Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Chem Biol ; 31(4): 683-698.e7, 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38151019

RESUMO

Mycobacterial bioenergetics is a validated target space for antitubercular drug development. Here, we identify BB2-50F, a 6-substituted 5-(N,N-hexamethylene)amiloride derivative as a potent, multi-targeting bioenergetic inhibitor of Mycobacterium tuberculosis. We show that BB2-50F rapidly sterilizes both replicating and non-replicating cultures of M. tuberculosis and synergizes with several tuberculosis drugs. Target identification experiments, supported by docking studies, showed that BB2-50F targets the membrane-embedded c-ring of the F1Fo-ATP synthase and the catalytic subunit (substrate-binding site) of succinate dehydrogenase. Biochemical assays and metabolomic profiling showed that BB2-50F inhibits succinate oxidation, decreases the activity of the tricarboxylic acid (TCA) cycle, and results in succinate secretion from M. tuberculosis. Moreover, we show that the lethality of BB2-50F under aerobic conditions involves the accumulation of reactive oxygen species. Overall, this study identifies BB2-50F as an effective inhibitor of M. tuberculosis and highlights that targeting multiple components of the mycobacterial respiratory chain can produce fast-acting antimicrobials.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Humanos , Succinato Desidrogenase/metabolismo , Succinato Desidrogenase/farmacologia , Antituberculosos/química , Tuberculose/tratamento farmacológico , Trifosfato de Adenosina , Inibidores Enzimáticos/farmacologia , Succinatos
2.
Cell Rep ; 42(5): 112444, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37115669

RESUMO

The bioenergetic mechanisms by which Mycobacterium tuberculosis survives hypoxia are poorly understood. Current models assume that the bacterium shifts to an alternate electron acceptor or fermentation to maintain membrane potential and ATP synthesis. Counterintuitively, we find here that oxygen itself is the principal terminal electron acceptor during hypoxic dormancy. M. tuberculosis can metabolize oxygen efficiently at least two orders of magnitude below the concentration predicted to occur in hypoxic lung granulomas. Despite a difference in apparent affinity for oxygen, both the cytochrome bcc:aa3 and cytochrome bd oxidase respiratory branches are required for hypoxic respiration. Simultaneous inhibition of both oxidases blocks oxygen consumption, reduces ATP levels, and kills M. tuberculosis under hypoxia. The capacity of mycobacteria to scavenge trace levels of oxygen, coupled with the absence of complex regulatory mechanisms to achieve hierarchal control of the terminal oxidases, may be a key determinant of long-term M. tuberculosis survival in hypoxic lung granulomas.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Humanos , Mycobacterium tuberculosis/metabolismo , Oxigênio/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Oxirredutases/metabolismo , Homeostase , Tuberculose/microbiologia , Hipóxia , Trifosfato de Adenosina/metabolismo , Citocromos/metabolismo
3.
Commun Biol ; 5(1): 166, 2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35210534

RESUMO

Increasing antimicrobial resistance compels the search for next-generation inhibitors with differing or multiple molecular targets. In this regard, energy conservation in Mycobacterium tuberculosis has been clinically validated as a promising new drug target for combatting drug-resistant strains of M. tuberculosis. Here, we show that HM2-16F, a 6-substituted derivative of the FDA-approved drug amiloride, is an anti-tubercular inhibitor with bactericidal properties comparable to the FDA-approved drug bedaquiline (BDQ; Sirturo®) and inhibits the growth of bedaquiline-resistant mutants. We show that HM2-16F weakly inhibits the F1Fo-ATP synthase, depletes ATP, and affects the entry of acetyl-CoA into the Krebs cycle. HM2-16F synergizes with the cytochrome bcc-aa3 oxidase inhibitor Q203 (Telacebec) and co-administration with Q203 sterilizes in vitro cultures in 14 days. Synergy with Q203 occurs via direct inhibition of the cytochrome bd oxidase by HM2-16F. This study shows that amiloride derivatives represent a promising discovery platform for targeting energy generation in drug-resistant tuberculosis.


Assuntos
Mycobacterium tuberculosis , Trifosfato de Adenosina , Amilorida/farmacologia , Antituberculosos/farmacologia , Citocromos , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Mycobacterium tuberculosis/metabolismo , Oxirredutases
4.
Prog Biophys Mol Biol ; 152: 35-44, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31733221

RESUMO

Cellular bioenergetics is an area showing promise for the development of new antimicrobials, antimalarials and cancer therapy. Enzymes involved in central carbon metabolism and energy generation are essential mediators of bacterial physiology, persistence and pathogenicity, lending themselves natural interest for drug discovery. In particular, succinate and malate are two major focal points in both the central carbon metabolism and the respiratory chain of Mycobacterium tuberculosis. Both serve as direct links between the citric acid cycle and the respiratory chain due to the quinone-linked reactions of succinate dehydrogenase, fumarate reductase and malate:quinone oxidoreductase. Inhibitors against these enzymes therefore hold the promise of disrupting two distinct, but essential, cellular processes at the same time. In this review, we discuss the roles and unique adaptations of these enzymes and critically evaluate the role that future inhibitors of these complexes could play in the bioenergetics target space.


Assuntos
Antituberculosos/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/farmacologia , Succinato Desidrogenase/farmacologia , Tuberculose/tratamento farmacológico , Benzoquinonas/metabolismo , Ciclo do Ácido Cítrico/efeitos dos fármacos , Descoberta de Drogas , Humanos , Malatos/metabolismo , Oxirredução , Ligação Proteica , Ácido Succínico/metabolismo
5.
Sci Rep ; 9(1): 16759, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31727946

RESUMO

The dynamic interaction of the N- and C-terminal domains of mycobacterial F-ATP synthase subunit ε is proposed to contribute to efficient coupling of H+-translocation and ATP synthesis. Here, we investigate crosstalk between both subunit ε domains by introducing chromosomal atpC missense mutations in the C-terminal helix 2 of ε predicted to disrupt inter domain and subunit ε-α crosstalk and therefore coupling. The ε mutant εR105A,R111A,R113A,R115A (ε4A) showed decreased intracellular ATP, slower growth rates and lower molar growth yields on non-fermentable carbon sources. Cellular respiration and metabolism were all accelerated in the mutant strain indicative of dysregulated oxidative phosphorylation. The ε4A mutant exhibited an altered colony morphology and was hypersusceptible to cell wall-acting antimicrobials suggesting defective cell wall biosynthesis. In silico screening identified a novel mycobacterial F-ATP synthase inhibitor disrupting ε's coupling activity demonstrating the potential to advance this regulation as a new area for mycobacterial F-ATP synthase inhibitor development.


Assuntos
Trifosfato de Adenosina/metabolismo , Mutação , Mycobacterium/crescimento & desenvolvimento , ATPases Translocadoras de Prótons/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Parede Celular , Simulação por Computador , Metabolismo Energético , Modelos Moleculares , Mycobacterium/enzimologia , Mycobacterium/genética , Conformação Proteica , Estrutura Secundária de Proteína , ATPases Translocadoras de Prótons/química , ATPases Translocadoras de Prótons/genética
6.
Proc Natl Acad Sci U S A ; 115(28): 7326-7331, 2018 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-29941569

RESUMO

Bedaquiline (BDQ), an inhibitor of the mycobacterial F1Fo-ATP synthase, has revolutionized the antitubercular drug discovery program by defining energy metabolism as a potent new target space. Several studies have recently suggested that BDQ ultimately causes mycobacterial cell death through a phenomenon known as uncoupling. The biochemical basis underlying this, in BDQ, is unresolved and may represent a new pathway to the development of effective therapeutics. In this communication, we demonstrate that BDQ can inhibit ATP synthesis in Escherichia coli by functioning as a H+/K+ ionophore, causing transmembrane pH and potassium gradients to be equilibrated. Despite the apparent lack of a BDQ-binding site, incorporating the E. coli Fo subunit into liposomes enhanced the ionophoric activity of BDQ. We discuss the possibility that localization of BDQ at F1Fo-ATP synthases enables BDQ to create an uncoupled microenvironment, by antiporting H+/K+ Ionophoric properties may be desirable in high-affinity antimicrobials targeting integral membrane proteins.


Assuntos
Trifosfato de Adenosina/biossíntese , Antituberculosos/farmacologia , Diarilquinolinas/farmacologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Ionóforos/farmacologia , ATPases Translocadoras de Prótons/metabolismo , Concentração de Íons de Hidrogênio
7.
Drug Resist Updat ; 36: 1-12, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29499834

RESUMO

From the war on drug resistance, through cancer biology, even to agricultural and environmental protection: there is a huge demand for rapid and effective solutions to control infections and diseases. The development of small molecule inhibitors was once an accepted "one-size fits all" approach to these varied problems, but persistence and resistance threaten to return society to a pre-antibiotic era. Only five essential cellular targets in bacteria have been developed for the majority of our clinically-relevant antibiotics. These include: cell wall synthesis, cell membrane function, protein and nucleic acid biosynthesis, and antimetabolites. Many of these targets are now compromised through rapidly spreading antimicrobial resistance and the need to target non-replicating cells (persisters). Recently, an unprecedented medical breakthrough was achieved by the FDA approval of the drug bedaquiline (BDQ, trade name Sirturo) for the treatment of multidrug-resistant tuberculosis disease. BDQ targets the membrane-bound F1Fo-ATP synthase, validating cellular energy generating machinery as a new target space for drug discovery. Recently, BDQ and several other FDA-approved drugs have been demonstrated to be respiratory "uncouplers" disrupting transmembrane electrochemical gradients, in addition to binding to enzyme targets. In this review, we summarize the role of bioenergetic systems in mycobacterial persistence and discuss the multi-targeting nature of uncouplers and the place these molecules may have in future drug development.


Assuntos
Antituberculosos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Metabolismo Energético/efeitos dos fármacos , Mycobacterium tuberculosis/fisiologia , Tuberculose Resistente a Múltiplos Medicamentos/tratamento farmacológico , Antituberculosos/uso terapêutico , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Diarilquinolinas/farmacologia , Diarilquinolinas/uso terapêutico , Desenho de Fármacos , Descoberta de Drogas/métodos , Farmacorresistência Bacteriana Múltipla , Complexo I de Transporte de Elétrons , Humanos , Potenciais da Membrana/efeitos dos fármacos , Terapia de Alvo Molecular/métodos , Mycobacterium tuberculosis/efeitos dos fármacos , ATPases Translocadoras de Prótons/antagonistas & inibidores , Tuberculose Resistente a Múltiplos Medicamentos/microbiologia , Desacopladores/farmacologia , Desacopladores/uso terapêutico
8.
Proc Natl Acad Sci U S A ; 114(28): 7426-7431, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28652330

RESUMO

The recent discovery of small molecules targeting the cytochrome bc1 :aa3 in Mycobacterium tuberculosis triggered interest in the terminal respiratory oxidases for antituberculosis drug development. The mycobacterial cytochrome bc1 :aa3 consists of a menaquinone:cytochrome c reductase (bc1 ) and a cytochrome aa3 -type oxidase. The clinical-stage drug candidate Q203 interferes with the function of the subunit b of the menaquinone:cytochrome c reductase. Despite the affinity of Q203 for the bc1 :aa3 complex, the drug is only bacteriostatic and does not kill drug-tolerant persisters. This raises the possibility that the alternate terminal bd-type oxidase (cytochrome bd oxidase) is capable of maintaining a membrane potential and menaquinol oxidation in the presence of Q203. Here, we show that the electron flow through the cytochrome bd oxidase is sufficient to maintain respiration and ATP synthesis at a level high enough to protect M. tuberculosis from Q203-induced bacterial death. Upon genetic deletion of the cytochrome bd oxidase-encoding genes cydAB, Q203 inhibited mycobacterial respiration completely, became bactericidal, killed drug-tolerant mycobacterial persisters, and rapidly cleared M. tuberculosis infection in vivo. These results indicate a synthetic lethal interaction between the two terminal respiratory oxidases that can be exploited for anti-TB drug development. Our findings should be considered in the clinical development of drugs targeting the cytochrome bc1 :aa3 , as well as for the development of a drug combination targeting oxidative phosphorylation in M. tuberculosis.


Assuntos
Mycobacterium tuberculosis/metabolismo , Oxirredutases/química , Mutações Sintéticas Letais , Trifosfato de Adenosina/química , Animais , Antineoplásicos/farmacologia , Antituberculosos/farmacologia , Redutases do Citocromo/metabolismo , Diarilquinolinas/farmacologia , Transporte de Elétrons , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Deleção de Genes , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Mitocondriais , Infecções por Mycobacterium/microbiologia , Mycobacterium bovis , Mycobacterium tuberculosis/genética , Fosforilação Oxidativa , Oxirredutases/genética , Oxigênio/química , Proteínas de Plantas , Células THP-1
9.
Proc Natl Acad Sci U S A ; 112(33): 10497-502, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26240343

RESUMO

The majority of microbial cells in global soils exist in a spectrum of dormant states. However, the metabolic processes that enable them to survive environmental challenges, such as nutrient-limitation, remain to be elucidated. In this work, we demonstrate that energy-starved cultures of Pyrinomonas methylaliphatogenes, an aerobic heterotrophic acidobacterium isolated from New Zealand volcanic soils, persist by scavenging the picomolar concentrations of H2 distributed throughout the atmosphere. Following the transition from exponential to stationary phase due to glucose limitation, the bacterium up-regulates by fourfold the expression of an eight-gene operon encoding an actinobacteria-type H2-uptake [NiFe]-hydrogenase. Whole-cells of the organism consume atmospheric H2 in a first-order kinetic process. Hydrogen oxidation occurred most rapidly under oxic conditions and was weakly associated with the cell membrane. We propose that atmospheric H2 scavenging serves as a mechanism to sustain the respiratory chain of P. methylaliphatogenes when organic electron donors are scarce. As the first observation of H2 oxidation to our knowledge in the Acidobacteria, the second most dominant soil phylum, this work identifies new sinks in the biogeochemical H2 cycle and suggests that trace gas oxidation may be a general mechanism for microbial persistence.


Assuntos
Acidobacteria/metabolismo , Gases , Microbiologia do Solo , Sequência de Aminoácidos , Atmosfera , Carbono/química , Cromatografia Gasosa , Transporte de Elétrons , Elétrons , Regulação Bacteriana da Expressão Gênica , Hidrogênio/química , Hidrogenase/metabolismo , Cinética , Dados de Sequência Molecular , Oxirredução , Oxigênio/química , Filogenia , Reprodutibilidade dos Testes , Homologia de Sequência de Aminoácidos , Solo/química
10.
Microbiol Spectr ; 2(3)2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25346874

RESUMO

Mycobacteria inhabit a wide range of intracellular and extracellular environments. Many of these environments are highly dynamic and therefore mycobacteria are faced with the constant challenge of redirecting their metabolic activity to be commensurate with either replicative growth or a non-replicative quiescence. A fundamental feature in this adaptation is the ability of mycobacteria to respire, regenerate reducing equivalents and generate ATP via oxidative phosphorylation. Mycobacteria harbor multiple primary dehydrogenases to fuel the electron transport chain and two terminal respiratory oxidases, an aa3 -type cytochrome c oxidase and cytochrome bd-type menaquinol oxidase, are present for dioxygen reduction coupled to the generation of a protonmotive force. Hypoxia leads to the downregulation of key respiratory complexes, but the molecular mechanisms regulating this expression are unknown. Despite being obligate aerobes, mycobacteria have the ability to metabolize in the absence of oxygen and a number of reductases are present to facilitate the turnover of reducing equivalents under these conditions (e.g. nitrate reductase, succinate dehydrogenase/fumarate reductase). Hydrogenases and ferredoxins are also present in the genomes of mycobacteria suggesting the ability of these bacteria to adapt to an anaerobic-type of metabolism in the absence of oxygen. ATP synthesis by the membrane-bound F1FO-ATP synthase is essential for growing and non-growing mycobacteria and the enzyme is able to function over a wide range of protonmotive force values (aerobic to hypoxic). The discovery of lead compounds that target respiration and oxidative phosphorylation in Mycobacterium tuberculosis highlights the importance of this area for the generation of new front line drugs to combat tuberculosis.


Assuntos
Trifosfato de Adenosina/metabolismo , Metabolismo Energético , Mycobacterium tuberculosis/metabolismo , Fosforilação Oxidativa , Aerobiose , Redes e Vias Metabólicas , Oxirredutases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA