is considered to be responsible for the onset of chronic obstructive pulmonary disease (COPD). A novel series of C-3-, C-4-, and N-1-substituted azetidin-2-ones were prepared as potential mechanism-based inhibitors of HLE to restore the protease/antiprotease imbalance. N-Acyloxyalkylazetidin-2-ones, 4, and their carbamate counterparts, 5, are weak HLE inhibitors, being 5 times less active than their bicyclic oxazolidin-2,4-dione-substituted analogues, 6, containing an electron-withdrawing substituent at C-4. Compounds 6 containing a C-4 substituent exist as two diastereomeric pairs of enantiomers, each pair presenting similar inhibitory activity against HLE. Comparative docking experiments with the C-4-substituted oxazolidin-2,4-dione inhibitors 6 suggest that only the 4R,5'S and 4S,5'S diastereomers consistently interact with the beta-lactam carbonyl carbon atom accessible to the serine hydroxyl oxygen.


Asunto(s)
Lactamas/síntesis química , Elastasa de Leucocito/antagonistas & inhibidores , Oxazoles/síntesis química , Azetidinas/síntesis química , Azetidinas/química , Diseño de Fármacos , Humanos , Lactamas/química , Elastasa de Leucocito/química , Modelos Moleculares , Oxazoles/química , Estereoisomerismo , Relación Estructura-Actividad
18.
ACS Chem Biol ; 10(7): 1631-6, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25906160

RESUMEN

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.


Asunto(s)
Oxidorreductasas de Alcohol/análisis , Oxidorreductasas de Alcohol/metabolismo , Antituberculosos/farmacología , Proteínas Bacterianas/análisis , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Mycobacterium tuberculosis/citología , Mycobacterium tuberculosis/enzimología , Tuberculosis/microbiología , Pared Celular/efectos de los fármacos , Humanos , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis/tratamiento farmacológico
19.
ACS Chem Biol ; 10(3): 705-14, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25427196

RESUMEN

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.


Asunto(s)
Oxidorreductasas de Alcohol/antagonistas & inhibidores , Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Quinoxalinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Antituberculosos/síntesis química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Pared Celular/efectos de los fármacos , Pared Celular/enzimología , Cristalografía por Rayos X , Descubrimiento de Drogas , Inhibidores Enzimáticos/síntesis química , Expresión Génica , Enlace de Hidrógeno , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Mutación , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/crecimiento & desarrollo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Quinoxalinas/síntesis química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA