Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Chem Biol ; 18(5): 482-491, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35194207

RESUMEN

Molecular profiling of small molecules offers invaluable insights into the function of compounds and allows for hypothesis generation about small-molecule direct targets and secondary effects. However, current profiling methods are limited in either the number of measurable parameters or throughput. Here we developed a multiplexed, unbiased framework that, by linking genetic to drug-induced changes in nearly a thousand metabolites, allows for high-throughput functional annotation of compound libraries in Escherichia coli. First, we generated a reference map of metabolic changes from CRISPR interference (CRISPRi) with 352 genes in all major essential biological processes. Next, on the basis of the comparison of genetic changes with 1,342 drug-induced metabolic changes, we made de novo predictions of compound functionality and revealed antibacterials with unconventional modes of action (MoAs). We show that our framework, combining dynamic gene silencing with metabolomics, can be adapted as a general strategy for comprehensive high-throughput analysis of compound functionality from bacteria to human cell lines.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Escherichia coli , Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Metabolómica/métodos
2.
Proc Natl Acad Sci U S A ; 115(39): 9779-9784, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30143580

RESUMEN

Reactive oxygen species (ROS)-mediated oxidative stress and DNA damage have recently been recognized as contributing to the efficacy of most bactericidal antibiotics, irrespective of their primary macromolecular targets. Inhibitors of targets involved in both combating oxidative stress as well as being required for in vivo survival may exhibit powerful synergistic action. This study demonstrates that the de novo arginine biosynthetic pathway in Mycobacterium tuberculosis (Mtb) is up-regulated in the early response to the oxidative stress-elevating agent isoniazid or vitamin C. Arginine deprivation rapidly sterilizes the Mtb de novo arginine biosynthesis pathway mutants ΔargB and ΔargF without the emergence of suppressor mutants in vitro as well as in vivo. Transcriptomic and flow cytometry studies of arginine-deprived Mtb have indicated accumulation of ROS and extensive DNA damage. Metabolomics studies following arginine deprivation have revealed that these cells experienced depletion of antioxidant thiols and accumulation of the upstream metabolite substrate of ArgB or ArgF enzymes. ΔargB and ΔargF were unable to scavenge host arginine and were quickly cleared from both immunocompetent and immunocompromised mice. In summary, our investigation revealed in vivo essentiality of the de novo arginine biosynthesis pathway for Mtb and a promising drug target space for combating tuberculosis.


Asunto(s)
Arginina/deficiencia , Mycobacterium tuberculosis/metabolismo , Estrés Oxidativo , Antioxidantes/metabolismo , Antituberculosos/farmacología , Arginina/metabolismo , Daño del ADN , Farmacorresistencia Bacteriana , Citometría de Flujo , Perfilación de la Expresión Génica , Técnicas In Vitro , Redes y Vías Metabólicas , Especies Reactivas de Oxígeno/metabolismo , Compuestos de Sulfhidrilo/metabolismo
3.
J Bacteriol ; 202(22)2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32900827

RESUMEN

Phenotypic testing for drug susceptibility of Mycobacterium tuberculosis is critical to basic research and managing the evolving problem of antimicrobial resistance in tuberculosis management, but it remains a specialized technique to which access is severely limited. Here, we report on the development and validation of an improved phage-mediated detection system for M. tuberculosis We incorporated a nanoluciferase (Nluc) reporter gene cassette into the TM4 mycobacteriophage genome to create phage TM4-nluc. We assessed the performance of this reporter phage in the context of cellular limit of detection and drug susceptibility testing using multiple biosafety level 2 drug-sensitive and -resistant auxotrophs as well as virulent M. tuberculosis strains. For both limit of detection and drug susceptibility testing, we developed a standardized method consisting of a 96-hour cell preculture followed by a 72-hour experimental window for M. tuberculosis detection with or without antibiotic exposure. The cellular limit of detection of M. tuberculosis in a 96-well plate batch culture was ≤102 CFU. Consistent with other phenotypic methods for drug susceptibility testing, we found TM4-nluc to be compatible with antibiotics representing multiple classes and mechanisms of action, including inhibition of core central dogma functions, cell wall homeostasis, metabolic inhibitors, compounds currently in clinical trials (SQ109 and Q203), and susceptibility testing for bedaquiline, pretomanid, and linezolid (components of the BPaL regimen for the treatment of multi- and extensively drug-resistant tuberculosis). Using the same method, we accurately identified rifampin-resistant and multidrug-resistant M. tuberculosis strains.IMPORTANCEMycobacterium tuberculosis, the causative agent of tuberculosis disease, remains a public health crisis on a global scale, and development of new interventions and identification of drug resistance are pillars in the World Health Organization End TB Strategy. Leveraging the tractability of the TM4 mycobacteriophage and the sensitivity of the nanoluciferase reporter enzyme, the present work describes an evolution of phage-mediated detection and drug susceptibility testing of M. tuberculosis, adding a valuable tool in drug discovery and basic biology research. With additional validation, this system may play a role as a quantitative phenotypic reference method and complement to genotypic methods for diagnosis and antibiotic susceptibility testing.


Asunto(s)
Antituberculosos/farmacología , Farmacorresistencia Bacteriana , Pruebas de Sensibilidad Microbiana/métodos , Micobacteriófagos/genética , Mycobacterium tuberculosis/efectos de los fármacos , Rifampin/farmacología , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/virología , Tuberculosis Resistente a Múltiples Medicamentos/microbiología , Tuberculosis Pulmonar/microbiología
4.
J Biol Chem ; 294(6): 1936-1943, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30530783

RESUMEN

Energy metabolism has recently gained interest as a target space for antibiotic drug development in mycobacteria. Of particular importance is bedaquiline (Sirturo), which kills mycobacteria by inhibiting the F1F0 ATP synthase. Other components of the electron transport chain such as the NADH dehydrogenases (NDH-2 and NdhA) and the terminal respiratory oxidase bc1:aa3 are also susceptible to chemical inhibition. Because antituberculosis drugs are prescribed as part of combination therapies, the interaction between novel drugs targeting energy metabolism and classical first and second line antibiotics must be considered to maximize treatment efficiency. Here, we show that subinhibitory concentration of drugs targeting the F1F0 ATP synthase and the cytochrome bc1:aa3, as well as energy uncouplers, interfere with the bactericidal potency of isoniazid and moxifloxacin. Isoniazid- and moxifloxacin-induced mycobacterial death correlated with a transient increase in intracellular ATP that was dissipated by co-incubation with energy metabolism inhibitors. Although oxidative phosphorylation is a promising target space for drug development, a better understanding of the link between energy metabolism and antibiotic-induced mycobacterial death is essential to develop potent drug combinations for the treatment of tuberculosis.


Asunto(s)
Antibacterianos/farmacología , Metabolismo Energético/efectos de los fármacos , Mycobacterium/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Diseño de Fármacos , Proteínas del Complejo de Cadena de Transporte de Electrón/antagonistas & inhibidores , Isoniazida/farmacología , Moxifloxacino/farmacología , Mycobacterium/citología , Fosforilación Oxidativa/efectos de los fármacos , ATPasas de Translocación de Protón/antagonistas & inhibidores
5.
Proc Natl Acad Sci U S A ; 114(28): 7426-7431, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28652330

RESUMEN

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.


Asunto(s)
Mycobacterium tuberculosis/metabolismo , Oxidorreductasas/química , Mutaciones Letales Sintéticas , Adenosina Trifosfato/química , Animales , Antineoplásicos/farmacología , Antituberculosos/farmacología , Reductasas del Citocromo/metabolismo , Diarilquinolinas/farmacología , Transporte de Electrón , Complejo IV de Transporte de Electrones/metabolismo , Eliminación de Gen , Humanos , Inflamación , Ratones , Ratones Endogámicos BALB C , Proteínas Mitocondriales , Infecciones por Mycobacterium/microbiología , Mycobacterium bovis , Mycobacterium tuberculosis/genética , Fosforilación Oxidativa , Oxidorreductasas/genética , Oxígeno/química , Proteínas de Plantas , Células THP-1
6.
Mol Syst Biol ; 14(11): e8623, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30397005

RESUMEN

In natural environments, microbes are typically non-dividing and gauge when nutrients permit division. Current models are phenomenological and specific to nutrient-rich, exponentially growing cells, thus cannot predict the first division under limiting nutrient availability. To assess this regime, we supplied starving Escherichia coli with glucose pulses at increasing frequencies. Real-time metabolomics and microfluidic single-cell microscopy revealed unexpected, rapid protein, and nucleic acid synthesis already from minuscule glucose pulses in non-dividing cells. Additionally, the lag time to first division shortened as pulsing frequency increased. We pinpointed division timing and dependence on nutrient frequency to the changing abundance of the division protein FtsZ. A dynamic, mechanistic model quantitatively relates lag time to FtsZ synthesis from nutrient pulses and FtsZ protease-dependent degradation. Lag time changed in model-congruent manners, when we experimentally modulated the synthesis or degradation of FtsZ. Thus, limiting abundance of FtsZ can quantitatively predict timing of the first cell division.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Escherichia coli/metabolismo , Glucosa/metabolismo , División Celular , Escherichia coli/citología , Metabolómica/métodos , Técnicas Analíticas Microfluídicas , Proteolisis , Análisis de la Célula Individual
8.
Proc Natl Acad Sci U S A ; 112(32): 10008-13, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26221021

RESUMEN

Multidrug resistance, strong side effects, and compliance problems in TB chemotherapy mandate new ways to kill Mycobacterium tuberculosis (Mtb). Here we show that deletion of the gene encoding homoserine transacetylase (metA) inactivates methionine and S-adenosylmethionine (SAM) biosynthesis in Mtb and renders this pathogen exquisitely sensitive to killing in immunocompetent or immunocompromised mice, leading to rapid clearance from host tissues. Mtb ΔmetA is unable to proliferate in primary human macrophages, and in vitro starvation leads to extraordinarily rapid killing with no appearance of suppressor mutants. Cell death of Mtb ΔmetA is faster than that of other auxotrophic mutants (i.e., tryptophan, pantothenate, leucine, biotin), suggesting a particularly potent mechanism of killing. Time-course metabolomics showed complete depletion of intracellular methionine and SAM. SAM depletion was consistent with a significant decrease in methylation at the DNA level (measured by single-molecule real-time sequencing) and with the induction of several essential methyltransferases involved in biotin and menaquinone biosynthesis, both of which are vital biological processes and validated targets of antimycobacterial drugs. Mtb ΔmetA could be partially rescued by biotin supplementation, confirming a multitarget cell death mechanism. The work presented here uncovers a previously unidentified vulnerability of Mtb-the incapacity to scavenge intermediates of SAM and methionine biosynthesis from the host. This vulnerability unveils an entirely new drug target space with the promise of rapid killing of the tubercle bacillus by a new mechanism of action.


Asunto(s)
Metionina/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/fisiología , S-Adenosilmetionina/farmacología , Acetiltransferasas/metabolismo , Animales , Línea Celular , Femenino , Humanos , Inmunocompetencia/efectos de los fármacos , Metaboloma/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones SCID , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/patogenicidad , Factores de Tiempo , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Virulencia
9.
J Biol Chem ; 291(13): 7060-9, 2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-26858255

RESUMEN

Mycobacterium tuberculosis (Mtb) displays a high degree of metabolic plasticity to adapt to challenging host environments. Genetic evidence suggests thatMtbrelies mainly on fatty acid catabolism in the host. However,Mtbalso maintains a functional glycolytic pathway and its role in the cellular metabolism ofMtbhas yet to be understood. Pyruvate kinase catalyzes the last and rate-limiting step in glycolysis and theMtbgenome harbors one putative pyruvate kinase (pykA, Rv1617). Here we show thatpykAencodes an active pyruvate kinase that is allosterically activated by glucose 6-phosphate (Glc-6-P) and adenosine monophosphate (AMP). Deletion ofpykApreventsMtbgrowth in the presence of fermentable carbon sources and has a cidal effect in the presence of glucose that correlates with elevated levels of the toxic catabolite methylglyoxal. Growth attenuation was also observed in media containing a combination of short chain fatty acids and glucose and surprisingly, in media containing odd and even chain fatty acids alone. Untargeted high sensitivity metabolomics revealed that inactivation of pyruvate kinase leads to accumulation of phosphoenolpyruvate (P-enolpyruvate), citrate, and aconitate, which was consistent with allosteric inhibition of isocitrate dehydrogenase by P-enolpyruvate. This metabolic block could be relieved by addition of the α-ketoglutarate precursor glutamate. Taken together, our study identifies an essential role of pyruvate kinase in preventing metabolic block during carbon co-catabolism inMtb.


Asunto(s)
Proteínas Bacterianas/metabolismo , Carbono/metabolismo , Glucólisis/genética , Mycobacterium tuberculosis/metabolismo , Piruvato Quinasa/metabolismo , Ácido Aconítico/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/farmacología , Regulación Alostérica , Animales , Proteínas Bacterianas/genética , Ácido Cítrico/metabolismo , Medios de Cultivo/química , Activación Enzimática , Ácidos Grasos Volátiles/farmacología , Femenino , Eliminación de Gen , Expresión Génica , Glucosa/metabolismo , Glucosa-6-Fosfato/metabolismo , Glucosa-6-Fosfato/farmacología , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Glucólisis/efectos de los fármacos , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Ácidos Cetoglutáricos/metabolismo , Ratones , Ratones SCID , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/genética , Fosfoenolpiruvato/metabolismo , Piruvaldehído/metabolismo , Piruvato Quinasa/genética , Análisis de Supervivencia , Tuberculosis/microbiología , Tuberculosis/mortalidad
10.
Proc Natl Acad Sci U S A ; 111(11): 4257-61, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24591586

RESUMEN

In the Earth's lower atmosphere, H2 is maintained at trace concentrations (0.53 ppmv/0.40 nM) and rapidly turned over (lifetime ≤ 2.1 y(-1)). It is thought that soil microbes, likely actinomycetes, serve as the main global sink for tropospheric H2. However, no study has ever unambiguously proven that a hydrogenase can oxidize this trace gas. In this work, we demonstrate, by using genetic dissection and sensitive GC measurements, that the soil actinomycete Mycobacterium smegmatis mc(2)155 constitutively oxidizes subtropospheric concentrations of H2. We show that two membrane-associated, oxygen-dependent [NiFe] hydrogenases mediate this process. Hydrogenase-1 (Hyd1) (MSMEG_2262-2263) is well-adapted to rapidly oxidize H2 at a range of concentrations [Vmax(app) = 12 nmol⋅g⋅dw(-1)⋅min(-1); Km(app) = 180 nM; threshold = 130 pM in the Δhyd23 (Hyd1 only) strain], whereas Hyd2 (MSMEG_2719-2720) catalyzes a slower-acting, higher-affinity process [Vmax(app) = 2.5 nmol⋅g⋅dw(-1)⋅min(-1); Km(app) = 50 nM; threshold = 50 pM in the Δhyd13 (Hyd2 only) strain]. These observations strongly support previous studies that have linked group 5 [NiFe] hydrogenases (e.g., Hyd2) to the oxidation of tropospheric H2 in soil ecosystems. We further reveal that group 2a [NiFe] hydrogenases (e.g., Hyd1) can contribute to this process. Hydrogenase expression and activity increases in carbon-limited cells, suggesting that scavenging of trace H2 helps to sustain dormancy. Distinct physiological roles for Hyd1 and Hyd2 during the adaptation to this condition are proposed. Soil organisms harboring high-affinity hydrogenases may be especially competitive, given that they harness a highly dependable fuel source in otherwise unstable environments.


Asunto(s)
Atmósfera/química , Hidrógeno/metabolismo , Hidrogenasas/metabolismo , Mycobacterium smegmatis/enzimología , Microbiología del Suelo , Cromatografía de Gases , Hidrógeno/análisis , Oxidación-Reducción , Oxígeno/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(31): 11479-84, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25049411

RESUMEN

Oxygen availability is a major factor and evolutionary force determining the metabolic strategy of bacteria colonizing an environmental niche. In the soil, conditions can switch rapidly between oxia and anoxia, forcing soil bacteria to remodel their energy metabolism accordingly. Mycobacterium is a dominant genus in the soil, and all its species are obligate aerobes. Here we show that an obligate aerobe, the soil actinomycete Mycobacterium smegmatis, adopts an anaerobe-type strategy by activating fermentative hydrogen production to adapt to hypoxia. This process is controlled by the two-component system DosR-DosS/DosT, an oxygen and redox sensor that is well conserved in mycobacteria. We show that DosR tightly regulates the two [NiFe]-hydrogenases: Hyd3 (MSMEG_3931-3928) and Hyd2 (MSMEG_2719-2718). Using genetic manipulation and high-sensitivity GC, we demonstrate that Hyd3 facilitates the evolution of H2 when oxygen is depleted. Combined activity of Hyd2 and Hyd3 was necessary to maintain an optimal NAD(+)/NADH ratio and enhanced adaptation to and survival of hypoxia. We demonstrate that fermentatively-produced hydrogen can be recycled when fumarate or oxygen become available, suggesting Mycobacterium smegmatis can switch between fermentation, anaerobic respiration, and aerobic respiration. Hydrogen metabolism enables this obligate aerobe to rapidly meet its energetic needs when switching between microoxic and anoxic conditions and provides a competitive advantage in low oxygen environments.


Asunto(s)
Bacterias Anaerobias/fisiología , Fermentación , Hidrógeno/metabolismo , Mycobacterium smegmatis/fisiología , Microbiología del Suelo , Estrés Fisiológico , Aerobiosis , Anaerobiosis , Bacterias Anaerobias/enzimología , Bacterias Anaerobias/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Electrones , Regulación Bacteriana de la Expresión Génica , Homeostasis , Hidrogenasas , Espacio Intracelular/metabolismo , Viabilidad Microbiana , Modelos Biológicos , Datos de Secuencia Molecular , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/genética , Oxidación-Reducción , Oxígeno/metabolismo , Regulón/genética , Transcripción Genética
12.
PLoS Pathog ; 10(11): e1004510, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25412183

RESUMEN

In chronic infection, Mycobacterium tuberculosis bacilli are thought to enter a metabolic program that provides sufficient energy for maintenance of the protonmotive force, but is insufficient to meet the demands of cellular growth. We sought to understand this metabolic downshift genetically by targeting succinate dehydrogenase, the enzyme which couples the growth processes controlled by the TCA cycle with the energy production resulting from the electron transport chain. M. tuberculosis contains two operons which are predicted to encode succinate dehydrogenase enzymes (sdh-1 and sdh-2); we found that deletion of Sdh1 contributes to an inability to survive long term stationary phase. Stable isotope labeling and mass spectrometry revealed that Sdh1 functions as a succinate dehydrogenase during aerobic growth, and that Sdh2 is dispensable for this catalysis, but partially overlapping activities ensure that the loss of one enzyme can incompletely compensate for loss of the other. Deletion of Sdh1 disturbs the rate of respiration via the mycobacterial electron transport chain, resulting in an increased proportion of reduced electron carrier (menaquinol) which leads to increased oxygen consumption. The loss of respiratory control leads to an inability to recover from stationary phase. We propose a model in which succinate dehydrogenase is a governor of cellular respiration in the adaptation to low oxygen environments.


Asunto(s)
Proteínas Bacterianas/metabolismo , Modelos Biológicos , Mycobacterium tuberculosis/enzimología , Consumo de Oxígeno/fisiología , Succinato Deshidrogenasa/metabolismo , Animales , Proteínas Bacterianas/genética , Ratones , Ratones Noqueados , Viabilidad Microbiana/genética , Mycobacterium tuberculosis/genética , Succinato Deshidrogenasa/genética
13.
Microbiology (Reading) ; 161(Pt 3): 648-61, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25525207

RESUMEN

Mycobacterium smegmatis is a fast-growing, saprophytic, mycobacterial species that contains two cAMP-receptor protein (CRP) homologues designated herein as Crp1 and Crp2. Phylogenetic analysis suggests that Crp1 (Msmeg_0539) is uniquely present in fast-growing environmental mycobacteria, whereas Crp2 (Msmeg_6189) occurs in both fast- and slow-growing species. A crp1 mutant of M. smegmatis was readily obtained, but crp2 could not be deleted, suggesting it was essential for growth. A total of 239 genes were differentially regulated in response to crp1 deletion (loss of function), including genes coding for mycobacterial energy generation, solute transport and catabolism of carbon sources. To assess the role of Crp2 in M. smegmatis, the crp2 gene was overexpressed (gain of function) and transcriptional profiling studies revealed that 58 genes were differentially regulated. Identification of the CRP promoter consensus in M. smegmatis showed that both Crp1 and Crp2 recognized the same consensus sequence (TGTGN8CACA). Comparison of the Crp1- and Crp2-regulated genes revealed distinct but overlapping regulons with 11 genes in common, including those of the succinate dehydrogenase operon (MSMEG_0417-0420, sdh1). Expression of the sdh1 operon was negatively regulated by Crp1 and positively regulated by Crp2. Electrophoretic mobility shift assays with purified Crp1 and Crp2 demonstrated that Crp1 binding to the sdh1 promoter was cAMP-independent whereas Crp2 binding was cAMP-dependent. These data suggest that Crp1 and Crp2 respond to distinct signalling pathways in M. smegmatis to coordinate gene expression in response to carbon and energy supply.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteína Receptora de AMP Cíclico/metabolismo , Mycobacterium smegmatis/crecimiento & desarrollo , Mycobacterium smegmatis/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Carbono/metabolismo , Proteína Receptora de AMP Cíclico/química , Proteína Receptora de AMP Cíclico/genética , Regulación Bacteriana de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium smegmatis/genética , Operón , Regiones Promotoras Genéticas , Alineación de Secuencia
14.
J Antimicrob Chemother ; 70(7): 2028-37, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25754998

RESUMEN

OBJECTIVES: It is not fully understood why inhibiting ATP synthesis in Mycobacterium species leads to death in non-replicating cells. We investigated the bactericidal mode of action of the anti-tubercular F1Fo-ATP synthase inhibitor bedaquiline (Sirturo™) in order to further understand the lethality of ATP synthase inhibition. METHODS: Mycobacterium smegmatis strains were used for all the experiments. Growth and survival during a bedaquiline challenge were performed in multiple media types. A time-course microarray was performed during initial bedaquiline challenge in minimal medium. Oxygen consumption and proton-motive force measurements were performed on whole cells and inverted membrane vesicles, respectively. RESULTS: A killing of 3 log10 cfu/mL was achieved 4-fold more quickly in minimal medium (a glycerol carbon source) versus rich medium (LB with Tween 80) during bedaquiline challenge. Assessing the accelerated killing condition, we identified a transcriptional remodelling of metabolism that was consistent with respiratory dysfunction but inconsistent with ATP depletion. In glycerol-energized cell suspensions, bedaquiline caused an immediate 2.3-fold increase in oxygen consumption. Bedaquiline collapsed the transmembrane pH gradient, but not the membrane potential, in a dose-dependent manner. Both these effects were dependent on binding to the F1Fo-ATP synthase. CONCLUSIONS: Challenge with bedaquiline results in an electroneutral uncoupling of respiration-driven ATP synthesis. This may be a determinant of the bactericidal effects of bedaquiline, while ATP depletion may be a determinant of its delayed onset of killing. We propose that bedaquiline binds to and perturbs the a-c subunit interface of the Fo, leading to futile proton cycling, which is known to be lethal to mycobacteria.


Asunto(s)
Antituberculosos/farmacología , Diarilquinolinas/farmacología , Viabilidad Microbiana/efectos de los fármacos , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium smegmatis/fisiología , Desacopladores/farmacología , Medios de Cultivo/química , Perfilación de la Expresión Génica , Humanos , Análisis por Micromatrices , Técnicas Microbiológicas
15.
Appl Environ Microbiol ; 81(4): 1190-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25501483

RESUMEN

We have known for 40 years that soils can consume the trace amounts of molecular hydrogen (H2) found in the Earth's atmosphere.This process is predicted to be the most significant term in the global hydrogen cycle. However, the organisms and enzymes responsible for this process were only recently identified. Pure culture experiments demonstrated that several species of Actinobacteria, including streptomycetes and mycobacteria, can couple the oxidation of atmospheric H2 to the reduction of ambient O2. A combination of genetic, biochemical, and phenotypic studies suggest that these organisms primarily use this fuel source to sustain electron input into the respiratory chain during energy starvation. This process is mediated by a specialized enzyme, the group 5 [NiFe]-hydrogenase, which is unusual for its high affinity, oxygen insensitivity, and thermostability. Atmospheric hydrogen scavenging is a particularly dependable mode of energy generation, given both the ubiquity of the substrate and the stress tolerance of its catalyst. This minireview summarizes the recent progress in understanding how and why certain organisms scavenge atmospheric H2. In addition, it provides insight into the wider significance of hydrogen scavenging in global H2 cycling and soil microbial ecology.


Asunto(s)
Actinobacteria/metabolismo , Proteínas Bacterianas/metabolismo , Hidrógeno/metabolismo , Hidrogenasas/metabolismo , Actinobacteria/enzimología , Actinobacteria/genética , Microbiología del Aire , Atmósfera/química , Proteínas Bacterianas/genética , Ecosistema , Hidrogenasas/genética
16.
J Bacteriol ; 196(17): 3091-7, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24936051

RESUMEN

Mycobacteria are obligate aerobes and respire using two terminal respiratory oxidases, an aa3-type cytochrome c oxidase and a cytochrome bd-type menaquinol oxidase. Cytochrome bd is encoded by cydAB from the cydABDC gene cluster that is conserved throughout the mycobacterial genus. Here we report that cydAB and cydDC in Mycobacterium smegmatis constitute two separate operons under hypoxic growth conditions. The transcriptional start sites of both operons were mapped, and a series of cydA-lacZ and cydD-lacZ transcriptional reporter fusions were made to identify regulatory promoter elements. A 51-bp region was identified in the cydAB promoter that was required for maximal cydA-lacZ expression in response to hypoxia. A cyclic AMP receptor protein (CRP)-binding site (viz. GTGAN6CCACC) was identified in this region, and mutation of this site to CCCAN6CTTTC abolished cydA-lacZ expression in response to hypoxia. Binding of purified CRP (MSMEG_0539) to the cydAB promoter DNA was analyzed using electrophoretic mobility shift assays. CRP binding was dependent on GTGAN6CCACC and showed cyclic AMP (cAMP) dependency. No CRP site was present in the cydDC promoter, and a 10-bp inverted repeat (CGGTGGTACCGGTACCACCG) was required for maximal cydD-lacZ expression. Taken together, the data indicate that CRP is a direct regulator of cydAB expression in response to hypoxia and that the regulation of cydDC expression is CRP independent and under the control of an unknown regulator.


Asunto(s)
Proteína Receptora de AMP Cíclico/metabolismo , Citocromos/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Mycobacterium smegmatis/enzimología , Oxígeno/metabolismo , Proteína Receptora de AMP Cíclico/genética , Citocromos/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/metabolismo , Transcripción Genética
17.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 4): 968-80, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24699642

RESUMEN

The proline-utilization pathway in Mycobacterium tuberculosis (Mtb) has recently been identified as an important factor in Mtb persistence in vivo, suggesting that this pathway could be a valuable therapeutic target against tuberculosis (TB). In Mtb, two distinct enzymes perform the conversion of proline into glutamate: the first step is the oxidation of proline into Δ(1)-pyrroline-5-carboxylic acid (P5C) by the flavoenzyme proline dehydrogenase (PruB), and the second reaction involves converting the tautomeric form of P5C (glutamate-γ-semialdehyde) into glutamate using the NAD(+)-dependent Δ(1)-pyrroline-5-carboxylic dehydrogenase (PruA). Here, the three-dimensional structures of Mtb-PruA, determined by X-ray crystallography, in the apo state and in complex with NAD(+) are described at 2.5 and 2.1 Šresolution, respectively. The structure reveals a conserved NAD(+)-binding mode, common to other related enzymes. Species-specific conformational differences in the active site, however, linked to changes in the dimer interface, suggest possibilities for selective inhibition of Mtb-PruA despite its reasonably high sequence identity to other PruA enzymes. Using recombinant PruA and PruB, the proline-utilization pathway in Mtb has also been reconstituted in vitro. Functional validation using a novel NMR approach has demonstrated that the PruA and PruB enzymes are together sufficient to convert proline to glutamate, the first such demonstration for monofunctional proline-utilization enzymes.


Asunto(s)
1-Pirrolina-5-Carboxilato Deshidrogenasa/química , Mycobacterium tuberculosis/enzimología , 1-Pirrolina-5-Carboxilato Deshidrogenasa/metabolismo , Cristalografía por Rayos X , Modelos Moleculares , NAD/química , NAD/metabolismo , Resonancia Magnética Nuclear Biomolecular , Prolina/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Homología Estructural de Proteína
18.
Environ Microbiol ; 16(1): 318-30, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24536093

RESUMEN

Mycobacterium smegmatis is an obligate aerobe that harbours three predicted [NiFe] hydrogenases, Hyd1 (MSMEG_2262­2263), Hyd2 (MSMEG_2720-2719) and Hyd3 (MSMEG_3931-3928). We show here that these three enzymes differ in their phylogeny, regulation and catalytic activity. Phylogenetic analysis revealed that Hyd1 groups with hydrogenases that oxidize H2 produced by metabolic processes, and Hyd2 is homologous to a novel group of putative high-affinity hydrogenases. Hyd1 and Hyd2 respond to carbon and oxygen limitation, and, in the case of Hyd1, hydrogen supplementation. Hydrogen consumption measurements confirmed that both enzymes can oxidize hydrogen. In contrast, the phylogenetic analysis and activity measurements of Hyd3 are consistent with the enzyme evolving hydrogen. Hyd3 is controlled by DosR, a regulator that responds to hypoxic conditions. The strict dependence of hydrogen oxidation of Hyd1 and Hyd2 on oxygen suggests that the enzymes are oxygen tolerant and linked to the respiratory chain. This unique combination of hydrogenases allows M. smegmatis to oxidize hydrogen at high (Hyd1) and potentially tropospheric (Hyd2) concentrations, as well as recycle reduced equivalents by evolving hydrogen (Hyd3). The distribution of these hydrogenases throughout numerous soil and marine species of actinomycetes suggests that oxic hydrogen metabolism provides metabolic flexibility in environments with changing nutrient fluxes.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hidrogenasas/metabolismo , Mycobacterium smegmatis/enzimología , Aerobiosis , Proteínas Bacterianas/genética , Hidrógeno/metabolismo , Hidrogenasas/genética , Familia de Multigenes , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/metabolismo , Operón , Oxidación-Reducción , Oxígeno/metabolismo , Filogenia
19.
Nat Microbiol ; 9(6): 1607-1618, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38740932

RESUMEN

Phthiocerol dimycocerosate (PDIM) is an essential virulence lipid of Mycobacterium tuberculosis. In vitro culturing rapidly selects for spontaneous PDIM-negative mutants that have attenuated virulence and increased cell wall permeability, thus impacting the relevance of experimental findings. PDIM loss can also reduce the efficacy of the BCG Pasteur vaccine. Here we show that vancomycin susceptibility can rapidly screen for M. tuberculosis PDIM production. We find that metabolic deficiency of methylmalonyl-CoA impedes the growth of PDIM-producing bacilli, selecting for PDIM-negative variants. Supplementation with odd-chain fatty acids, cholesterol or vitamin B12 restores PDIM-positive bacterial growth. Specifically, we show that propionate supplementation enhances PDIM-producing bacterial growth and selects against PDIM-negative mutants, analogous to in vivo conditions. Our study provides a simple approach to screen for and maintain PDIM production, and reveals how discrepancies between the host and in vitro nutrient environments can attenuate bacterial pathogenicity.


Asunto(s)
Mycobacterium tuberculosis , Propionatos , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crecimiento & desarrollo , Propionatos/farmacología , Propionatos/metabolismo , Virulencia , Lípidos/química , Ésteres del Colesterol/metabolismo , Tuberculosis/microbiología , Tuberculosis/prevención & control , Ácidos Grasos/metabolismo , Vitamina B 12/farmacología , Vitamina B 12/metabolismo , Humanos , Mutación , Factores de Virulencia/metabolismo , Factores de Virulencia/genética , Colesterol/metabolismo , Acilcoenzima A
20.
Cell Chem Biol ; 31(4): 683-698.e7, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38151019

RESUMEN

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.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Succinato Deshidrogenasa/metabolismo , Succinato Deshidrogenasa/farmacología , Antituberculosos/química , Tuberculosis/tratamiento farmacológico , Adenosina Trifosfato , Inhibidores Enzimáticos/farmacología , Succinatos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA