Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 567
Filtrar
Más filtros

Intervalo de año de publicación
1.
EMBO J ; 42(15): e113687, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37377118

RESUMEN

Mycobacteria, such as Mycobacterium tuberculosis, depend on the activity of adenosine triphosphate (ATP) synthase for growth. The diarylquinoline bedaquiline (BDQ), a mycobacterial ATP synthase inhibitor, is an important medication for treatment of drug-resistant tuberculosis but suffers from off-target effects and is susceptible to resistance mutations. Consequently, both new and improved mycobacterial ATP synthase inhibitors are needed. We used electron cryomicroscopy and biochemical assays to study the interaction of Mycobacterium smegmatis ATP synthase with the second generation diarylquinoline TBAJ-876 and the squaramide inhibitor SQ31f. The aryl groups of TBAJ-876 improve binding compared with BDQ, while SQ31f, which blocks ATP synthesis ~10 times more potently than ATP hydrolysis, binds a previously unknown site in the enzyme's proton-conducting channel. Remarkably, BDQ, TBAJ-876, and SQ31f all induce similar conformational changes in ATP synthase, suggesting that the resulting conformation is particularly suited for drug binding. Further, high concentrations of the diarylquinolines uncouple the transmembrane proton motive force while for SQ31f they do not, which may explain why high concentrations of diarylquinolines, but not SQ31f, have been reported to kill mycobacteria.


Asunto(s)
Diarilquinolinas , Mycobacterium tuberculosis , Diarilquinolinas/farmacología , Antituberculosos/farmacología , Antituberculosos/química , Antituberculosos/metabolismo , Adenosina Trifosfato/metabolismo , Mycobacterium tuberculosis/genética
2.
Nature ; 589(7840): 143-147, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33299175

RESUMEN

Tuberculosis-the world's leading cause of death by infectious disease-is increasingly resistant to current first-line antibiotics1. The bacterium Mycobacterium tuberculosis (which causes tuberculosis) can survive low-energy conditions, allowing infections to remain dormant and decreasing their susceptibility to many antibiotics2. Bedaquiline was developed in 2005 from a lead compound identified in a phenotypic screen against Mycobacterium smegmatis3. This drug can sterilize even latent M. tuberculosis infections4 and has become a cornerstone of treatment for multidrug-resistant and extensively drug-resistant tuberculosis1,5,6. Bedaquiline targets the mycobacterial ATP synthase3, which is an essential enzyme in the obligate aerobic Mycobacterium genus3,7, but how it binds the intact enzyme is unknown. Here we determined cryo-electron microscopy structures of M. smegmatis ATP synthase alone and in complex with bedaquiline. The drug-free structure suggests that hook-like extensions from the α-subunits prevent the enzyme from running in reverse, inhibiting ATP hydrolysis and preserving energy in hypoxic conditions. Bedaquiline binding induces large conformational changes in the ATP synthase, creating tight binding pockets at the interface of subunits a and c that explain the potency of this drug as an antibiotic for tuberculosis.


Asunto(s)
Complejos de ATP Sintetasa/química , Antituberculosos/química , Microscopía por Crioelectrón , Diarilquinolinas/química , Mycobacterium smegmatis/enzimología , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología , Complejos de ATP Sintetasa/antagonistas & inhibidores , Complejos de ATP Sintetasa/metabolismo , Adenosina Trifosfato/metabolismo , Antituberculosos/metabolismo , Antituberculosos/farmacología , Diarilquinolinas/metabolismo , Diarilquinolinas/farmacología , Hidrólisis/efectos de los fármacos , Modelos Moleculares , Mycobacterium smegmatis/efectos de los fármacos , Rotación
3.
Proc Natl Acad Sci U S A ; 120(5): e2212755120, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36693100

RESUMEN

Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), a disease that claims ~1.6 million lives annually. The current treatment regime is long and expensive, and missed doses contribute to drug resistance. Therefore, development of new anti-TB drugs remains one of the highest public health priorities. Mtb has evolved a complex cell envelope that represents a formidable barrier to antibiotics. The Mtb cell envelop consists of four distinct layers enriched for Mtb specific lipids and glycans. Although the outer membrane, comprised of mycolic acid esters, has been extensively studied, less is known about the plasma membrane, which also plays a critical role in impacting antibiotic efficacy. The Mtb plasma membrane has a unique lipid composition, with mannosylated phosphatidylinositol lipids (phosphatidyl-myoinositol mannosides, PIMs) comprising more than 50% of the lipids. However, the role of PIMs in the structure and function of the membrane remains elusive. Here, we used multiscale molecular dynamics (MD) simulations to understand the structure-function relationship of the PIM lipid family and decipher how they self-organize to shape the biophysical properties of mycobacterial plasma membranes. We assess both symmetric and asymmetric assemblies of the Mtb plasma membrane and compare this with residue distributions of Mtb integral membrane protein structures. To further validate the model, we tested known anti-TB drugs and demonstrated that our models agree with experimental results. Thus, our work sheds new light on the organization of the mycobacterial plasma membrane. This paves the way for future studies on antibiotic development and understanding Mtb membrane protein function.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Fosfatidilinositoles/metabolismo , Mycobacterium tuberculosis/metabolismo , Membrana Celular/metabolismo , Tuberculosis/microbiología , Antituberculosos/metabolismo
4.
Mol Cell ; 66(2): 169-179.e8, 2017 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-28392175

RESUMEN

Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis, which kills 1.8 million annually. Mtb RNA polymerase (RNAP) is the target of the first-line antituberculosis drug rifampin (Rif). We report crystal structures of Mtb RNAP, alone and in complex with Rif, at 3.8-4.4 Å resolution. The results identify an Mtb-specific structural module of Mtb RNAP and establish that Rif functions by a steric-occlusion mechanism that prevents extension of RNA. We also report non-Rif-related compounds-Nα-aroyl-N-aryl-phenylalaninamides (AAPs)-that potently and selectively inhibit Mtb RNAP and Mtb growth, and we report crystal structures of Mtb RNAP in complex with AAPs. AAPs bind to a different site on Mtb RNAP than Rif, exhibit no cross-resistance with Rif, function additively when co-administered with Rif, and suppress resistance emergence when co-administered with Rif.


Asunto(s)
Proteínas Bacterianas/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Regulación Bacteriana de la Expresión Génica , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/genética , Transcripción Genética , Antituberculosos/metabolismo , Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Sitios de Unión , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , ARN Polimerasas Dirigidas por ADN/química , Farmacorresistencia Bacteriana , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Modelos Moleculares , Mycobacterium tuberculosis/efectos de los fármacos , Unión Proteica , Conformación Proteica , Rifampin/metabolismo , Rifampin/farmacología , Relación Estructura-Actividad , Transcripción Genética/efectos de los fármacos
5.
J Cell Mol Med ; 28(8): e18279, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38634203

RESUMEN

The rise of pyrazinamide (PZA)-resistant strains of Mycobacterium tuberculosis (MTB) poses a major challenge to conventional tuberculosis (TB) treatments. PZA, a cornerstone of TB therapy, must be activated by the mycobacterial enzyme pyrazinamidase (PZase) to convert its active form, pyrazinoic acid, which targets the ribosomal protein S1. Resistance, often associated with mutations in the RpsA protein, complicates treatment and highlights a critical gap in the understanding of structural dynamics and mechanisms of resistance, particularly in the context of the G97D mutation. This study utilizes a novel integration of computational techniques, including multiscale biomolecular and molecular dynamics simulations, physicochemical and medicinal chemistry predictions, quantum computations and virtual screening from the ZINC and Chembridge databases, to elucidate the resistance mechanism and identify lead compounds that have the potential to improve treatment outcomes for PZA-resistant MTB, namely ZINC15913786, ZINC20735155, Chem10269711, Chem10279789 and Chem10295790. These computational methods offer a cost-effective, rapid alternative to traditional drug trials by bypassing the need for organic subjects while providing highly accurate insight into the binding sites and efficacy of new drug candidates. The need for rapid and appropriate drug development emphasizes the need for robust computational analysis to justify further validation through in vitro and in vivo experiments.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis Resistente a Múltiples Medicamentos , Tuberculosis , Humanos , Pirazinamida/química , Pirazinamida/metabolismo , Pirazinamida/farmacología , Mycobacterium tuberculosis/genética , Antituberculosos/química , Antituberculosos/metabolismo , Antituberculosos/farmacología , Tuberculosis/microbiología , Mutación , Pruebas de Sensibilidad Microbiana
6.
Drug Metab Dispos ; 52(8): 858-874, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38769017

RESUMEN

Alpibectir (also known as BVL-GSK098 and GSK3729098) is a new chemical entity (NCE) with a novel mechanism for the treatment of tuberculosis. The disposition of alpibectir was determined in subjects from a first-time-in-human trial after a single oral dose of 40 mg and after 7 days repeat dosing at 30 mg. Here we present a combined approach of 19F-NMR (nuclear magnetic resonance), 1H-NMR, and high-resolution mass spectrometry (HRMS) to confidently determine the human metabolic fate of alpibectir. Utilizing multiple sites of fluorination in the molecule, it was possible to fractionate human urine and plasma to confidently detect and quantify the metabolite responses using 19F-NMR. Qualitative detection and structural characterization of F-containing NMR fractions were performed using complementary high-resolution ultra-high performance liquid chromatography tandem mass spectrometry (UHPLC-MS/MS) analyses to further add confidence to the metabolite responses in these fractions. Subsequent 1H-NMR then provided unequivocal standard-free structural confirmation for key metabolites, which would not be possible with conventional radioactivity detection and LC-MS/MS techniques. Alpibectir was shown to undergo extensive hydrolysis of the central amide moiety, where the resultant N-dealkylated amine and trifluorobutyric acid products were detected initially by unbiased 19F-NMR detection along with major downstream biotransformations to form a carbamoyl glucuronide conjugate and trifluoroacetic acid, respectively. Parallel UHPLC-MS/MS analyses provided confirmatory or additional structural characterization only where relevant. These concerted data allowed for the qualitative metabolic profile and quantitative determination of drug-related material (DRM) in urine and plasma, along with the percentage of dose excreted in urine, to be reported in a comprehensive, efficient, and data-led manner. SIGNIFICANCE STATEMENT: Combining the selectivity of 19F-NMR (nuclear magnetic resonance) for unfractionated samples as first-intent, data-led sample fractionation prior to 19F-NMR and structure-rich 1H-NMR detection, along with the sensitivity of high-resolution ultra-high performance liquid chromatography tandem mass spectrometry (UHPLC-MS/MS), a novel alternative for time-efficient detection and quantification of drug-related material (DRM) in human without use of radiolabeled drug is reported. This allowed more complete data rationalization of human metabolism, permitting early risk assessment and progression of the development of antitubercular agent, alpibectir.


Asunto(s)
Espectroscopía de Resonancia Magnética , Humanos , Espectroscopía de Resonancia Magnética/métodos , Espectrometría de Masas en Tándem/métodos , Cromatografía Líquida de Alta Presión/métodos , Masculino , Antituberculosos/farmacocinética , Antituberculosos/orina , Antituberculosos/metabolismo , Metabolómica/métodos , Adulto
7.
Molecules ; 29(3)2024 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-38338462

RESUMEN

Tuberculosis is one of the most common infectious diseases in the world, caused by Mycobacterium tuberculosis. The outbreak of multiple drug-resistant tuberculosis has become a major challenge to prevent this disease worldwide. ClpC1 is a Clp ATPase protein of Mycobacterium tuberculosis, functioning as a chaperon when combined with the Clp complex. ClpC1 has emerged as a new target to discover anti-tuberculosis drugs. This study aimed to explore the ClpC1 inhibitors from actinomycetes, which have been known to provide abundant sources of antibiotics. Two cyclic peptides, including nocardamin (1), halolitoralin A (3), and a lactone pleurone (2), were isolated from the culture of Streptomyces aureus (VTCC43181). The structures of these compounds were determined based on the detailed analysis of their spectral data and comparison with references. This is the first time these compounds have been isolated from S. aureus. Compounds 1-3 were evaluated for their affection of ATPase activity of the recombinant ClpC1 protein. Of these compounds, halolitoralin A (1), a macrocyclic peptide, was effective for the ATPase hydrolysis of the ClpC1 protein.


Asunto(s)
Mycobacterium tuberculosis , Streptomyces , Staphylococcus aureus/metabolismo , Antituberculosos/farmacología , Antituberculosos/metabolismo , Proteínas Bacterianas/química , Adenosina Trifosfatasas/metabolismo
8.
Microbiology (Reading) ; 169(1)2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36748627

RESUMEN

DprE2 is an essential enzyme in the synthesis of decaprenylphosphoryl-ß-d-arabinofuranose (DPA) and subsequently arabinogalactan, and is a significant new drug target for M. tuberculosis. Two compounds from the GSK-177 box set, GSK301A and GSK032A, were identified through Mt-DprE2-target overexpression studies. The Mt-DprE1-DprE2 complex was co-purified and a new in vitro DprE2 assay developed, based on the oxidation of the reduced nicotinamide adenine dinucleotide cofactor of DprE2 (NADH/NADPH). The Mt-DprE1-DprE2 complex showed interesting kinetics in both the DprE1 resazurin-based assay, where Mt-DprE2 was found to enhance Mt-DprE1 activity and reduce substrate inhibition; and also in the DprE2 assay, which similarly exhibited substrate inhibition and a difference in kinetics of the two potential cofactors, NADH and NADPH. Although, no inhibition was observed in the DprE2 assay by the two GSK set compounds, spontaneous mutant generation indicated a possible explanation in the form of a pro-drug activation pathway, involving fgd1 and fbiC.


Asunto(s)
Mycobacterium tuberculosis , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , NAD/metabolismo , NADP/metabolismo , Antituberculosos/farmacología , Antituberculosos/metabolismo , Proteínas Bacterianas/química
9.
Bioorg Chem ; 138: 106648, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37315451

RESUMEN

CtpF is a Ca2+ transporter P-type ATPase key to the response to stress conditions and to Mycobacterium tuberculosis virulence, therefore, an interesting target for the design of novel anti-Mtb compounds. In this work, molecular dynamics simulations of four previously identified CtpF inhibitors allowed recognizing the key protein-ligand (P-L) interactions, which were then used to perform a pharmacophore-based virtual screening (PBVS) of 22 million compounds from ZINCPharmer. The top-rated compounds were then subjected to molecular docking, and their scores were refined by MM-GBSA calculations. In vitro assays showed that ZINC04030361 (Compound 7) was the best promising candidate, showing a MIC of 25.0 µg/mL, inhibition of Ca2+-ATPase activity (IC50) of 3.3 µM, cytotoxic activity of 27.2 %, and hemolysis of red blood cells lower than 0.2 %. Interestingly, the ctpF gene is upregulated in the presence of compound 7, compared to other alkali/alkaline P-type ATPases coding genes, strongly suggesting that CtpF is a compound 7-specific target.


Asunto(s)
Mycobacterium tuberculosis , Mycobacterium tuberculosis/metabolismo , Simulación del Acoplamiento Molecular , Unión Proteica , Simulación de Dinámica Molecular , Proteínas de Transporte de Membrana/metabolismo , Adenosina Trifosfatasas/metabolismo , Membrana Celular/metabolismo , Antituberculosos/farmacología , Antituberculosos/metabolismo , Proteínas Bacterianas/metabolismo
10.
J Am Chem Soc ; 144(46): 21157-21173, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-36367461

RESUMEN

The mechanism of action (MoA) of a clickable fatty acid analogue 8-(2-cyclobuten-1-yl)octanoic acid (DA-CB) has been investigated for the first time. Proteomics, metabolomics, and lipidomics were combined with a network analysis to investigate the MoA of DA-CB against Mycobacterium smegmatis (Msm). The metabolomics results showed that DA-CB has a general MoA related to that of ethionamide (ETH), a mycolic acid inhibitor that targets enoyl-ACP reductase (InhA), but DA-CB likely inhibits a step downstream from InhA. Our combined multi-omics approach showed that DA-CB appears to disrupt the pathway leading to the biosynthesis of mycolic acids, an essential mycobacterial fatty acid for both Msm and Mycobacterium tuberculosis (Mtb). DA-CB decreased keto-meromycolic acid biosynthesis. This intermediate is essential in the formation of mature mycolic acid, which is a key component of the mycobacterial cell wall in a process that is catalyzed by the essential polyketide synthase Pks13 and the associated ligase FadD32. The multi-omics analysis revealed further collateral alterations in bacterial metabolism, including the overproduction of shorter carbon chain hydroxy fatty acids and branched chain fatty acids, alterations in pyrimidine metabolism, and a predominate downregulation of proteins involved in fatty acid biosynthesis. Overall, the results with DA-CB suggest the exploration of this and related compounds as a new class of tuberculosis (TB) therapeutics. Furthermore, the clickable nature of DA-CB may be leveraged to trace the cellular fate of the modified fatty acid or any derived metabolite or biosynthetic intermediate.


Asunto(s)
Mycobacterium tuberculosis , Ácidos Micólicos , Ácidos Micólicos/metabolismo , Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/metabolismo , Mycobacterium smegmatis/metabolismo , Ácidos Grasos/metabolismo , Antituberculosos/farmacología , Antituberculosos/metabolismo
11.
Antimicrob Agents Chemother ; 66(10): e0103422, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36190243

RESUMEN

Contezolid (MRX-I), a safer antibiotic of the oxazolidinone class, is a promising new antibiotic with potent activity against Mycobacterium tuberculosis (MTB) both in vitro and in vivo. To identify resistance mechanisms of contezolid in MTB, we isolated several in vitro spontaneous contezolid-resistant MTB mutants, which exhibited 16-fold increases in the MIC of contezolid compared with the parent strain but were still unexpectedly susceptible to linezolid. Whole-genome sequencing revealed that most of the contezolid-resistant mutants bore mutations in the mce3R gene, which encodes a transcriptional repressor. The mutations in mce3R led to markedly increased expression of a monooxygenase encoding gene Rv1936. We then characterized Rv1936 as a putative flavin-dependent monooxygenase that catalyzes the degradation of contezolid into its inactive 2,3-dihydropyridin-4-one (DHPO) ring-opened metabolites, thereby conferring drug resistance. While contezolid is an attractive drug candidate with potent antimycobacterial activity and low toxicity, the occurrence of mutations in Mce3R should be considered when designing combination therapy using contezolid for treating tuberculosis.


Asunto(s)
Mycobacterium tuberculosis , Oxazolidinonas , Linezolid , Antibacterianos , Mutación , Oxigenasas de Función Mixta/metabolismo , Flavinas/genética , Flavinas/metabolismo , Antituberculosos/farmacología , Antituberculosos/metabolismo , Pruebas de Sensibilidad Microbiana
12.
Antimicrob Agents Chemother ; 66(1): e0146521, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34780266

RESUMEN

Tuberculosis (TB), caused by Mycobacterium tuberculosis, is one of the most fatal diseases in the world. Methylenetetrahydrofolate reductase (MTHFR) catalyzes the production of 5-methyltetrahydrofolate (5-CH3-THF), which is required for the de novo biosynthesis of methionine in bacteria. Here, we identified Rv2172c as an MTHFR in M. tuberculosis through in vitro and in vivo analyses and determined that the protein is essential for the in vitro growth of the bacterium. Subsequently, we constructed rv2172c R159N and L214A mutants in M. tuberculosis and found that these mutants were more sensitive to the antifolates para-aminosalicylic acid (PAS) and sulfamethoxazole (SMX). Combining biochemical and genetic methods, we found that rv2172c R159N or L214A mutation impaired methionine production, leading to increased susceptibility of M. tuberculosis to PAS, which was largely restored by adding exogenous methionine. Moreover, overexpression of rv2172c in M. tuberculosis could increase methionine production and lead to PAS resistance. This research is the first to identify an MTHFR in M. tuberculosis and reveals that the activity of this enzyme is associated with susceptibility to antifolates. These findings have particular value for antitubercular drug design for the treatment of drug-resistant TB.


Asunto(s)
Ácido Aminosalicílico , Mycobacterium tuberculosis , Ácido Aminosalicílico/metabolismo , Ácido Aminosalicílico/farmacología , Antituberculosos/metabolismo , Antituberculosos/farmacología , Proteínas Bacterianas/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/metabolismo
13.
J Comput Chem ; 43(9): 619-630, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35167132

RESUMEN

In this study, we assess the effective inhibition of a series of thiazolidine derivatives (1a-1q) were adopting structure-based drug design. Thiazolidine is a five-membered ring structure with thioether and amino groups at positions 1 and 3. Although, thiazolidine may bind to a wide range of protein targets, it is a major heterocyclic core in medicinal chemistry. Different scoring utilities including AutoDock Vina, Glide, and MM/GBSA analysis were performed to commensurate the improvement of screening progress. The evaluated binding affinities were validated by molecular dynamics simulations over a period of 20 ns for the interactions between the Mycobacterium tuberculosis protein PrpR with three novel scaffolds (1b, 1j, and 1k). All the scaffolds exhibited distinct stable interactions with the significant residues like Arg169, Arg197, Tyr248, Arg308, and Gly311 respectively. Further, the inhibitory activities of scaffolds were predicted and evaluated by machine learning based algorithm to rank the above proposed compounds. This study reveals the potential of 1k and 1j as effective inhibitor candidates for the treatment of tuberculosis.


Asunto(s)
Mycobacterium tuberculosis , Antituberculosos/química , Antituberculosos/metabolismo , Antituberculosos/farmacología , Diseño de Fármacos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Mycobacterium tuberculosis/metabolismo
14.
Bioorg Med Chem ; 74: 117046, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36228522

RESUMEN

Tuberculosis (TB) remains a leading cause of infectious disease-related mortality and morbidity. Pyrazinamide (PZA) is a critical component of the first-line TB treatment regimen because of its sterilizing activity against non-replicating Mycobacterium tuberculosis (Mtb), but its mechanism of action has remained enigmatic. PZA is a prodrug converted by pyrazinamidase encoded by pncA within Mtb to the active moiety, pyrazinoic acid (POA) and PZA resistance is caused by loss-of-function mutations to pyrazinamidase. We have recently shown that POA induces targeted protein degradation of the enzyme PanD, a crucial component of the coenzyme A biosynthetic pathway essential in Mtb. Based on the newly identified mechanism of action of POA, along with the crystal structure of PanD bound to POA, we designed several POA analogs using structure for interpretation to improve potency and overcome PZA resistance. We prepared and tested ring and carboxylic acid bioisosteres as well as 3, 5, 6 substitutions on the ring to study the structure activity relationships of the POA scaffold. All the analogs were evaluated for their whole cell antimycobacterial activity, and a few representative molecules were evaluated for their binding affinity, towards PanD, through isothermal titration calorimetry. We report that analogs with ring and carboxylic acid bioisosteres did not significantly enhance the antimicrobial activity, whereas the alkylamino-group substitutions at the 3 and 5 position of POA were found to be up to 5 to 10-fold more potent than POA. Further development and mechanistic analysis of these analogs may lead to a next generation POA analog for treating TB.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Pirazinamida/farmacología , Pirazinamida/metabolismo , Antituberculosos/farmacología , Antituberculosos/metabolismo , Amidohidrolasas/metabolismo , Tuberculosis/microbiología , Mutación , Relación Estructura-Actividad , Ácidos Carboxílicos/metabolismo , Pruebas de Sensibilidad Microbiana , Farmacorresistencia Bacteriana
15.
Microbiology (Reading) ; 167(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33555244

RESUMEN

Treatment of tuberculosis requires a multi-drug regimen administered for at least 6 months. The long-term chemotherapy is attributed in part to a minor subpopulation of nonreplicating Mycobacterium tuberculosis cells that exhibit phenotypic tolerance to antibiotics. The origins of these cells in infected hosts remain unclear. Here we discuss some recent evidence supporting the hypothesis that hibernation of ribosomes in M. tuberculosis, induced by zinc starvation, could be one of the primary mechanisms driving the development of nonreplicating persisters in hosts. We further analyse inconsistencies in previously reported studies to clarify the molecular principles underlying mycobacterial ribosome hibernation.


Asunto(s)
Mycobacterium/fisiología , Tuberculosis/microbiología , Antituberculosos/metabolismo , Antituberculosos/uso terapéutico , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , Humanos , Mycobacterium/efectos de los fármacos , Mycobacterium/metabolismo , Proteínas Ribosómicas/química , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Tuberculosis/tratamiento farmacológico , Zinc/deficiencia
16.
Chem Res Toxicol ; 34(8): 1879-1889, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34319702

RESUMEN

A chemical activation study of the thiocarbonyl-type antitubercular prodrugs, ethionamide (ETH), thioacetazone (TAZ), and isoxyl (ISO), was performed. Biomimetic oxidation of ethionamide using H2O2 (1 equiv) led to ETH-SO as the only stable S-oxide compound, which was found to occur in solution in the preferential form of a sulfine (ETH═S═O vs the sulfenic acid tautomer ETH-S-OH), as previously observed in the crystal state. It was also demonstrated that ETH-SO is capable of reacting with amines, as the putative sulfinic derivative (ETH-SO2H) was supposed to do. Unlike ETH, oxidation of TAZ did not allow observation of the mono-oxygenated species (TAZ-SO), leading directly to the more stable sulfinic acid derivative (TAZ-SO2H), which can then lose a SOxH group after further oxidation or when placed in a basic medium. It was also noticed that the unstable TAZ-SO intermediate can lead to the carbodiimide derivative as another electrophilic species. It is suggested that TAZ-SOH, TAZ-SO2H, and the carbodiimide compound can also react with NH2-containing nucleophilic species, and therefore be involved in toxic effects. Finally, ISO showed a very complex reactivity, here assigned to the coexistence of two mono-oxygenated structures, the sulfine and sulfenic acid tautomers. The mono- and dioxygenated derivatives of ISO are also highly unstable, leading to a panel of multiple metabolites, which are still reactive and likely contribute to the toxicity of this prodrug.


Asunto(s)
Antituberculosos/metabolismo , Etionamida/metabolismo , Feniltiourea/análogos & derivados , Profármacos/metabolismo , Tioacetazona/metabolismo , Antituberculosos/química , Etionamida/química , Peróxido de Hidrógeno/metabolismo , Modelos Moleculares , Oxidación-Reducción , Feniltiourea/química , Feniltiourea/metabolismo , Profármacos/química , Tioacetazona/química
17.
Org Biomol Chem ; 19(8): 1818-1826, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33565547

RESUMEN

An in situ screening assay for UDP-galactopyranose mutase (UGM, an essential enzyme of M. tuberculosis cell wall biosynthesis) has been developed to discover novel UGM inhibitors. The approach is based on the amide-forming reaction of an amino acid core with various cinnamic acids, followed by a direct fluorescence polarization assay to identify the best UGM binders without isolation and purification of the screened ligands. This assay allows us to perform one-pot high-throughput synthesis and screening of enzyme inhibitors in a 384-well plate format. UGM ligands were successfully identified by this technology and their inhibition levels were established from pure synthetic compounds in vitro and in a whole cell antibacterial assay. This study provides a blueprint for designing enamide structures as new UGM inhibitors and anti-mycobacterial agents.


Asunto(s)
Aminoácidos/farmacología , Antituberculosos/farmacología , Cinamatos/farmacología , Inhibidores Enzimáticos/farmacología , Transferasas Intramoleculares/antagonistas & inhibidores , Aminoácidos/síntesis química , Aminoácidos/metabolismo , Antituberculosos/síntesis química , Antituberculosos/metabolismo , Cinamatos/síntesis química , Cinamatos/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Transferasas Intramoleculares/química , Transferasas Intramoleculares/metabolismo , Cinética , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Mycobacterium bovis/efectos de los fármacos , Mycobacterium bovis/enzimología , Mycobacterium tuberculosis/enzimología , Unión Proteica
18.
J Immunol ; 202(2): 494-502, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30552162

RESUMEN

Drug-resistant mycobacteria are a rising problem worldwide. There is an urgent need to understand the immune response to tuberculosis to identify host targets that, if targeted therapeutically, could be used to tackle these currently untreatable infections. In this study we use an Il-1ß fluorescent transgenic line to show that there is an early innate immune proinflammatory response to well-established zebrafish models of inflammation and Mycobacterium marinum infection. We demonstrate that host-derived hypoxia signaling, mediated by the Hif-1α transcription factor, can prime macrophages with increased levels of Il-1ß in the absence of infection, upregulating neutrophil antimicrobial NO production, leading to greater protection against infection. Our data link Hif-1α to proinflammatory macrophage Il-1ß transcription in vivo during early mycobacterial infection and importantly highlight a host protective mechanism, via antimicrobial NO, that decreases disease outcomes and that could be targeted therapeutically to stimulate the innate immune response to better deal with infections.


Asunto(s)
Hipoxia/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium marinum/fisiología , Mycobacterium tuberculosis/fisiología , Tuberculosis/inmunología , Animales , Animales Modificados Genéticamente , Antituberculosos/metabolismo , Células Cultivadas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Proteínas Fluorescentes Verdes/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunidad Innata , Interleucina-1beta/genética , Óxido Nítrico/metabolismo , Pez Cebra
19.
Phys Chem Chem Phys ; 23(40): 23233-23241, 2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34623361

RESUMEN

Ethionamide (ETH) is a high-profile drug for the treatment of patients with multidrug-resistant Mycobacterium tuberculosis and, in order to produce its inhibitory effects, it needs to be bioactivated by monooxygenase EthA. This process is under the control of the transcriptional repressors EthR and EthR2, so that their inhibition results in the boosting of ethionamide activation. Herein, through crystallographic data and computer simulations, we calculated the interaction binding energies of four inhibitors with improved in vitro potency, namely BDM76060 (PDB ID: 6HS1), BDM72201 (PDB ID: 6HRX), BDM76150 (PDB ID: 6HS2) and BDM72719 (PDB ID: 6HRY), in complexes with the transcriptional repressor EthR2, using density functional theory (DFT) within the molecular fractionation with conjugated caps (MFCC) approach. It was observed that these ligands share the same binding site within a 10.0 Å radius of the EthR2 protein; however, their structural particularities have a significant impact on the global energies of systems. The BDM72201 and BDM72719 components are weakly attached to the binding site, while BDM76060 and BDM76150 components produce stronger bonds, corroborating with experimental studies demonstrating that BDM76060 and BDM76150 are more successful in producing inhibitory effects. BDM76060 and BDM76150 have many functional groups that increase the contact surface with the protein and attract a more significant number of amino acid residues, being able to produce polarities that generate stronger interactions. In the current scenario of a growing number of cases of bacterial resistance, the obtained data can be used to guide clinical trials of these inhibitors and other inhibitors that act on the alternative EthR2 pathway, focusing on improving the activity of ethionamide, its effectiveness, a reduction in the treatment time and exposure to cytotoxic effects.


Asunto(s)
Antituberculosos/química , Etionamida/química , Proteínas Represoras/química , Antituberculosos/metabolismo , Antituberculosos/uso terapéutico , Sitios de Unión , Teoría Funcional de la Densidad , Etionamida/metabolismo , Etionamida/uso terapéutico , Humanos , Ligandos , Simulación de Dinámica Molecular , Mycobacterium tuberculosis/metabolismo , Proteínas Represoras/metabolismo , Tuberculosis/tratamiento farmacológico
20.
Bioorg Chem ; 115: 105133, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34329993

RESUMEN

Novel chemotherapeutic agents against multidrug resistant-tuberculosis (MDR-TB) are urgently needed at this juncture to save the life of TB-infected patients. In this work, we have synthesized and characterized novel isatin hydrazones 4(a-o) and their thiomorpholine tethered analogues 5(a-o). All the synthesized compounds were initially screened for their anti-mycobacterial activity against the H37Rv strain of Mycobacterium tuberculosis (MTB) under level-I testing. Remarkably, five compounds 4f, 4h, 4n, 5f and 5m (IC50 = 1.9 µM to 9.8 µM) were found to be most active, with 4f (IC50 = 1.9 µM) indicating highest inhibition of H37Rv. These compounds were further evaluated at level-II testing against the five drug-resistant strains such as isoniazid-resistant strains (INH-R1 and INH-R2), rifampicin-resistant strains (RIF-R1 and RIF-R2) and fluoroquinolone-resistant strain (FQ-R1) of MTB. Interestingly, 4f and 5f emerged as the most potent compounds with IC50 of 3.6 µM and 1.9 µM against RIF-R1 MTB strain, followed by INH-R1 MTB strain with IC50 of 3.5 µM and 3.4 µM, respectively. Against FQ-R1 MTB strain, the lead compounds 4f and 5f displayed excellent inhibition at IC50 5.9 µM and 4.9 µM, respectively indicating broad-spectrum of activity. Further, molecular docking, ADME pharmacokinetic and molecular dynamics simulations of the compounds were performed against the DNA gyrase B and obtained encouraging results.


Asunto(s)
Antituberculosos/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Hidrazonas/química , Isatina/química , Morfolinas/química , Mycobacterium tuberculosis/efectos de los fármacos , Antituberculosos/química , Antituberculosos/metabolismo , Sitios de Unión , Supervivencia Celular/efectos de los fármacos , Girasa de ADN/química , Girasa de ADN/metabolismo , Diseño de Fármacos , Semivida , Humanos , Hidrazonas/metabolismo , Hidrazonas/farmacología , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Rifampin/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA