Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 300(3): 105710, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38309504

RESUMEN

The bacterial envelope is an essential compartment involved in metabolism and metabolites transport, virulence, and stress defense. Its roles become more evident when homeostasis is challenged during host-pathogen interactions. In particular, the presence of free radical groups and excess copper in the periplasm causes noxious reactions, such as sulfhydryl group oxidation leading to enzymatic inactivation and protein denaturation. In response to this, canonical and accessory oxidoreductase systems are induced, performing quality control of thiol groups, and therefore contributing to restoring homeostasis and preserving survival under these conditions. Here, we examine recent advances in the characterization of the Dsb-like, Salmonella-specific Scs system. This system includes the ScsC/ScsB pair of Cu+-binding proteins with thiol-oxidoreductase activity, an alternative ScsB-partner, the membrane-linked ScsD, and a likely associated protein, ScsA, with a role in peroxide resistance. We discuss the acquisition of the scsABCD locus and its integration into a global regulatory pathway directing envelope response to Cu stress during the evolution of pathogens that also harbor the canonical Dsb systems. The evidence suggests that the canonical Dsb systems cannot satisfy the extra demands that the host-pathogen interface imposes to preserve functional thiol groups. This resulted in the acquisition of the Scs system by Salmonella. We propose that the ScsABCD complex evolved to connect Cu and redox stress responses in this pathogen as well as in other bacterial pathogens.


Asunto(s)
Proteínas Bacterianas , Proteínas Portadoras , Cobre , Salmonella , Proteínas Bacterianas/metabolismo , Cobre/metabolismo , Homeostasis , Oxidación-Reducción , Oxidorreductasas/metabolismo , Salmonella/metabolismo , Compuestos de Sulfhidrilo , Proteínas Portadoras/metabolismo
2.
J Biol Inorg Chem ; 27(6): 509-528, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35802193

RESUMEN

Copper is essential in cells as a cofactor for key redox enzymes. Bacteria have acquired molecular components that sense, uptake, distribute, and expel copper ensuring that cuproenzymes are metallated and steady-state metal levels are maintained. Toward preventing deleterious reactions, proteins bind copper ions with high affinities and transfer the metal via ligand exchange, warranting that copper ions are always complexed. Consequently, the directional copper distribution within cell compartments and across cell membranes requires specific dynamic interactions and metal exchange between cognate holo-apo protein partners. These metal exchange reactions are determined by thermodynamic and kinetics parameters and influenced by mass action. Then, copper distribution can be conceptualized as a molecular system of singular interacting elements that maintain a physiological copper homeostasis. This review focuses on the impact of copper high-affinity binding and exchange reactions on the homeostatic mechanisms, the conceptual models to describe the cell as a homeostatic system, the various molecule functions that contribute to copper homeostasis, and the alternative system architectures responsible for copper homeostasis in model bacteria.


Asunto(s)
Quelantes , Cobre , Bacterias , Quelantes/química , Cobre/química , Homeostasis , Cinética , Termodinámica
3.
Trends Microbiol ; 29(5): 384-387, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33516594

RESUMEN

Copper (Cu) plays a key role at the host-pathogen interface as both an essential element and a toxic element. Intracellular strains of pathogenic Salmonella have acquired the periplasmic Cu chaperone, CueP, and the thiol oxidoreductases complex Scs, while losing the ancestral Cu-detoxification Cus system. Coregulation of these species-specific factors link Cu with redox stress and allows Salmonella to counteract Cu toxicity during infection.


Asunto(s)
Membrana Celular/metabolismo , Cobre/metabolismo , Interacciones Huésped-Patógeno , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Animales , Proteínas Bacterianas/metabolismo , Humanos , Oxidación-Reducción , Periplasma/metabolismo , Virulencia
4.
mSphere ; 5(6)2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33361129

RESUMEN

Two-component systems control periplasmic Cu+ homeostasis in Gram-negative bacteria. In characterized systems such as Escherichia coli CusRS, upon Cu+ binding to the periplasmic sensing region of CusS, a cytoplasmic phosphotransfer domain of the sensor phosphorylates the response regulator CusR. This drives the expression of efflux transporters, chaperones, and redox enzymes to ameliorate metal toxic effects. Here, we show that the Pseudomonas aeruginosa two-component sensor histidine kinase CopS exhibits a Cu-dependent phosphatase activity that maintains CopR in a nonphosphorylated state when the periplasmic Cu levels are below the activation threshold of CopS. Upon Cu+ binding to the sensor, the phosphatase activity is blocked and the phosphorylated CopR activates transcription of the CopRS regulon. Supporting the model, mutagenesis experiments revealed that the ΔcopS strain exhibits maximal expression of the CopRS regulon, lower intracellular Cu+ levels, and increased Cu tolerance compared to wild-type cells. The invariant phosphoacceptor residue His235 of CopS was not required for the phosphatase activity itself but was necessary for its Cu dependency. To sense the metal, the periplasmic domain of CopS binds two Cu+ ions at its dimeric interface. Homology modeling of CopS based on CusS structure (four Ag+ binding sites) clearly supports the different binding stoichiometries in the two systems. Interestingly, CopS binds Cu+/2+ with 3 × 10-14 M affinity, pointing to the absence of free (hydrated) Cu+/2+ in the periplasm.IMPORTANCE Copper is a micronutrient required as cofactor in redox enzymes. When free, copper is toxic, mismetallating proteins and generating damaging free radicals. Consequently, copper overload is a strategy that eukaryotic cells use to combat pathogens. Bacteria have developed copper-sensing transcription factors to control copper homeostasis. The cell envelope is the first compartment that has to cope with copper stress. Dedicated two-component systems control the periplasmic response to metal overload. This paper shows that the sensor kinase of the copper-sensing two-component system present in Pseudomonadales exhibits a signal-dependent phosphatase activity controlling the activation of its cognate response regulator, distinct from previously described periplasmic Cu sensors. Importantly, the data show that the system is activated by copper levels compatible with the absence of free copper in the cell periplasm. These observations emphasize the diversity of molecular mechanisms that have evolved in bacteria to manage the copper cellular distribution.


Asunto(s)
Cobre/química , Cobre/metabolismo , Escherichia coli/enzimología , Periplasma/enzimología , Pseudomonas aeruginosa/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Transporte Biológico/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Escherichia coli/genética , Histidina Quinasa/metabolismo , Homeostasis , Periplasma/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Pseudomonas aeruginosa/genética , Regulón/genética
5.
New Phytol ; 228(1): 194-209, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32367515

RESUMEN

Iron is an essential cofactor for symbiotic nitrogen fixation, required by many of the enzymes involved, including signal transduction proteins, O2 homeostasis systems, and nitrogenase itself. Consequently, host plants have developed a transport network to deliver essential iron to nitrogen-fixing nodule cells. Ferroportin family members in model legume Medicago truncatula were identified and their expression was determined. Yeast complementation assays, immunolocalization, characterization of a tnt1 insertional mutant line, and synchrotron-based X-ray fluorescence assays were carried out in the nodule-specific M. truncatula ferroportin Medicago truncatula nodule-specific gene Ferroportin2 (MtFPN2) is an iron-efflux protein. MtFPN2 is located in intracellular membranes in the nodule vasculature and in inner nodule tissues, as well as in the symbiosome membranes in the interzone and early-fixation zone of the nodules. Loss-of-function of MtFPN2 alters iron distribution and speciation in nodules, reducing nitrogenase activity and biomass production. Using promoters with different tissular activity to drive MtFPN2 expression in MtFPN2 mutants, we determined that expression in the inner nodule tissues is sufficient to restore the phenotype, while confining MtFPN2 expression to the vasculature did not improve the mutant phenotype. These data indicate that MtFPN2 plays a primary role in iron delivery to nitrogen-fixing bacteroids in M. truncatula nodules.


Asunto(s)
Medicago truncatula , Regulación de la Expresión Génica de las Plantas , Hierro/metabolismo , Medicago truncatula/genética , Medicago truncatula/metabolismo , Fijación del Nitrógeno , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Nódulos de las Raíces de las Plantas/genética , Nódulos de las Raíces de las Plantas/metabolismo , Simbiosis
6.
J Biol Chem ; 294(13): 4934-4945, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30718281

RESUMEN

Copper homeostasis in pathogenic bacteria is critical for cuproprotein assembly and virulence. However, in vivo biochemical analyses of these processes are challenging, which has prevented defining and quantifying the homeostatic interplay between Cu+-sensing transcriptional regulators, chaperones, and sequestering molecules. The cytoplasm of Pseudomonas aeruginosa contains a Cu+-sensing transcriptional regulator, CueR, and two homologous metal chaperones, CopZ1 and CopZ2, forming a unique system for studying Cu+ homeostasis. We found here that both chaperones exchange Cu+, albeit at a slow rate, reaching equilibrium after 3 h, a time much longer than P. aeruginosa duplication time. Therefore, they appeared as two separate cellular Cu+ pools. Although both chaperones transferred Cu+ to CueR in vitro, experiments in vivo indicated that CopZ1 metallates CueR, eliciting the translation of Cu+ efflux transporters involved in metal tolerance. Although this observation was consistent with the relative Cu+ affinities of the three proteins (CopZ1 < CueR < CopZ2), in vitro and in silico analyses also indicated a stronger interaction between CopZ1 and CueR that was independent of Cu+ In contrast, CopZ2 function was defined by its distinctly high abundance during Cu2+ stress. Under resting conditions, CopZ2 remained largely in its apo form. Metal stress quickly induced CopZ2 expression, and its holo form predominated, reaching levels commensurate with the cytoplasmic Cu+ levels. In summary, these results show that CopZ1 acts as chaperone delivering Cu+ to the CueR sensor, whereas CopZ2 functions as a fast-response Cu+-sequestering storage protein. We propose that equivalent proteins likely play similar roles in most bacterial systems.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Cobre/metabolismo , Proteínas de Unión al ADN/biosíntesis , Regulación Bacteriana de la Expresión Génica , Homeostasis , Chaperonas Moleculares/biosíntesis , Pseudomonas aeruginosa/metabolismo , Proteínas Bacterianas/genética , Proteínas de Unión al ADN/genética , Chaperonas Moleculares/genética , Pseudomonas aeruginosa/genética
7.
Front Plant Sci ; 10: 1780, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32082345

RESUMEN

Symbiotic nitrogen fixation carried out by the interaction between legumes and diazotrophic bacteria known as rhizobia requires relatively large levels of transition metals. These elements are cofactors of many key enzymes involved in this process. Metallic micronutrients are obtained from soil by the roots and directed to sink organs by the vasculature, in a process mediated by a number of metal transporters and small organic molecules that facilitate metal delivery in the plant fluids. Among the later, nicotianamine is one of the most important. Synthesized by nicotianamine synthases (NAS), this molecule forms metal complexes participating in intracellular metal homeostasis and long-distance metal trafficking. Here we characterized the NAS2 gene from model legume Medicago truncatula. MtNAS2 is located in the root vasculature and in all nodule tissues in the infection and fixation zones. Symbiotic nitrogen fixation requires of MtNAS2 function, as indicated by the loss of nitrogenase activity in the insertional mutant nas2-1, phenotype reverted by reintroduction of a wild-type copy of MtNAS2. This would result from the altered iron distribution in nas2-1 nodules shown with X-ray fluorescence. Moreover, iron speciation is also affected in these nodules. These data suggest a role of nicotianamine in iron delivery for symbiotic nitrogen fixation.

8.
Mol Microbiol ; 110(3): 357-369, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30047562

RESUMEN

Biological systems require precise copper homeostasis enabling metallation of cuproproteins while preventing metal toxicity. In bacteria, sensing, transport, and storage molecules act in coordination to fulfill these roles. However, there is not yet a kinetic schema explaining the system integration. Here, we report a model emerging from experimental and computational approaches that describes the dynamics of copper distribution in Pseudomonas aeruginosa. Based on copper uptake experiments, a minimal kinetic model describes well the copper distribution in the wild-type bacteria but is unable to explain the behavior of the mutant strain lacking CopA1, a key Cu+ efflux ATPase. The model was expanded through an iterative hypothesis-driven approach, arriving to a mechanism that considers the induction of compartmental pools and the parallel function of CopA and Cus efflux systems. Model simulations support the presence of a periplasmic copper storage with a crucial role under dyshomeostasis conditions in P. aeruginosa. Importantly, the model predicts not only the interplay of periplasmic and cytoplasmic pools but also the existence of a threshold in the concentration of external copper beyond which cells lose their ability to control copper levels.


Asunto(s)
Cobre/metabolismo , Homeostasis , Periplasma/metabolismo , Pseudomonas aeruginosa/metabolismo , Oligoelementos/metabolismo , Transporte Biológico , Simulación por Computador , ATPasas Transportadoras de Cobre/genética , ATPasas Transportadoras de Cobre/metabolismo , Citoplasma/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Modelos Biológicos
9.
J Biol Chem ; 292(38): 15691-15704, 2017 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-28760827

RESUMEN

Bacterial copper (Cu+) homeostasis enables both precise metallation of diverse cuproproteins and control of variable metal levels. To this end, protein networks mobilize Cu+ to cellular targets with remarkable specificity. However, the understanding of these processes is rather fragmented. Here, we use genome-wide transcriptomic analysis by RNA-Seq to characterize the response of Pseudomonas aeruginosa to external 0.5 mm CuSO4, a condition that did not generate pleiotropic effects. Pre-steady-state (5-min) and steady-state (2-h) Cu+ fluxes resulted in distinct transcriptome landscapes. Cells quickly responded to Cu2+ stress by slowing down metabolism. This was restored once steady state was reached. Specific Cu+ homeostasis genes were strongly regulated in both conditions. Our system-wide analysis revealed induction of three Cu+ efflux systems (a P1B-ATPase, a porin, and a resistance-nodulation-division (RND) system) and of a putative Cu+-binding periplasmic chaperone and the unusual presence of two cytoplasmic CopZ proteins. Both CopZ chaperones could bind Cu+ with high affinity. Importantly, novel transmembrane transporters probably mediating Cu+ influx were among those largely repressed upon Cu+ stress. Compartmental Cu+ levels appear independently controlled; the cytoplasmic Cu+ sensor CueR controls cytoplasmic chaperones and plasma membrane transporters, whereas CopR/S responds to periplasmic Cu+ Analysis of ΔcopR and ΔcueR mutant strains revealed a CopR regulon composed of genes involved in periplasmic Cu+ homeostasis and its putative DNA recognition sequence. In conclusion, our study establishes a system-wide model of a network of sensors/regulators, soluble chaperones, and influx/efflux transporters that control the Cu+ levels in P. aeruginosa compartments.


Asunto(s)
Cobre/metabolismo , Homeostasis , Pseudomonas aeruginosa/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Transporte Biológico/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Sulfato de Cobre/farmacología , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Genómica , Homeostasis/efectos de los fármacos , Modelos Moleculares , Conformación Proteica , Pseudomonas aeruginosa/citología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética , Regulón/genética
10.
Metallomics ; 8(9): 906-14, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27465346

RESUMEN

The early discovery of the human Cu(+)-ATPases and their link to Menkes and Wilson's diseases brought attention to the unique role of these transporters in copper homeostasis. The characterization of bacterial Cu(+)-ATPases has significantly furthered our understanding of the structure, selectivity and transport mechanism of these enzymes, as well as their interplay with other elements of Cu(+) distribution networks. This review focuses on the structural-functional insights that have emerged from studies of bacterial Cu(+)-ATPases at the molecular level and how these observations have contributed to drawing up a comprehensive picture of cellular copper homeostasis.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Bacterias/enzimología , ATPasas Transportadoras de Cobre/química , ATPasas Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Transporte Biológico , Modelos Moleculares , Estructura Molecular , Conformación Proteica , Relación Estructura-Actividad
11.
Mol Microbiol ; 100(6): 1066-79, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26946370

RESUMEN

Listeria monocytogenes FrvA (Lmo0641) is critical for virulence in the mouse model and is an ortholog of the Bacillus subtilis Fur- and PerR-regulated Fe(II) efflux P1B4 -type ATPase PfeT. Previously, FrvA was suggested to protect against heme toxicity. Here, we demonstrate that an frvA mutant is sensitive to iron intoxication, but not to other metals. Expression of frvA is induced by high iron and this induction requires Fur. FrvA functions in vitro as a divalent cation specific ATPase most strongly activated by ferrous iron. When expressed in B. subtilis, FrvA increases resistance to iron both in wild-type and in a pfeT null strain. FrvA is a high affinity Fe(II) exporter and its induction imposes severe iron limitation in B. subtilis resulting in derepression of both Fur- and PerR-regulated genes. FrvA also recognizes Co(II) and Zn(II) as substrates and can complement B. subtilis strains defective in the endogenous export systems for these cations. Building on these results, we conclude that FrvA functions in the efflux of Fe(II), and not heme during listerial infection.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Compuestos Ferrosos/metabolismo , Listeria monocytogenes/metabolismo , Factores de Virulencia/metabolismo , Adenosina Trifosfatasas/genética , Bacillus subtilis/enzimología , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Compuestos Ferrosos/toxicidad , Regulación Bacteriana de la Expresión Génica , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/enzimología , Listeria monocytogenes/genética , Mutación , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Proteínas Represoras/metabolismo , Virulencia , Factores de Virulencia/genética
12.
J Biol Chem ; 291(22): 11529-39, 2016 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-27022029

RESUMEN

Little is known about iron efflux transporters within bacterial systems. Recently, the participation of Bacillus subtilis PfeT, a P1B4-ATPase, in cytoplasmic Fe(2+) efflux has been proposed. We report here the distinct roles of mycobacterial P1B4-ATPases in the homeostasis of Co(2+) and Fe(2+) Mutation of Mycobacterium smegmatis ctpJ affects the homeostasis of both ions. Alternatively, an M. tuberculosis ctpJ mutant is more sensitive to Co(2+) than Fe(2+), whereas mutation of the homologous M. tuberculosis ctpD leads to Fe(2+) sensitivity but no alterations in Co(2+) homeostasis. In vitro, the three enzymes are activated by both Fe(2+) and Co(2+) and bind 1 eq of either ion at their transport site. However, equilibrium binding affinities and activity kinetics show that M. tuberculosis CtpD has higher affinity for Fe(2+) and twice the Fe(2+)-stimulated activity than the CtpJs. These parameters are paralleled by a lower activation and affinity for Co(2+) Analysis of Fe(2+) and Co(2+) binding to CtpD by x-ray absorption spectroscopy shows that both ions are five- to six-coordinate, constrained within oxygen/nitrogen environments with similar geometries. Mutagenesis studies suggest the involvement of invariant Ser, His, and Glu residues in metal coordination. Interestingly, replacement of the conserved Cys at the metal binding pocket leads to a large reduction in Fe(2+) but not Co(2+) binding affinity. We propose that CtpJ ATPases participate in the control of steady state Fe(2+) levels. CtpD, required for M. tuberculosis virulence, is a high affinity Fe(2+) transporter involved in the rapid response to iron dyshomeostasis generated upon redox stress.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/metabolismo , Hierro/metabolismo , Metales/metabolismo , Mycobacterium tuberculosis/enzimología , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Hierro/química , Metales/química , Mutación/genética , Especificidad por Sustrato , Tuberculosis/metabolismo , Tuberculosis/microbiología , Virulencia , Espectroscopía de Absorción de Rayos X
13.
Methods Mol Biol ; 1377: 267-77, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26695039

RESUMEN

P1B-type ATPases transport transition metals across biological membranes. The chemical characteristics of these substrates, as well as, physiological requirements have contributed to the evolution of high metal binding affinities (fM) in these enzymes. Metal binding determinations are consequently facilitated by the stable metal-protein interaction, while affinity measurements require careful analysis of metal levels. In the cell, transition metals are associated with chaperone proteins. Metals reach the ATPase transport sites following specific protein-protein interactions and ligand exchange enabling the metal transfer from the chaperone to the transporter. Here, we describe methods for analyzing the binding of Cu(+) to Cu(+)-ATPases, as well as the approach to monitor Cu(+) transfer from soluble Cu(+)-chaperones donors to and from membrane Cu(+)-ATPases.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cobre/metabolismo , Adenosina Trifosfatasas/genética , Unión Competitiva , Transporte Biológico , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Níquel/química , Ácido Nitrilotriacético/química , Unión Proteica , Quinolinas/química , Solubilidad
14.
Mol Microbiol ; 98(4): 787-803, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26261021

RESUMEN

Iron is an essential element for nearly all cells and limited iron availability often restricts growth. However, excess iron can also be deleterious, particularly when cells expressing high affinity iron uptake systems transition to iron rich environments. Bacillus subtilis expresses numerous iron importers, but iron efflux has not been reported. Here, we describe the B. subtilis PfeT protein (formerly YkvW/ZosA) as a P1B4 -type ATPase in the PerR regulon that serves as an Fe(II) efflux pump and protects cells against iron intoxication. Iron and manganese homeostasis in B. subtilis are closely intertwined: a pfeT mutant is iron sensitive, and this sensitivity can be suppressed by low levels of Mn(II). Conversely, a pfeT mutant is more resistant to Mn(II) overload. In vitro, the PfeT ATPase is activated by both Fe(II) and Co(II), although only Fe(II) efflux is physiologically relevant in wild-type cells, and null mutants accumulate elevated levels of intracellular iron. Genetic studies indicate that PfeT together with the ferric uptake repressor (Fur) cooperate to prevent iron intoxication, with iron sequestration by the MrgA mini-ferritin playing a secondary role. Protection against iron toxicity may also be a key role for related P1B4 -type ATPases previously implicated in bacterial pathogenesis.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Bacillus subtilis/enzimología , Proteínas Bacterianas/metabolismo , Hierro/toxicidad , Adenosina Trifosfatasas/genética , Bacillus subtilis/genética , Bacillus subtilis/crecimiento & desarrollo , Proteínas Bacterianas/genética , Secuencia de Bases , Regulación Bacteriana de la Expresión Génica , Hierro/metabolismo , Manganeso/metabolismo , Mutación , Regulón , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
15.
Antimicrob Agents Chemother ; 59(4): 2256-64, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25645825

RESUMEN

Efficient iron acquisition is crucial for the pathogenesis of Mycobacterium tuberculosis. Mycobacterial iron uptake and metabolism are therefore attractive targets for antitubercular drug development. Resistance mutations against a novel pyrazolopyrimidinone compound (PZP) that is active against M. tuberculosis have been identified within the gene cluster encoding the ESX-3 type VII secretion system. ESX-3 is required for mycobacterial iron acquisition through the mycobactin siderophore pathway, which could indicate that PZP restricts mycobacterial growth by targeting ESX-3 and thus iron uptake. Surprisingly, we show that ESX-3 is not the cellular target of the compound. We demonstrate that PZP indeed targets iron metabolism; however, we found that instead of inhibiting uptake of iron, PZP acts as an iron chelator, and we present evidence that the compound restricts mycobacterial growth by chelating intrabacterial iron. Thus, we have unraveled the unexpected mechanism of a novel antimycobacterial compound.


Asunto(s)
Antibacterianos/farmacología , Quelantes del Hierro/farmacología , Mycobacterium smegmatis/efectos de los fármacos , Pirazoles/farmacología , Pirimidinonas/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Ferrozina/metabolismo , Hierro/metabolismo , Pruebas de Sensibilidad Microbiana , Mycobacterium smegmatis/genética , Oxazoles/metabolismo , Pirazoles/síntesis química , Pirimidinonas/síntesis química , ARN Bacteriano/metabolismo , Sideróforos/metabolismo
16.
Proc Natl Acad Sci U S A ; 111(50): E5480-7, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25468978

RESUMEN

Metallochaperones traffic copper (Cu(+)) from its point of entry at the plasma membrane to its destination. In plants, one destination is the chloroplast, which houses plastocyanin, a Cu-dependent electron transfer protein involved in photosynthesis. We present a previously unidentified Cu(+) chaperone that evolved early in the plant lineage by an alternative-splicing event of the pre-mRNA encoding the chloroplast P-type ATPase in Arabidopsis 1 (PAA1). In several land plants, recent duplication events created a separate chaperone-encoding gene coincident with loss of alternative splicing. The plant-specific Cu(+) chaperone delivers Cu(+) with specificity for PAA1, which is flipped in the envelope relative to prototypical bacterial ATPases, compatible with a role in Cu(+) import into the stroma and consistent with the canonical catalytic mechanism of these enzymes. The ubiquity of the chaperone suggests conservation of this Cu(+)-delivery mechanism and provides a unique snapshot into the evolution of a Cu(+) distribution pathway. We also provide evidence for an interaction between PAA2, the Cu(+)-ATPase in thylakoids, and the Cu(+)-chaperone for Cu/Zn superoxide dismutase (CCS), uncovering a Cu(+) network that has evolved to fine-tune Cu(+) distribution.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Cloroplastos/fisiología , Cobre/metabolismo , Evolución Molecular , Homeostasis/fisiología , Metalochaperonas/genética , Proteínas de Arabidopsis/metabolismo , Chlamydomonas reinhardtii/genética , ATPasas de Translocación de Protón de Cloroplastos/metabolismo , Cloroplastos/metabolismo , Clonación Molecular , Biología Computacional , Immunoblotting , Metalochaperonas/metabolismo , Superóxido Dismutasa/metabolismo
17.
J Biol Chem ; 289(30): 20492-501, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24917681

RESUMEN

Cellular copper homeostasis requires transmembrane transport and compartmental trafficking while maintaining the cell essentially free of uncomplexed Cu(2+/+). In bacteria, soluble cytoplasmic and periplasmic chaperones bind and deliver Cu(+) to target transporters or metalloenzymes. Transmembrane Cu(+)-ATPases couple the hydrolysis of ATP to the efflux of cytoplasmic Cu(+). Cytosolic Cu(+) chaperones (CopZ) interact with a structural platform in Cu(+)-ATPases (CopA) and deliver copper into the ion permeation path. CusF is a periplasmic Cu(+) chaperone that supplies Cu(+) to the CusCBA system for efflux to the extracellular milieu. In this report, using Escherichia coli CopA and CusF, direct Cu(+) transfer from the ATPase to the periplasmic chaperone was observed. This required the specific interaction of the Cu(+)-bound form of CopA with apo-CusF for subsequent metal transfer upon ATP hydrolysis. As expected, the reverse Cu(+) transfer from CusF to CopA was not observed. Mutation of CopA extracellular loops or the electropositive surface of CusF led to a decrease in Cu(+) transfer efficiency. On the other hand, mutation of Met and Glu residues proposed to be part of the metal exit site in the ATPase yielded enzymes with lower turnover rates, although Cu(+) transfer was minimally affected. These results show how soluble chaperones obtain Cu(+) from transmembrane transporters. Furthermore, by explaining the movement of Cu(+) from the cytoplasmic pool to the extracellular milieu, these data support a mechanism by which cytoplasmic Cu(+) can be precisely directed to periplasmic targets via specific transporter-chaperone interactions.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Periplasmáticas/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas Transportadoras de Cobre , ATPasas Transportadoras de Cobre , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Transporte Iónico/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/genética , Mutación , Proteínas Periplasmáticas/genética , Estructura Secundaria de Proteína
18.
Microbiology (Reading) ; 160(Pt 6): 1237-1251, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24662147

RESUMEN

Copper is an important element in host-microbe interactions, acting both as a catalyst in enzymes and as a potential toxin. Cu(+)-ATPases drive cytoplasmic Cu(+) efflux and protect bacteria against metal overload. Many pathogenic and symbiotic bacteria contain multiple Cu(+)-ATPase genes within particular genetic environments, suggesting alternative roles for each resulting protein. This hypothesis was tested by characterizing five homologous Cu(+)-ATPases present in the symbiotic organism Sinorhizobium meliloti. Mutation of each gene led to different phenotypes and abnormal nodule development in the alfalfa host. Distinct responses were detected in free-living S. meliloti mutant strains exposed to metal and redox stresses. Differential gene expression was detected under Cu(+), oxygen or nitrosative stress. These observations suggest that CopA1a maintains the cytoplasmic Cu(+) quota and its expression is controlled by Cu(+) levels. CopA1b is also regulated by Cu(+) concentrations and is required during symbiosis for bacteroid maturation. CopA2-like proteins, FixI1 and FixI2, are necessary for the assembly of two different cytochrome c oxidases at different stages of bacterial life. CopA3 is a phylogenetically distinct Cu(+)-ATPase that does not contribute to Cu(+) tolerance. It is regulated by redox stress and required during symbiosis. We postulated a model where non-redundant homologous Cu(+)-ATPases, operating under distinct regulation, transport Cu(+) to different target proteins.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Sinorhizobium meliloti/enzimología , Adenosina Trifosfatasas/genética , Proteínas de Transporte de Catión/genética , ATPasas Transportadoras de Cobre , Técnicas de Inactivación de Genes , Medicago sativa/microbiología , Metales/metabolismo , Metales/toxicidad , Compuestos Nitrosos/metabolismo , Compuestos Nitrosos/toxicidad , Oxidantes/metabolismo , Oxidantes/toxicidad , Nodulación de la Raíz de la Planta , Sinorhizobium meliloti/efectos de los fármacos , Sinorhizobium meliloti/genética , Estrés Fisiológico
19.
Mol Microbiol ; 91(1): 185-97, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24255990

RESUMEN

The genome of Mycobacterium tuberculosis encodes two paralogous P1 B 4 -ATPases, CtpD (Rv1469) and CtpJ (Rv3743). Both proteins showed ATPase activation by Co(2+) and Ni(2+) , and both appear to be required for metal efflux from the cell. However, using a combination of biochemical and genetic studies we found that these proteins play non-redundant roles in virulence and metal efflux. CtpJ expression is induced by Co(2+) and this protein possesses a relatively high turnover rate. A ctpJ deletion mutant accumulated Co(2+) , indicating that this ATPase controls cytoplasmic metal levels. In contrast, CtpD expression is induced by redox stressors and this protein displays a relatively low turnover rate. A ctpD mutant failed to accumulate metal, suggesting an alternative cellular function. ctpD is cotranscribed with two thioredoxin genes trxA (Rv1470), trxB (Rv1471), and an enoyl-coA hydratase (Rv1472), indicating a possible role for CtpD in the metallation of these redox-active proteins. Supporting this, in vitro metal binding assays showed that TrxA binds Co(2+) and Ni(2+) . Mutation of ctpD, but not ctpJ, reduced bacterial fitness in the mouse lung, suggesting that redox maintenance, but not Co(2+) accumulation, is important for growth in vivo.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/fisiología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/fisiología , Cobalto/metabolismo , Mycobacterium tuberculosis/enzimología , Níquel/metabolismo , Tuberculosis/microbiología , Factores de Virulencia/metabolismo , Adenosina Trifosfatasas/genética , Animales , Proteínas Bacterianas/genética , Citoplasma/metabolismo , Modelos Animales de Enfermedad , Femenino , Aptitud Genética , Genoma Bacteriano , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Mutación , Mycobacterium tuberculosis/fisiología , Especies de Nitrógeno Reactivo/metabolismo , Tiorredoxinas , Factores de Virulencia/genética
20.
Artículo en Inglés | MEDLINE | ID: mdl-24205499

RESUMEN

Copper is an important micronutrient required as a redox co-factor in the catalytic centers of enzymes. However, free copper is a potential hazard because of its high chemical reactivity. Consequently, organisms exert a tight control on Cu(+) transport (entry-exit) and traffic through different compartments, ensuring the homeostasis required for cuproprotein synthesis and prevention of toxic effects. Recent studies based on biochemical, bioinformatics, and metalloproteomics approaches, reveal a highly regulated system of transcriptional regulators, soluble chaperones, membrane transporters, and target cuproproteins distributed in the various bacterial compartments. As a result, new questions have emerged regarding the diversity and apparent redundancies of these components, their irregular presence in different organisms, functional interactions, and resulting system architectures.


Asunto(s)
Bacterias/metabolismo , Cobre/metabolismo , Homeostasis , Bacterias/genética , Regulación Bacteriana de la Expresión Génica , Redes y Vías Metabólicas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...