Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
PLoS Pathog ; 16(10): e1009008, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33064782

RESUMEN

RNA thermometers (RNATs) trigger bacterial virulence factor expression in response to the temperature shift on entering a warm-blooded host. At lower temperatures these secondary structures sequester ribosome-binding sites (RBSs) to prevent translation initiation, whereas at elevated temperatures they "melt" allowing translation. Campylobacter jejuni is the leading bacterial cause of human gastroenteritis worldwide yet little is known about how it interacts with the host including host induced gene regulation. Here we demonstrate that an RNAT regulates a C. jejuni gene, Cj1163c or czcD, encoding a member of the Cation Diffusion Facilitator family. The czcD upstream untranslated region contains a predicted stem loop within the mRNA that sequesters the RBS to inhibit translation at temperatures below 37°C. Mutations that disrupt or enhance predicted secondary structure have significant and predictable effects on temperature regulation. We also show that in an RNAT independent manner, CzcD expression is induced by Zn(II). Mutants lacking czcD are hypersensitive to Zn(II) and also over-accumulate Zn(II) relative to wild-type, all consistent with CzcD functioning as a Zn(II) exporter. Importantly, we demonstrate that C. jejuni Zn(II)-tolerance at 32°C, a temperature at which the RNAT limits CzcD production, is increased by RNAT disruption. Finally we show that czcD inactivation attenuates larval killing in a Galleria infection model and that at 32°C disrupting RNAT secondary structure to allow CzcD production can enhance killing. We hypothesise that CzcD regulation by metals and temperature provides a mechanism for C. jejuni to overcome innate immune system-mediated Zn(II) toxicity in warm-blooded animal hosts.


Asunto(s)
Regulación de la Temperatura Corporal/genética , Campylobacter jejuni/genética , Zinc/metabolismo , Bacterias/genética , Infecciones por Campylobacter/genética , Regulación Bacteriana de la Expresión Génica/genética , Conformación de Ácido Nucleico , ARN/genética , ARN/metabolismo , ARN Bacteriano/genética , ARN Mensajero/genética , Temperatura , Virulencia , Factores de Virulencia/metabolismo
2.
Appl Environ Microbiol ; 86(18)2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32680873

RESUMEN

Biomineralization of Cu has been shown to control contaminant dynamics and transport in soils. However, very little is known about the role that subsurface microorganisms may play in the biogeochemical cycling of Cu. In this study, we investigate the bioreduction of Cu(II) by the subsurface metal-reducing bacterium Geobacter sulfurreducens Rapid removal of Cu from solution was observed in cell suspensions of G. sulfurreducens when Cu(II) was supplied, while transmission electron microscopy (TEM) analyses showed the formation of electron-dense nanoparticles associated with the cell surface. Energy-dispersive X-ray spectroscopy (EDX) point analysis and EDX spectrum image maps revealed that the nanoparticles are rich in both Cu and S. This finding was confirmed by X-ray absorption near-edge structure (XANES) and extended X-ray absorption fine structure (EXAFS) analyses, which identified the nanoparticles as Cu2S. Biomineralization of CuxS nanoparticles in soils has been reported to enhance the colloidal transport of a number of contaminants, including Pb, Cd, and Hg. However, formation of these CuxS nanoparticles has only been observed under sulfate-reducing conditions and could not be repeated using isolates of implicated organisms. As G. sulfurreducens is unable to respire sulfate, and no reducible sulfur was supplied to the cells, these data suggest a novel mechanism for the biomineralization of Cu2S under anoxic conditions. The implications of these findings for the biogeochemical cycling of Cu and other metals as well as the green production of Cu catalysts are discussed.IMPORTANCE Dissimilatory metal-reducing bacteria are ubiquitous in soils and aquifers and are known to utilize a wide range of metals as terminal electron acceptors. These transformations play an important role in the biogeochemical cycling of metals in pristine and contaminated environments and can be harnessed for bioremediation and metal bioprocessing purposes. However, relatively little is known about their interactions with Cu. As a trace element that becomes toxic in excess, Cu can adversely affect soil biota and fertility. In addition, biomineralization of Cu nanoparticles has been reported to enhance the mobilization of other toxic metals. Here, we demonstrate that when supplied with acetate under anoxic conditions, the model metal-reducing bacterium Geobacter sulfurreducens can transform soluble Cu(II) to Cu2S nanoparticles. This study provides new insights into Cu biomineralization by microorganisms and suggests that contaminant mobilization enhanced by Cu biomineralization could be facilitated by Geobacter species and related organisms.


Asunto(s)
Biomineralización , Cobre/metabolismo , Geobacter/metabolismo , Nanopartículas del Metal , Sulfuros/metabolismo
3.
Protein Expr Purif ; 174: 105663, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32387341

RESUMEN

SapM from Mycobacterium tuberculosis is a secreted phosphatase critical for pathogen survival inside the host, representing an attractive target for the development of anti-tuberculosis drugs. The main limitation to biochemical and structural studies of SapM has been the lack of a suitable protocol to produce soluble recombinant protein. The aim of the present work was to produce SapM in Escherichia coli in a soluble and catalytically active form. We describe here the construct design, expression and purification of soluble SapM using Sarkosyl as a solubility-enhancing agent and auto-induction media. We demonstrate that solubilisation of the recombinant protein with Sarkosyl, and further purification, yields a catalytically active enzyme with high purity and monodisperse. The identity and molecular weight of the recombinant SapM was confirmed by mass spectrometry analyses, and we provide evidence that SapM behaves as a monomer in solution. Overall, this work lays the foundation for further studies to exploit SapM as a drug target, and provides a protocol for producing active and soluble recombinant enzymes that are hard to solubilise in E. coli.


Asunto(s)
Proteínas Bacterianas , Expresión Génica , Mycobacterium tuberculosis/genética , Monoéster Fosfórico Hidrolasas , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Escherichia coli/enzimología , Escherichia coli/genética , Mycobacterium tuberculosis/enzimología , Monoéster Fosfórico Hidrolasas/biosíntesis , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Solubilidad
6.
Infect Immun ; 85(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28652309

RESUMEN

Copper is an essential yet potentially toxic trace element that is required by all aerobic organisms. A key regulator of copper homeostasis in mammalian cells is the copper-transporting P-type ATPase ATP7A, which mediates copper transport from the cytoplasm into the secretory pathway, as well as copper export across the plasma membrane. Previous studies have shown that ATP7A-dependent copper transport is required for killing phagocytosed Escherichia coli in a cultured macrophage cell line. In this investigation, we expanded on these studies by generating Atp7aLysMcre mice, in which the Atp7a gene was specifically deleted in cells of the myeloid lineage, including macrophages. Primary macrophages isolated from Atp7aLysMcre mice exhibit decreased copper transport into phagosomal compartments and a reduced ability to kill Salmonella enterica serovar Typhimurium compared to that of macrophages isolated from wild-type mice. The Atp7aLysMcre mice were also more susceptible to systemic infection by S Typhimurium than wild-type mice. Deletion of the S Typhimurium copper exporters, CopA and GolT, was found to decrease infection in wild-type mice but not in the Atp7aLysMcre mice. These studies suggest that ATP7A-dependent copper transport into the phagosome mediates host defense against S Typhimurium, which is counteracted by copper export from the bacteria via CopA and GolT. These findings reveal unique and opposing functions for copper transporters of the host and pathogen during infection.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Interacciones Huésped-Patógeno , Macrófagos/enzimología , Salmonella typhimurium/enzimología , Adenosina Trifosfatasas/genética , Animales , Proteínas de Transporte de Catión/genética , Cobre/toxicidad , Femenino , Macrófagos/inmunología , Masculino , Ratones Noqueados , Salmonelosis Animal/microbiología , Salmonelosis Animal/patología , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/fisiología , Virulencia
7.
Infect Immun ; 85(11)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28808161

RESUMEN

Listeria monocytogenes is a foodborne pathogen responsible for a number of life-threatening infections of humans. During an infection, it invades epithelial cells before spreading from the intestine to the cells of the liver and spleen. This requires an ability to adapt to varying oxygen levels. Here, we demonstrate that L. monocytogenes has two terminal oxidases, a cytochrome bd-type (CydAB) and a cytochrome aa 3-type menaquinol (QoxAB) oxidase, and that both are used for respiration under different oxygen tensions. Furthermore, we show that possession of both terminal oxidases is important in infection. In air, the CydAB bd-type oxidase is essential for aerobic respiration and intracellular replication, and cydAB mutants are highly attenuated in mice. In contrast, the QoxAB aa 3-type oxidase is required neither for aerobic respiration in air nor for intracellular growth. However, the qoxAB mutants are attenuated in mice, with a delay in the onset of disease signs and with increased survival time, indicating a role for the QoxAB aa 3-type oxidase in the initial stages of infection. Growth of bacteria under defined oxygen conditions revealed that at 1% (vol/vol), both oxidases are functional, and the presence of either is sufficient for aerobic respiration and intracellular replication. However, at 0.2% (vol/vol), both oxidases are necessary for maximum growth. These findings are consistent with the ability of L. monocytogenes to switch between terminal oxidases under different oxygen conditions, providing exquisite adaptation to different conditions encountered within the infected host.

8.
Microbiology (Reading) ; 168(7)2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35786425
9.
Mol Microbiol ; 87(3): 466-77, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23171030

RESUMEN

Periplasmic Cu,Zn-superoxide dismutases (Cu,Zn-SODs) are implicated in bacterial virulence. It has been proposed that some bacterial P(1B)-type ATPases supply copper to periplasmic cupro-proteins and such transporters have also been implicated in virulence. Here we show that either of two P(1B)-type ATPases, CopA or GolT, is needed to activate a periplasmic Cu,Zn-SOD (SodCII) in Salmonella enterica serovar Typhimurium. A ΔcopA/ΔgolT mutant accumulates inactive Zn-SodCII which can be activated by copper-supplementation in vitro. In contrast, either single ATPase mutant accumulates fully active Cu,Zn-SodCII. A contribution of GolT to copper handling is consistent with its copper-responsive transcription mediated by DNA-binding metal-responsive activator GolS. The requirement for duplicate transcriptional activators CueR and GolS remains unclear since both have similar tight K(Cu). Mutants lacking periplasmic cupro-protein CueP also accumulate inactive Zn-SodCII and while CopA and GolT show functional redundancy, both require CueP to activate SodCII in vivo. Zn-SodCII is also activated in vitro by incubation with Cu-CueP and this coincides with copper transfer as monitored by electron paramagnetic resonance spectroscopy. These experiments establish a role for CueP within the copper supply pathway for Salmonella Cu,Zn-SodCII. Copper binding by CueP in this pathogen may confer protection of the periplasm from copper-mediated damage while sustaining vital cupro-enzyme activity.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Cobre/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Salmonella typhimurium/enzimología , Salmonella typhimurium/metabolismo , Superóxido Dismutasa/metabolismo , Adenosina Trifosfatasas/genética , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Eliminación de Gen , Proteínas de Transporte de Membrana/genética , Salmonella typhimurium/genética
10.
ACS Infect Dis ; 10(1): 170-183, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38085851

RESUMEN

Treatment of Mycobacterium tuberculosis and Mycobacterium avium infections requires multiple drugs for long time periods. Mycobacterium protein-tyrosine-phosphatase B (MptpB) is a key M. tuberculosis virulence factor that subverts host antimicrobial activity to promote intracellular survival. Inhibition of MptpB reduces the infection burden in vivo and offers new opportunities to improve current treatments. Here, we demonstrate that M. avium produces an MptpB orthologue and that the MptpB inhibitor C13 reduces the M. avium infection burden in macrophages. Combining C13 with the antibiotics rifampicin or bedaquiline showed an additive effect, reducing intracellular infection of both M. tuberculosis and M. avium by 50%, compared to monotreatment with antibiotics alone. This additive effect was not observed with pretomanid. Combining C13 with the minor groove-binding compounds S-MGB-362 and S-MGB-363 also reduced the M. tuberculosis intracellular burden. Similar additive effects of C13 and antibiotics were confirmed in vivo using Galleria mellonella infections. We demonstrate that the reduced mycobacterial burden in macrophages observed with C13 treatments is due to the increased trafficking to lysosomes.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Antibacterianos/farmacología , Proteínas Bacterianas , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología , Proteínas Tirosina Fosfatasas , Micobacterias no Tuberculosas
11.
Infect Immun ; 80(1): 14-21, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22025520

RESUMEN

We report here the identification and characterization of two zinc uptake systems, ZurAM and ZinABC, in the intracellular pathogen Listeria monocytogenes. Transcription of both operons was zinc responsive and regulated by the zinc-sensing repressor Zur. Deletion of either zurAM or zinA had no detectable effect on growth in defined media, but a double zurAM zinA mutant was unable to grow in the absence of zinc supplementation. Deletion of zinA had no detectable effect on intracellular growth in HeLa epithelial cells. In contrast, growth of the zurAM mutant was significantly impaired in these cells, indicating the importance of the ZurAM system during intracellular growth. Notably, the deletion of both zinA and zurAM severely attenuated intracellular growth, with the double mutant being defective in actin-based motility and unable to spread from cell to cell. Deletion of either zurAM or zinA had a significant effect on virulence in an oral mouse model, indicating that both zinc uptake systems are important in vivo and establishing the importance of zinc acquisition during infection by L. monocytogenes. The presence of two zinc uptake systems may offer a mechanism by which L. monocytogenes can respond to zinc deficiency within a variety of environments and during different stages of infection, with each system making distinct contributions under different stress conditions.


Asunto(s)
Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Proteínas de Transporte de Membrana/metabolismo , Zinc/metabolismo , Animales , Transporte Biológico , Recuento de Colonia Microbiana , Citoplasma/microbiología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Listeria monocytogenes/genética , Listeriosis/microbiología , Listeriosis/mortalidad , Listeriosis/patología , Proteínas de Transporte de Membrana/genética , Ratones , Operón , Análisis de Supervivencia , Transcripción Genética , Virulencia
12.
Mol Microbiol ; 81(2): 457-72, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21564342

RESUMEN

We have characterized the csoR-copA-copZ copper resistance operon of the important human intracellular pathogen Listeria monocytogenes. Transcription of the operon is specifically induced by copper, and mutants lacking the P1-type ATPase CopA have reduced copper tolerance and over-accumulate copper relative to wild type. The copper-responsive repressor CsoR autoregulates transcription by binding to a single 32 bp site spanning the -10 and -35 elements of the promoter. Copper co-ordination by CsoR derepresses transcription of the operon and alters CsoR:DNA complex assembly as determined by DNase I footprinting and electrophoretic mobility shift assays, with some DNA-binding capacity being retained in the presence of 2 mole equivalents of copper. Analysis of the CsoR copper sensory site demonstrated that substitution of Cys4² with Ala generated a CsoR variant that was unresponsive to copper. Importantly, in the absence of CopZ, copper responsiveness of csoR-copA-copZ expression is substantially increased, implying that CopZ reduces the access of CsoR to copper. Furthermore, CopZ is shown to confer copper resistance in mutants lacking copper-inducible csoR-copA-copZ expression, thus providing protection from the deleterious effects of copper within the cytoplasm.


Asunto(s)
Proteínas Bacterianas/metabolismo , Cobre/metabolismo , Regulación Bacteriana de la Expresión Génica , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Metalochaperonas/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Estructuras Animales/microbiología , Animales , Fusión Artificial Génica , Carga Bacteriana , Proteínas Bacterianas/genética , Huella de ADN , ADN Bacteriano/metabolismo , Modelos Animales de Enfermedad , Ensayo de Cambio de Movilidad Electroforética , Genes Reporteros , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Listeriosis/mortalidad , Listeriosis/patología , Metalochaperonas/genética , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Operón , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Represoras/genética , Enfermedades de los Roedores/microbiología , Enfermedades de los Roedores/mortalidad , Enfermedades de los Roedores/patología , Alineación de Secuencia , Análisis de Supervivencia , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
13.
J Biol Chem ; 285(33): 25259-68, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20534583

RESUMEN

Salmonella enterica sv. typhimurium (S. enterica sv. Typhimurium) has two metal-transporting P(1)-type ATPases whose actions largely overlap with respect to growth in elevated copper. Mutants lacking both ATPases over-accumulate copper relative to wild-type or either single mutant. Such duplication of ATPases is unusual in bacterial copper tolerance. Both ATPases are under the control of MerR family metal-responsive transcriptional activators. Analyses of periplasmic copper complexes identified copper-CueP as one of the predominant metal pools. Expression of cueP was recently shown to be controlled by the same metal-responsive activator as one of the P(1)-type ATPase genes (copA), and copper-CueP is a further atypical feature of copper homeostasis in S. enterica sv. Typhimurium. Elevated copper is detected by a reporter construct driven by the promoter of copA in wild-type S. enterica sv. Typhimurium during infection of macrophages. Double mutants missing both ATPases also show reduced survival inside cultured macrophages. It is hypothesized that elevated copper within macrophages may have selected for specialized copper-resistance systems in pathogenic microorganism such as S. enterica sv. Typhimurium.


Asunto(s)
Proteínas Bacterianas/metabolismo , Cobre/metabolismo , Periplasma/metabolismo , Salmonella typhimurium/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Línea Celular , ATPasas Transportadoras de Cobre , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Mutación , Salmonella typhimurium/genética , Salmonella typhimurium/crecimiento & desarrollo
14.
Sci Rep ; 11(1): 7667, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33828158

RESUMEN

SapM is a secreted virulence factor from Mycobacterium tuberculosis critical for pathogen survival and persistence inside the host. Its full potential as a target for tuberculosis treatment has not yet been exploited because of the lack of potent inhibitors available. By screening over 1500 small molecules, we have identified new potent and selective inhibitors of SapM with an uncompetitive mechanism of inhibition. The best inhibitors share a trihydroxy-benzene moiety essential for activity. Importantly, the inhibitors significantly reduce mycobacterial burden in infected human macrophages at 1 µM, and they are selective with respect to other mycobacterial and human phosphatases. The best inhibitor also reduces intracellular burden of Francisella tularensis, which secretes the virulence factor AcpA, a homologue of SapM, with the same mechanism of catalysis and inhibition. Our findings demonstrate that inhibition of SapM with small molecule inhibitors is efficient in reducing intracellular mycobacterial survival in host macrophages and confirm SapM as a potential therapeutic target. These initial compounds have favourable physico-chemical properties and provide a basis for exploration towards the development of new tuberculosis treatments. The efficacy of a SapM inhibitor in reducing Francisella tularensis intracellular burden suggests the potential for developing broad-spectrum antivirulence agents to treat microbial infections.


Asunto(s)
Mycobacterium tuberculosis/efectos de los fármacos , Factores de Virulencia/antagonistas & inhibidores , Fosfatasa Alcalina/antagonistas & inhibidores , Francisella tularensis/enzimología , Humanos , Terapia Molecular Dirigida , Mycobacterium tuberculosis/patogenicidad , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad , Tuberculosis/tratamiento farmacológico
15.
Biochemistry ; 49(31): 6617-26, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20586430

RESUMEN

Metal homeostasis and resistance in bacteria is maintained by a panel of metal-sensing transcriptional regulators that collectively control transition metal availability and mediate resistance to heavy metal xenobiotics, including As(III), Cd(II), Pb(II), and Hg(II). The ArsR family constitutes a superfamily of metal sensors that appear to conform to the same winged helical, homodimeric fold, that collectively "sense" a wide array of beneficial metal ions and heavy metal pollutants. The genomes of many actinomycetes, including the soil dwelling bacterium Streptomyces coelicolor and the human pathogen Mycobacterium tuberculosis, encode over ten ArsR family regulators, most of unknown function. Here, we present the characterization of a homologue of M. tuberculosis CmtR (CmtR(Mtb)) from S. coelicolor, denoted CmtR(Sc). We show that CmtR(Sc), in contrast to CmtR(Mtb), binds two monomer mol equivalents of Pb(II) or Cd(II) to form two pairs of sulfur-rich coordination complexes per dimer. Metal site 1 conforms exactly to the alpha4C site previously characterized in CmtR(Mtb) while metal site 2 is coordinated by a C-terminal vicinal thiolate pair, Cys110 and Cys111. Biological assays reveal that only Cd(II) and, to a lesser extent, Pb(II) mediate transcriptional derepression in the heterologous host Mycobacterium smegmatis in a way that requires metal site 1. In contrast, mutagenesis of metal site 2 ligands Cys110 or Cys111 significantly reduces Cd(II) responsiveness, with no detectable effect on Pb(II) sensing. The implications of these findings on the ability to predict metal specificity and function from metal-site signatures in the primary structure of ArsR family proteins are discussed.


Asunto(s)
Cadmio/metabolismo , Plomo/metabolismo , Metales Pesados/metabolismo , Streptomyces coelicolor/química , Sitios de Unión/genética , Homeostasis , Mutagénesis Sitio-Dirigida , Mycobacterium tuberculosis/química , Unión Proteica/genética , Streptomyces coelicolor/genética , Streptomyces coelicolor/metabolismo , Activación Transcripcional , Xenobióticos/metabolismo
16.
Nat Prod Rep ; 27(5): 668-80, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20442958

RESUMEN

Detecting deficiency and excess of different metal ions is fundamental for every organism. Our understanding of how metals are detected by bacteria is exceptionally well advanced, and multiple families of cytoplasmic DNA-binding, metal-sensing transcriptional regulators have been characterised(ArsR-SmtB, MerR, CsoR-RcnR, CopY, DtxR, Fur, NikR). Some of the sensors regulate a single gene while others act globally controlling transcription of regulons. They not only modulate the expression of genes directly associated with metal homeostasis, but can also alter metabolism to reduce the cellular demand for metals in short supply. Different representatives of each of the sensor families can regulate gene expression in response to different metals, and the residues that form the sensory metal-binding sites have been defined in a number of these proteins. Indeed, in the case of theArsR-SmtB family, multiple distinct metal-sensing motifs (and one non-metal-sensing motif) have been identified which correlate with the detection of different metals. This review summarises the different families of bacterial metal-sensing transcriptional regulators and discusses current knowledge regarding the mechanisms of metal-regulated gene expression and the structural features of sensory metal-binding sites focusing on the ArsR-SmtB family. In addition, recent progress in understanding the principles governing the ability of the sensors to detect specific metals within a cell and the coordination of the different sensors to control cellular metal levels is discussed.


Asunto(s)
Bacterias , Proteínas Bacterianas/metabolismo , Metales/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Bacterias/química , Bacterias/metabolismo , Modelos Moleculares
17.
Front Immunol ; 11: 486, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32265931

RESUMEN

Infants are more likely to develop lethal disseminated forms of tuberculosis compared with older children and adults. The reasons for this are currently unknown. In this study we test the hypothesis that antimycobacterial function is impaired in infant alveolar macrophages (AMϕs) compared with those of adults. We develop a method of obtaining AMϕs from healthy infants using rigid bronchoscopy and incubate the AMϕs with live virulent Mycobacterium tuberculosis (Mtb). Infant AMϕs are less able to restrict Mtb replication compared with adult AMϕs, despite having similar phagocytic capacity and immunophenotype. RNA-Seq showed that infant AMϕs exhibit lower expression of genes involved in mycobactericidal activity and IFNγ-induction pathways. Infant AMϕs also exhibit lower expression of genes encoding mononuclear cell chemokines such as CXCL9. Our data indicates that failure of AMϕs to contain Mtb and recruit additional mononuclear cells to the site of infection helps to explain the more fulminant course of tuberculosis in early life.


Asunto(s)
Sistema Inmunológico/crecimiento & desarrollo , Lactante , Macrófagos Alveolares/fisiología , Mycobacterium tuberculosis , Adulto , Anciano , Líquido del Lavado Bronquioalveolar , Quimiocinas/biosíntesis , Quimiocinas/genética , Quimiotaxis/genética , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Activación de Macrófagos , Persona de Mediana Edad , Mycobacterium tuberculosis/fisiología , Fagocitosis , ARN Mensajero/biosíntesis , RNA-Seq
18.
J Antimicrob Chemother ; 63(5): 928-36, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19240079

RESUMEN

OBJECTIVES: The secreted Mycobacterium tuberculosis protein tyrosine phosphatase (MptpB) is a virulence factor for M. tuberculosis and contributes to its survival within host macrophages. The aim of this study was to identify potent selective inhibitors of MptpB and to determine the efficacy of these compounds in mycobacterium-infected macrophages. METHODS: The inhibitory effect of a small library of compounds on MptpB was first examined in vitro. The efficacy of these compounds was further examined in mycobacterium-infected macrophages. RESULTS: We have identified a new family of double-site isoxazole-based compounds that are potent selective inhibitors of MptpB. Importantly, the inhibitors substantially reduce mycobacterial survival in infected macrophages. In contrast with current anti-tubercular drugs, these MptpB inhibitors do not have bactericidal action but rather, severely impair mycobacterial growth within macrophages. Docking analysis suggests a double-site binding mechanism of inhibition with the isoxazole head in the active site and a salicylate group in a secondary binding pocket that is a unique structural feature of MptpB. CONCLUSIONS: These results provide the first evidence that inhibition of phosphatases can be exploited against mycobacterial infections. The cell activity of the inhibitors together with the lack of MptpB human orthologues suggests a strong potential for these compounds to be developed as drug candidates against tuberculosis and promises a new therapeutic strategy to tackle clearance and reduce the persistence of M. tuberculosis infection.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Macrófagos/microbiología , Viabilidad Microbiana , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/inmunología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Animales , Línea Celular , Inhibidores Enzimáticos/química , Ratones , Modelos Moleculares , Estructura Molecular , Mycobacterium tuberculosis/efectos de los fármacos , Unión Proteica
19.
Sci Rep ; 9(1): 10315, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31312014

RESUMEN

Mycobacterium tuberculosis (Mtb) SapM is a secreted virulence factor critical for intracellular survival of the pathogen. The role of SapM in phagosome maturation arrest in host macrophages suggests its potential as a drug target to assist in the clearance of tuberculosis infection. However, the mechanism of action of SapM at the molecular level remains unknown. In this study, we provide new insights into the mechanism of catalysis, substrate specificity and inhibition of SapM, and we identify the critical residues for catalysis and substrate binding. Our findings demonstrate that SapM is an atypical monoester alkaline phosphatase, with a serine-based mechanism of catalysis probably metal-dependent. Particularly relevant to SapM function and pathogenesis, is its activity towards PI(4,5)P2 and PI3P, two phosphoinositides that function at the early stages of microbial phagocytosis and phagosome formation. This suggests that SapM may have a pleiotropic role with a wider importance on Mtb infection than initially thought. Finally, we have identified two inhibitors of SapM, L-ascorbic acid and 2-phospho-L-ascorbic, which define two different mechanisms by which the catalytic activity of this phosphatase could be regulated. Critically, we demonstrate that 2-phospho-L-ascorbic reduces mycobacterial survival in macrophage infections, hence confirming the potential of SapM as a therapeutic drug target.


Asunto(s)
Fosfatasa Ácida/genética , Antituberculosos/farmacología , Mycobacterium tuberculosis/patogenicidad , Virulencia/efectos de los fármacos , Fosfatasa Ácida/antagonistas & inhibidores , Fosfatasa Ácida/química , Ácido Ascórbico/análogos & derivados , Ácido Ascórbico/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Catálisis , Dominio Catalítico , Humanos , Concentración 50 Inhibidora , Mycobacterium tuberculosis/efectos de los fármacos , Fosfatidilinositoles/metabolismo , Especificidad por Sustrato , Células THP-1
20.
J Med Chem ; 61(18): 8337-8352, 2018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30153005

RESUMEN

Mycobacterium tuberculosis protein-tyrosine-phosphatase B (MptpB) is a secreted virulence factor that subverts antimicrobial activity in the host. We report here the structure-based design of selective MptpB inhibitors that reduce survival of multidrug-resistant tuberculosis strains in macrophages and enhance killing efficacy by first-line antibiotics. Monotherapy with an orally bioavailable MptpB inhibitor reduces infection burden in acute and chronic guinea pig models and improves the overall pathology. Our findings provide a new paradigm for tuberculosis treatment.


Asunto(s)
Antituberculosos/química , Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Diseño de Fármacos , Macrófagos/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Tuberculosis Resistente a Múltiples Medicamentos/tratamiento farmacológico , Animales , Proteínas Bacterianas/química , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Femenino , Cobayas , Macrófagos/microbiología , Macrófagos/patología , Masculino , Modelos Moleculares , Estructura Molecular , Conformación Proteica , Proteínas Tirosina Fosfatasas/química , Relación Estructura-Actividad , Tuberculosis Resistente a Múltiples Medicamentos/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA