Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
PLoS Biol ; 17(2): e3000123, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30716063

RESUMO

The diffusible signal factors (DSFs) are a family of quorum-sensing autoinducers (AIs) produced and detected by numerous gram-negative bacteria. The DSF family AIs are fatty acids, differing in their acyl chain length, branching, and substitution but having in common a cis-2 double bond that is required for their activity. In both human and plant pathogens, DSFs regulate diverse phenotypes, including virulence factor expression, antibiotic resistance, and biofilm dispersal. Despite their widespread relevance to both human health and agriculture, the molecular basis of DSF recognition by their cellular receptors remained a mystery. Here, we report the first structure-function studies of the DSF receptor regulation of pathogenicity factor R (RpfR). We present the X-ray crystal structure of the RpfR DSF-binding domain in complex with the Burkholderia DSF (BDSF), which to our knowledge is the first structure of a DSF receptor in complex with its AI. To begin to understand the mechanistic role of the BDSF-RpfR contacts observed in the biologically important complex, we have also determined the X-ray crystal structure of the RpfR DSF-binding domain in complex with the inactive, saturated isomer of BDSF, dodecanoic acid (C12:0). In addition to these ligand-receptor complex structures, we report the discovery of a previously overlooked RpfR domain and show that it binds to and negatively regulates the DSF synthase regulation of pathogenicity factor F (RpfF). We have named this RpfR region the RpfF interaction (FI) domain, and we have determined its X-ray crystal structure alone and in complex with RpfF. These X-ray crystal structures, together with extensive complementary in vivo and in vitro functional studies, reveal the molecular basis of DSF recognition and the importance of the cis-2 double bond to DSF function. Finally, we show that throughout cellular growth, the production of BDSF by RpfF is post-translationally controlled by the RpfR N-terminal FI domain, affecting the cellular concentration of the bacterial second messenger bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP). Thus, in addition to describing the molecular basis for the binding and specificity of a DSF for its receptor, we describe a receptor-synthase interaction regulating bacterial quorum-sensing signaling and second messenger signal transduction.


Assuntos
Proteínas de Bactérias/metabolismo , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Proteínas de Bactérias/química , Burkholderia/metabolismo , Cristalização , Cristalografia por Raios X , GMP Cíclico/biossíntese , Ácidos Láuricos/química , Ácidos Láuricos/metabolismo , Modelos Moleculares , Ligação Proteica , Domínios Proteicos , Percepção de Quorum
2.
mBio ; 8(1)2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28196957

RESUMO

Active tuberculosis (TB) and latent Mycobacterium tuberculosis infection both require lengthy treatments to achieve durable cures. This problem has partly been attributable to the existence of nonreplicating M. tuberculosis "persisters" that are difficult to kill using conventional anti-TB treatments. Compounds that target the respiratory pathway have the potential to kill both replicating and persistent M. tuberculosis and shorten TB treatment, as this pathway is essential in both metabolic states. We developed a novel respiratory pathway-specific whole-cell screen to identify new respiration inhibitors. This screen identified the biphenyl amide GSK1733953A (DG70) as a likely respiration inhibitor. DG70 inhibited both clinical drug-susceptible and drug-resistant M. tuberculosis strains. Whole-genome sequencing of DG70-resistant colonies identified mutations in menG (rv0558), which is responsible for the final step in menaquinone biosynthesis and required for respiration. Overexpression of menG from wild-type and DG70-resistant isolates increased the DG70 MIC by 4× and 8× to 30×, respectively. Radiolabeling and high-resolution mass spectrometry studies confirmed that DG70 inhibited the final step in menaquinone biosynthesis. DG70 also inhibited oxygen utilization and ATP biosynthesis, which was reversed by external menaquinone supplementation. DG70 was bactericidal in actively replicating cultures and in a nutritionally deprived persistence model. DG70 was synergistic with the first-line TB drugs isoniazid, rifampin, and the respiratory inhibitor bedaquiline. The combination of DG70 and isoniazid completely sterilized cultures in the persistence model by day 10. These results suggest that MenG is a good therapeutic target and that compounds targeting MenG along with standard TB therapy have the potential to shorten TB treatment duration.IMPORTANCE This study shows that MenG, which is responsible for the last enzymatic step in menaquinone biosynthesis, may be a good drug target for improving TB treatments. We describe the first small-molecule inhibitor (DG70) of Mycobacterium tuberculosis MenG and show that DG70 has characteristics that are highly desirable for a new antitubercular agent, including bactericidality against both actively growing and nonreplicating mycobacteria and synergy with several first-line drugs that are currently used to treat TB.


Assuntos
Antituberculosos/farmacologia , Compostos de Bifenilo/isolamento & purificação , Compostos de Bifenilo/farmacologia , Descoberta de Drogas , Metiltransferases/antagonistas & inibidores , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Trifosfato de Adenosina/biossíntese , Compostos de Bifenilo/química , Farmacorresistência Bacteriana , Humanos , Metiltransferases/química , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/enzimologia , Bibliotecas de Moléculas Pequenas/análise , Vitamina K 2/análogos & derivados , Vitamina K 2/metabolismo , Vitamina K 2/farmacologia
3.
PLoS One ; 10(10): e0141076, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26517557

RESUMO

Integrated computational approaches for Mycobacterium tuberculosis (Mtb) are useful to identify new molecules that could lead to future tuberculosis (TB) drugs. Our approach uses information derived from the TBCyc pathway and genome database, the Collaborative Drug Discovery TB database combined with 3D pharmacophores and dual event Bayesian models of whole-cell activity and lack of cytotoxicity. We have prioritized a large number of molecules that may act as mimics of substrates and metabolites in the TB metabolome. We computationally searched over 200,000 commercial molecules using 66 pharmacophores based on substrates and metabolites from Mtb and further filtering with Bayesian models. We ultimately tested 110 compounds in vitro that resulted in two compounds of interest, BAS 04912643 and BAS 00623753 (MIC of 2.5 and 5 µg/mL, respectively). These molecules were used as a starting point for hit-to-lead optimization. The most promising class proved to be the quinoxaline di-N-oxides, evidenced by transcriptional profiling to induce mRNA level perturbations most closely resembling known protonophores. One of these, SRI58 exhibited an MIC = 1.25 µg/mL versus Mtb and a CC50 in Vero cells of >40 µg/mL, while featuring fair Caco-2 A-B permeability (2.3 x 10-6 cm/s), kinetic solubility (125 µM at pH 7.4 in PBS) and mouse metabolic stability (63.6% remaining after 1 h incubation with mouse liver microsomes). Despite demonstration of how a combined bioinformatics/cheminformatics approach afforded a small molecule with promising in vitro profiles, we found that SRI58 did not exhibit quantifiable blood levels in mice.


Assuntos
Antituberculosos/farmacologia , Biologia Computacional/métodos , Metaboloma/efeitos dos fármacos , Mycobacterium tuberculosis/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antituberculosos/química , Teorema de Bayes , Células CACO-2 , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia , Células Vero
4.
J Chem Inf Model ; 55(3): 645-59, 2015 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-25636146

RESUMO

Isoniazid (INH) is usually administered to treat latent Mycobacterium tuberculosis (Mtb) infections and is used in combination therapy to treat active tuberculosis (TB). Unfortunately, resistance to this drug is hampering its clinical effectiveness. INH is a prodrug that must be activated by Mtb catalase-peroxidase (KatG) before it can inhibit InhA (Mtb enoyl-acyl-carrier-protein reductase). Isoniazid-resistant cases of TB found in clinical settings usually involve mutations in or deletion of katG, which abrogate INH activation. Compounds that inhibit InhA without requiring prior activation by KatG would not be affected by this resistance mechanism and hence would display continued potency against these drug-resistant isolates of Mtb. Virtual screening experiments versus InhA in the GO Fight Against Malaria (GO FAM) project were designed to discover new scaffolds that display base-stacking interactions with the NAD cofactor. GO FAM experiments included targets from other pathogens, including Mtb, when they had structural similarity to a malaria target. Eight of the 16 soluble compounds identified by docking against InhA plus visual inspection were modest inhibitors and did not require prior activation by KatG. The best two inhibitors discovered are both fragment-sized compounds and displayed Ki values of 54 and 59 µM, respectively. Importantly, the novel inhibitors discovered have low structural similarity to known InhA inhibitors and thus help expand the number of chemotypes on which future medicinal chemistry efforts can be focused. These new fragment hits could eventually help advance the fight against INH-resistant Mtb strains, which pose a significant global health threat.


Assuntos
Antituberculosos/química , Antituberculosos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Simulação de Acoplamento Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Oxirredutases/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Catalase/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Farmacorresistência Bacteriana , Isoniazida/farmacologia , Cinética , Testes de Sensibilidade Microbiana
5.
J Chem Inf Model ; 54(7): 2157-65, 2014 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-24968215

RESUMO

Tuberculosis is a major, neglected disease for which the quest to find new treatments continues. There is an abundance of data from large phenotypic screens in the public domain against Mycobacterium tuberculosis (Mtb). Since machine learning methods can learn from past data, we were interested in addressing whether more data builds better models. We now describe using Bayesian machine learning to assess whether we can improve our models by combining the large quantities of single-point data with the much smaller (higher quality) dual-event data sets, which use both dose-response data for both whole-cell antitubercular activity and Vero cell cytotoxicity. We have evaluated 12 models ranging from different single-point, dual-event dose-response, single-point and dual-event dose-response as well as combined data sets for three distinct data sets from the same laboratory. We used a fourth data set of active and inactive compounds from the same group as well as a smaller set of 177 active compounds from GlaxoSmithKline as test sets. Our data suggest combining single-point with dual-event dose-response data does not diminish the internal or external predictive ability of the models based on the receiver operator curve (ROC) for these models (internal ROC range 0.83-0.91, external ROC range 0.62-0.83) compared to the orders of magnitude smaller dual-event models (internal ROC range 0.6-0.83 and external ROC 0.54-0.83). In conclusion, models developed with 1200-5000 compounds appear to be as predictive as those generated with 25 000-350 000 molecules. Our results have implications for justifying further high-throughput screening versus focused testing based on model predictions.


Assuntos
Antituberculosos/farmacologia , Inteligência Artificial , Avaliação Pré-Clínica de Medicamentos/métodos , Informática/métodos , Mycobacterium tuberculosis/efeitos dos fármacos , Animais , Antituberculosos/toxicidade , Teorema de Bayes , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Células Vero
6.
Chem Biol ; 21(7): 819-30, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-24954008

RESUMO

In this study, we identified antifolates with potent, targeted activity against whole-cell Mycobacterium tuberculosis (MTB). Liquid chromatography-mass spectrometry analysis of antifolate-treated cultures revealed metabolic disruption, including decreased pools of methionine and S-adenosylmethionine. Transcriptomic analysis highlighted altered regulation of genes involved in the biosynthesis and utilization of these two compounds. Supplementation with amino acids or S-adenosylmethionine was sufficient to rescue cultures from antifolate treatment. Instead of the "thymineless death" that characterizes folate pathway inhibition in a wide variety of organisms, these data suggest that MTB is vulnerable to a critical disruption of the reactions centered around S-adenosylmethionione, the activated methyl cycle.


Assuntos
Antituberculosos/farmacologia , Antagonistas do Ácido Fólico/farmacologia , Ácido Fólico/metabolismo , Metionina/análogos & derivados , Metionina/metabolismo , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/metabolismo , Di-Hidropteroato Sintase/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Humanos , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/genética , S-Adenosilmetionina/metabolismo , Especificidade da Espécie , Tetra-Hidrofolato Desidrogenase/metabolismo , Triazinas/farmacologia
7.
Chem Biol ; 20(3): 370-8, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23521795

RESUMO

Identification of unique leads represents a significant challenge in drug discovery. This hurdle is magnified in neglected diseases such as tuberculosis. We have leveraged public high-throughput screening (HTS) data to experimentally validate a virtual screening approach employing Bayesian models built with bioactivity information (single-event model) as well as bioactivity and cytotoxicity information (dual-event model). We virtually screened a commercial library and experimentally confirmed actives with hit rates exceeding typical HTS results by one to two orders of magnitude. This initial dual-event Bayesian model identified compounds with antitubercular whole-cell activity and low mammalian cell cytotoxicity from a published set of antimalarials. The most potent hit exhibits the in vitro activity and in vitro/in vivo safety profile of a drug lead. These Bayesian models offer significant economies in time and cost to drug discovery.


Assuntos
Antituberculosos/farmacologia , Antituberculosos/toxicidade , Descoberta de Drogas , Animais , Teorema de Bayes , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos , Feminino , Concentração Inibidora 50 , Macrófagos/efeitos dos fármacos , Camundongos , Mycobacterium tuberculosis/efeitos dos fármacos , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA