Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Angew Chem Int Ed Engl ; 63(16): e202401379, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38407997

RESUMEN

Ferritins are multimeric cage-forming proteins that play a crucial role in cellular iron homeostasis. All H-chain-type ferritins harbour a diiron site, the ferroxidase centre, at the centre of a 4 α-helical bundle, but bacterioferritins are unique in also binding 12 hemes per 24 meric assembly. The ferroxidase centre is known to be required for the rapid oxidation of Fe2+ during deposition of an immobilised ferric mineral core within the protein's hollow interior. In contrast, the heme of bacterioferritin is required for the efficient reduction of the mineral core during iron release, but has little effect on the rate of either oxidation or mineralisation of iron. Thus, the current view is that these two cofactors function in iron uptake and release, respectively, with no functional overlap. However, rapid electron transfer between the heme and ferroxidase centre of bacterioferritin from Escherichia coli was recently demonstrated, suggesting that the two cofactors may be functionally connected. Here we report absorbance and (magnetic) circular dichroism spectroscopies, together with in vitro assays of iron-release kinetics, which demonstrate that the ferroxidase centre plays an important role in the reductive mobilisation of the bacterioferritin mineral core, which is dependent on the heme-ferroxidase centre electron transfer pathway.


Asunto(s)
Ceruloplasmina , Hierro , Hierro/química , Ceruloplasmina/química , Escherichia coli/metabolismo , Ferritinas/química , Proteínas Bacterianas/metabolismo , Grupo Citocromo b/química , Minerales , Oxidación-Reducción , Hemo/metabolismo
2.
Crit Rev Toxicol ; 53(10): 658-701, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38050998

RESUMEN

Tobacco use is a major cause of preventable morbidity and mortality globally. Tobacco products, including smokeless tobacco (ST), generally contain tobacco-specific N-nitrosamines (TSNAs), such as N'-nitrosonornicotine (NNN) and 4-(methylnitrosamino)-1-(3-pyridyl)-butanone (NNK), which are potent carcinogens that cause mutations in critical genes in human DNA. This review covers the series of biochemical and chemical transformations, related to TSNAs, leading from tobacco cultivation to cancer initiation. A key aim of this review is to provide a greater understanding of TSNAs: their precursors, the microbial and chemical mechanisms that contribute to their formation in ST, their mutagenicity leading to cancer due to ST use, and potential means of lowering TSNA levels in tobacco products. TSNAs are not present in harvested tobacco but can form due to nitrosating agents reacting with tobacco alkaloids present in tobacco during certain types of curing. TSNAs can also form during or following ST production when certain microorganisms perform nitrate metabolism, with dissimilatory nitrate reductases converting nitrate to nitrite that is then released into tobacco and reacts chemically with tobacco alkaloids. When ST usage occurs, TSNAs are absorbed and metabolized to reactive compounds that form DNA adducts leading to mutations in critical target genes, including the RAS oncogenes and the p53 tumor suppressor gene. DNA repair mechanisms remove most adducts induced by carcinogens, thus preventing many but not all mutations. Lastly, because TSNAs and other agents cause cancer, previously documented strategies for lowering their levels in ST products are discussed, including using tobacco with lower nornicotine levels, pasteurization and other means of eliminating microorganisms, omitting fermentation and fire-curing, refrigerating ST products, and including nitrite scavenging chemicals as ST ingredients.


Asunto(s)
Neoplasias , Nitrosaminas , Tabaco sin Humo , Humanos , Carcinógenos/toxicidad , Mutágenos , Neoplasias/inducido químicamente , Nitratos , Nitritos , Nitrosaminas/toxicidad , Nitrosaminas/química , Nitrosaminas/metabolismo , Tabaco sin Humo/toxicidad
3.
J Am Chem Soc ; 144(16): 7129-7145, 2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35416044

RESUMEN

Previously characterized nitrite reductases fall into three classes: siroheme-containing enzymes (NirBD), cytochrome c hemoproteins (NrfA and NirS), and copper-containing enzymes (NirK). We show here that the di-iron protein YtfE represents a physiologically relevant new class of nitrite reductases. Several functions have been previously proposed for YtfE, including donating iron for the repair of iron-sulfur clusters that have been damaged by nitrosative stress, releasing nitric oxide (NO) from nitrosylated iron, and reducing NO to nitrous oxide (N2O). Here, in vivo reporter assays confirmed that Escherichia coli YtfE increased cytoplasmic NO production from nitrite. Spectroscopic and mass spectrometric investigations revealed that the di-iron site of YtfE exists in a mixture of forms, including nitrosylated and nitrite-bound, when isolated from nitrite-supplemented, but not nitrate-supplemented, cultures. Addition of nitrite to di-ferrous YtfE resulted in nitrosylated YtfE and the release of NO. Kinetics of nitrite reduction were dependent on the nature of the reductant; the lowest Km, measured for the di-ferrous form, was ∼90 µM, well within the intracellular nitrite concentration range. The vicinal di-cysteine motif, located in the N-terminal domain of YtfE, was shown to function in the delivery of electrons to the di-iron center. Notably, YtfE exhibited very low NO reductase activity and was only able to act as an iron donor for reconstitution of apo-ferredoxin under conditions that damaged its di-iron center. Thus, YtfE is a high-affinity, low-capacity nitrite reductase that we propose functions to relieve nitrosative stress by acting in combination with the co-regulated NO-consuming enzymes Hmp and Hcp.


Asunto(s)
Proteínas de Escherichia coli , Estrés Nitrosativo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Hierro/química , Óxido Nítrico/metabolismo , Nitrito Reductasas/metabolismo , Nitritos/metabolismo
4.
Chembiochem ; 23(13): e202200257, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35510795

RESUMEN

Ferritins are ubiquitous diiron enzymes involved in iron(II) detoxification and oxidative stress responses and can act as metabolic iron stores. The overall reaction mechanisms of ferritin enzymes are still unclear, particularly concerning the role of the conserved, near catalytic center Tyr residue. Thus, we carried out a computational study of a ferritin using a large cluster model of well over 300 atoms including its first- and second-coordination sphere. The calculations reveal important insight into the structure and reactivity of ferritins. Specifically, the active site Tyr residue delivers a proton and electron in the catalytic cycle prior to iron(II) oxidation. In addition, the calculations highlight a likely cation binding site at Asp65 , which through long-range electrostatic interactions, influences the electronic configuration and charge distributions of the metal center. The results are consistent with experimental observations but reveal novel detail of early mechanistic steps that lead to an unusual mixed-valent iron(III)-iron(II) center.


Asunto(s)
Ferritinas , Oxígeno , Sitios de Unión , Cationes/metabolismo , Compuestos Férricos/química , Ferritinas/química , Compuestos Ferrosos/química , Hierro/química , Oxidación-Reducción , Oxígeno/metabolismo
5.
Appl Environ Microbiol ; 88(7): e0002922, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35285709

RESUMEN

Isoprene (2-methyl-1,3-butadiene) is a climate-active gas released to the atmosphere in large quantities, comparable to methane in magnitude. Several bacteria have been isolated which can grow on isoprene as a sole carbon and energy source, but very little information is available about the degradation of isoprene by these bacteria at the biochemical level. Isoprene utilization is dependent on a multistep pathway, with the first step being the oxidation of isoprene to epoxy-isoprene. This is catalyzed by a four-component soluble diiron monooxygenase, isoprene monooxygenase (IsoMO). IsoMO is a six-protein complex comprising an oxygenase (IsoABE), containing the di-iron active site, a Rieske-type ferredoxin (IsoC), a NADH reductase (IsoF), and a coupling/effector protein (IsoD), homologous to the soluble methane monooxygenase and alkene/aromatic monooxygenases. Here, we describe the purification of the IsoMO components from Rhodococcus sp. AD45 and reconstitution of isoprene-oxidation activity in vitro. Some IsoMO components were expressed and purified from the homologous host Rhodococcus sp. AD45-ID, a Rhodococcus sp. AD45 strain lacking the megaplasmid which contains the isoprene metabolic gene cluster. Others were expressed in Escherichia coli and purified as fusion proteins. We describe the characterization of these purified components and demonstrate their activity when combined with Rhodococcus sp. AD45 cell lysate. Demonstration of IsoMO activity in vitro provides a platform for further biochemical and biophysical characterization of this novel soluble diiron center monooxygenase, facilitating new insights into the enzymatic basis for the bacterial degradation of isoprene. IMPORTANCE Isoprene is a highly abundant climate-active gas and a carbon source for some bacteria. Analyses of the genes encoding isoprene monooxygenase (IsoMO) indicate this enzyme is a soluble diiron center monooxygenase in the same family of oxygenases as soluble methane monooxygenase, alkene monooxygenase, and toluene monooxygenase. We report the initial biochemical characterization of IsoMO from Rhodococcus, the first from any bacterium, describing the challenging purification and reconstitution of in vitro activity of its four components. This study lays the foundation for future detailed mechanistic studies of IsoMO, a key enzyme in the global isoprene cycle.


Asunto(s)
Rhodococcus , Butadienos , Carbono/metabolismo , Hemiterpenos/metabolismo , Oxigenasas de Función Mixta/metabolismo , Oxigenasas/metabolismo , Rhodococcus/metabolismo
6.
Proc Natl Acad Sci U S A ; 116(6): 2058-2067, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30659147

RESUMEN

The gene encoding the cyanobacterial ferritin SynFtn is up-regulated in response to copper stress. Here, we show that, while SynFtn does not interact directly with copper, it is highly unusual in several ways. First, its catalytic diiron ferroxidase center is unlike those of all other characterized prokaryotic ferritins and instead resembles an animal H-chain ferritin center. Second, as demonstrated by kinetic, spectroscopic, and high-resolution X-ray crystallographic data, reaction of O2 with the di-Fe2+ center results in a direct, one-electron oxidation to a mixed-valent Fe2+/Fe3+ form. Iron-O2 chemistry of this type is currently unknown among the growing family of proteins that bind a diiron site within a four α-helical bundle in general and ferritins in particular. The mixed-valent form, which slowly oxidized to the more usual di-Fe3+ form, is an intermediate that is continually generated during mineralization. Peroxide, rather than superoxide, is shown to be the product of O2 reduction, implying that ferroxidase centers function in pairs via long-range electron transfer through the protein resulting in reduction of O2 bound at only one of the centers. We show that electron transfer is mediated by the transient formation of a radical on Tyr40, which lies ∼4 Å from the diiron center. As well as demonstrating an expansion of the iron-O2 chemistry known to occur in nature, these data are also highly relevant to the question of whether all ferritins mineralize iron via a common mechanism, providing unequivocal proof that they do not.


Asunto(s)
Compuestos Férricos/química , Compuestos Ferrosos/química , Oxígeno/química , Peróxidos/química , Proteínas/química , Ceruloplasmina/química , Transporte de Electrón , Ferritinas/química , Hierro/química , Modelos Moleculares , Conformación Molecular , Oxidación-Reducción , Relación Estructura-Actividad
7.
J Biol Chem ; 295(28): 9752-9765, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32303639

RESUMEN

The bacterial protein WhiD belongs to the Wbl family of iron-sulfur [Fe-S] proteins present only in the actinomycetes. In Streptomyces coelicolor, it is required for the late stages of sporulation, but precisely how it functions is unknown. Here, we report results from in vitro and in vivo experiments with WhiD from Streptomyces venezuelae (SvWhiD), which differs from S. coelicolor WhiD (ScWhiD) only at the C terminus. We observed that, like ScWhiD and other Wbl proteins, SvWhiD binds a [4Fe-4S] cluster that is moderately sensitive to O2 and highly sensitive to nitric oxide (NO). However, although all previous studies have reported that Wbl proteins are monomers, we found that SvWhiD exists in a monomer-dimer equilibrium associated with its unusual C-terminal extension. Several Wbl proteins of Mycobacterium tuberculosis are known to interact with its principal sigma factor SigA. Using bacterial two-hybrid, gel filtration, and MS analyses, we demonstrate that SvWhiD interacts with domain 4 of the principal sigma factor of Streptomyces, σHrdB (σHrdB4). Using MS, we determined the dissociation constant (Kd ) for the SvWhiD-σHrdB4 complex as ∼0.7 µm, consistent with a relatively tight binding interaction. We found that complex formation was cluster dependent and that a reaction with NO, which was complete at 8-10 NO molecules per cluster, resulted in dissociation into the separate proteins. The SvWhiD [4Fe-4S] cluster was significantly less sensitive to reaction with O2 and NO when SvWhiD was bound to σHrdB4, consistent with protection of the cluster in the complex.


Asunto(s)
Proteínas Bacterianas , Proteínas de Unión al ADN , Factor sigma , Streptomyces , Factores de Transcripción , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/metabolismo , Factor sigma/química , Factor sigma/metabolismo , Streptomyces/química , Streptomyces/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo
8.
J Biol Chem ; 295(51): 17602-17623, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33454001

RESUMEN

Iron is an essential micronutrient, and, in the case of bacteria, its availability is commonly a growth-limiting factor. However, correct functioning of cells requires that the labile pool of chelatable "free" iron be tightly regulated. Correct metalation of proteins requiring iron as a cofactor demands that such a readily accessible source of iron exist, but overaccumulation results in an oxidative burden that, if unchecked, would lead to cell death. The toxicity of iron stems from its potential to catalyze formation of reactive oxygen species that, in addition to causing damage to biological molecules, can also lead to the formation of reactive nitrogen species. To avoid iron-mediated oxidative stress, bacteria utilize iron-dependent global regulators to sense the iron status of the cell and regulate the expression of proteins involved in the acquisition, storage, and efflux of iron accordingly. Here, we survey the current understanding of the structure and mechanism of the important members of each of these classes of protein. Diversity in the details of iron homeostasis mechanisms reflect the differing nutritional stresses resulting from the wide variety of ecological niches that bacteria inhabit. However, in this review, we seek to highlight the similarities of iron homeostasis between different bacteria, while acknowledging important variations. In this way, we hope to illustrate how bacteria have evolved common approaches to overcome the dual problems of the insolubility and potential toxicity of iron.


Asunto(s)
Bacterias/metabolismo , Hierro/metabolismo , Bacterias/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Ferritinas/química , Ferritinas/metabolismo , Hierro/química , Estrés Oxidativo , Especies de Nitrógeno Reactivo/química , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/química , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Sideróforos/química , Sideróforos/metabolismo
9.
Microbiology (Reading) ; 167(11)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34825885

RESUMEN

Ferritins are proteins forming 24meric rhombic dodecahedral cages that play a key role in iron storage and detoxification in all cell types. Their function requires the transport of Fe2+ from the exterior of the protein to buried di-iron catalytic sites, known as ferroxidase centres, where Fe2+ is oxidized to form Fe3+-oxo precursors of the ferritin mineral core. The route of iron transit through animal ferritins is well understood: the Fe2+ substrate enters the protein via channels at the threefold axes and conserved carboxylates on the inner surface of the protein cage have been shown to contribute to transient binding sites that guide Fe2+ to the ferroxidase centres. The routes of iron transit through prokaryotic ferritins are less well studied but for some, at least, there is evidence that channels at the twofold axes are the major route for Fe2+ uptake. SynFtn, isolated from the cyanobacterium Synechococcus CC9311, is an atypical prokaryotic ferritin that was recently shown to take up Fe2+ via its threefold channels. However, the transfer site carboxylate residues conserved in animal ferritins are absent, meaning that the route taken from the site of iron entry into SynFtn to the catalytic centre is yet to be defined. Here, we report the use of a combination of site-directed mutagenesis, absorbance-monitored activity assays and protein crystallography to probe the effect of substitution of two residues potentially involved in this pathway. Both Glu141 and Asp65 play a role in guiding the Fe2+ substrate to the ferroxidase centre. In the absence of Asp65, routes for Fe2+ to, and Fe3+ exit from, the ferroxidase centre are affected resulting in inefficient formation of the mineral core. These observations further define the iron transit route in what may be the first characterized example of a new class of ferritins peculiar to cyanobacteria.


Asunto(s)
Ferritinas , Hierro , Synechococcus , Animales , Dominio Catalítico , Ceruloplasmina/química , Ceruloplasmina/genética , Ferritinas/química , Ferritinas/genética , Hierro/metabolismo , Minerales/química , Oxidación-Reducción , Synechococcus/química
10.
Angew Chem Int Ed Engl ; 60(15): 8376-8379, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33460502

RESUMEN

The iron redox cycle in ferritins is not completely understood. Bacterioferritins are distinct from other ferritins in that they contain haem groups. It is acknowledged that the two iron motifs in bacterioferritins, the di-nuclear ferroxidase centre and the haem B group, play key roles in two opposing processes, iron sequestration and iron mobilisation, respectively, and the two redox processes are independent. Herein, we show that in Escherichia coli bacterioferritin, there is an electron transfer pathway from the haem to the ferroxidase centre suggesting a new role(s) haem might play in bacterioferritins.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ceruloplasmina/metabolismo , Grupo Citocromo b/metabolismo , Ferritinas/metabolismo , Hemo/metabolismo , Proteínas Bacterianas/química , Ceruloplasmina/química , Grupo Citocromo b/química , Transporte de Electrón , Escherichia coli/química , Escherichia coli/metabolismo , Ferritinas/química , Hemo/química
11.
Angew Chem Int Ed Engl ; 60(15): 8361-8369, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33482043

RESUMEN

Both O2 and H2 O2 can oxidize iron at the ferroxidase center (FC) of Escherichia coli bacterioferritin (EcBfr) but mechanistic details of the two reactions need clarification. UV/Vis, EPR, and Mössbauer spectroscopies have been used to follow the reactions when apo-EcBfr, pre-loaded anaerobically with Fe2+ , was exposed to O2 or H2 O2 . We show that O2 binds di-Fe2+ FC reversibly, two Fe2+ ions are oxidized in concert and a H2 O2 molecule is formed and released to the solution. This peroxide molecule further oxidizes another di-Fe2+ FC, at a rate circa 1000 faster than O2 , ensuring an overall 1:4 stoichiometry of iron oxidation by O2 . Initially formed Fe3+ can further react with H2 O2 (producing protein bound radicals) but relaxes within seconds to an H2 O2 -unreactive di-Fe3+ form. The data obtained suggest that the primary role of EcBfr in vivo may be to detoxify H2 O2 rather than sequester iron.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ceruloplasmina/metabolismo , Grupo Citocromo b/metabolismo , Escherichia coli/química , Ferritinas/metabolismo , Peróxido de Hidrógeno/metabolismo , Hierro/metabolismo , Oxígeno/metabolismo , Proteínas Bacterianas/química , Ceruloplasmina/química , Grupo Citocromo b/química , Escherichia coli/metabolismo , Ferritinas/química , Peróxido de Hidrógeno/química , Hierro/química , Modelos Moleculares , Oxidación-Reducción , Oxígeno/química
12.
J Am Chem Soc ; 142(11): 5104-5116, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32078310

RESUMEN

The [Fe2S2]-RsrR gene transcription regulator senses the redox status in bacteria by modulating DNA binding, while its cluster cycles between +1 and +2 states-only the latter binds DNA. We have previously shown that RsrR can undergo remarkable conformational changes involving a 100° rotation of tryptophan 9 between exposed (Out) and buried (In) states. Here, we have used the chemical modification of Trp9, site-directed mutagenesis, and crystallographic and computational chemical studies to show that (i) the Out and In states correspond to oxidized and reduced RsrR, respectively, (ii) His33 is protonated in the In state due to a change in its pKa caused by cluster reduction, and (iii) Trp9 rotation is conditioned by the response of its dipole moment to environmental electrostatic changes. Our findings illustrate a novel function of protonation resulting from electron transfer.


Asunto(s)
Proteínas de Unión al ADN/química , ADN/química , Electrones , Proteínas Hierro-Azufre/química , Protones , Factores de Transcripción/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histidina/química , Histidina/genética , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Simulación de Dinámica Molecular , Mutación , Oxidación-Reducción , Unión Proteica , Conformación Proteica , Streptomyces/enzimología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Microbiology (Reading) ; 166(10): 909-917, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32886603

RESUMEN

Nitrous oxide (N2O) is a potent greenhouse gas that is produced naturally as an intermediate during the process of denitrification carried out by some soil bacteria. It is consumed by nitrous oxide reductase (N2OR), the terminal enzyme of the denitrification pathway, which catalyses a reduction reaction to generate dinitrogen. N2OR contains two important copper cofactors (CuA and CuZ centres) that are essential for activity, and in copper-limited environments, N2OR fails to function, contributing to rising levels of atmospheric N2O and a major environmental challenge. Here we report studies of nosX, one of eight genes in the nos cluster of the soil dwelling α-proteobaterium Paraccocus denitrificans. A P. denitrificans ΔnosX deletion mutant failed to reduce N2O under both copper-sufficient and copper-limited conditions, demonstrating that NosX plays an essential role in N2OR activity. N2OR isolated from nosX-deficient cells was found to be unaffected in terms of the assembly of its copper cofactors, and to be active in in vitro assays, indicating that NosX is not required for the maturation of the enzyme; in particular, it plays no part in the assembly of either of the CuA and CuZ centres. Furthermore, quantitative Reverse Transcription PCR (qRT-PCR) studies showed that NosX does not significantly affect the expression of the N2OR-encoding nosZ gene. NosX is a homologue of the FAD-binding protein ApbE from Pseudomonas stutzeri, which functions in the flavinylation of another N2OR accessory protein, NosR. Thus, it is likely that NosX is a system-specific maturation factor of NosR, and so is indirectly involved in maintaining the reaction cycle of N2OR and cellular N2O reduction.


Asunto(s)
Proteínas Bacterianas/metabolismo , Óxido Nitroso/metabolismo , Paracoccus denitrificans/metabolismo , Proteínas Bacterianas/genética , Coenzimas/metabolismo , Cobre/metabolismo , Desnitrificación , Proteínas de la Membrana/metabolismo , Mutación , Oxidación-Reducción , Oxidorreductasas/metabolismo , Paracoccus denitrificans/enzimología , Paracoccus denitrificans/genética
14.
Proc Natl Acad Sci U S A ; 114(16): E3215-E3223, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28373574

RESUMEN

The iron-sulfur cluster containing protein Fumarate and Nitrate Reduction (FNR) is the master regulator for the switch between anaerobic and aerobic respiration in Escherichia coli and many other bacteria. The [4Fe-4S] cluster functions as the sensory module, undergoing reaction with O2 that leads to conversion to a [2Fe-2S] form with loss of high-affinity DNA binding. Here, we report studies of the FNR cluster conversion reaction using time-resolved electrospray ionization mass spectrometry. The data provide insight into the reaction, permitting the detection of cluster conversion intermediates and products, including a [3Fe-3S] cluster and persulfide-coordinated [2Fe-2S] clusters [[2Fe-2S](S) n , where n = 1 or 2]. Analysis of kinetic data revealed a branched mechanism in which cluster sulfide oxidation occurs in parallel with cluster conversion and not as a subsequent, secondary reaction to generate [2Fe-2S](S) n species. This methodology shows great potential for broad application to studies of protein cofactor-small molecule interactions.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Fumaratos/química , Proteínas Hierro-Azufre/química , Hierro/química , Nitratos/química , Oxígeno/farmacología , Sulfuros/química , Proteínas de Escherichia coli/metabolismo , Fumaratos/metabolismo , Hierro/metabolismo , Proteínas Hierro-Azufre/metabolismo , Cinética , Modelos Moleculares , Nitratos/metabolismo , Oxidación-Reducción , Conformación Proteica , Espectrometría de Masa por Ionización de Electrospray , Sulfuros/metabolismo
15.
J Am Chem Soc ; 141(6): 2367-2375, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30657661

RESUMEN

The recently discovered Rrf2 family transcriptional regulator RsrR coordinates a [2Fe-2S] cluster. Remarkably, binding of the protein to RsrR-regulated promoter DNA sequences is switched on and off through the facile cycling of the [2Fe-2S] cluster between +2 and +1 states. Here, we report high resolution crystal structures of the RsrR dimer, revealing that the [2Fe-2S] cluster is asymmetrically coordinated across the RsrR monomer-monomer interface by two Cys residues from one subunit and His and Glu residues from the other. To our knowledge, this is the first example of a protein bound [Fe-S] cluster with three different amino acid side chains as ligands, and of Glu acting as ligand to a [2Fe-2S] cluster. Analyses of RsrR structures revealed a conformational change, centered on Trp9, which results in a significant shift in the DNA-binding helix-turn-helix region.


Asunto(s)
Proteínas Bacterianas/química , Factores de Transcripción/química , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , ADN/metabolismo , Modelos Moleculares , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Factores de Transcripción/metabolismo
16.
Chemistry ; 25(14): 3675-3684, 2019 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-30600851

RESUMEN

Nitric oxide (NO) can function as both a cytotoxin and a signalling molecule. In both cases, reaction with iron-sulfur (Fe-S) cluster proteins plays an important role because Fe-S clusters are reactive towards NO and so are a primary site of general NO-induced damage (toxicity). This sensitivity to nitrosylation is harnessed in the growing group of regulatory proteins that function in sensing of NO via an Fe-S cluster. Although information about the products of cluster nitrosylation is now emerging, detection and identification of intermediates remains a major challenge, due to their transient nature and the difficulty in distinguishing spectroscopically similar iron-NO species. Here we report studies of the NO-sensing Fe-S cluster regulators NsrR and WhiD using non-denaturing mass spectrometry, in which non-covalent interactions between the protein and Fe/S/NO species are preserved. The data provide remarkable insight into the nitrosylation reactions, permitting identification, for the first time, of protein-bound mono-, di- and tetranitrosyl [4Fe-4S] cluster complexes ([4Fe-4S](NO), [4Fe-4S])(NO)2 and [4Fe-4S](NO)4 ) as intermediates along pathways to formation of product Roussin's red ester (RRE) and Roussin's black salt (RBS)-like species. The data allow the nitrosylation mechanisms of NsrR and WhiD to be elucidated and clearly distinguished.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Hierro-Azufre/metabolismo , Mycobacterium tuberculosis/metabolismo , Óxido Nítrico/metabolismo , Streptomyces coelicolor/metabolismo , Factores de Transcripción/metabolismo , Proteínas Bacterianas/química , Proteínas Hierro-Azufre/química , Modelos Moleculares , Mycobacterium tuberculosis/química , Conformación Proteica , Espectrometría de Masa por Ionización de Electrospray , Streptomyces coelicolor/química , Factores de Transcripción/química
17.
Nucleic Acids Res ; 45(11): 6600-6612, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28482027

RESUMEN

Mycobacterium tuberculosis (MTb) is the causative agent of pulmonary tuberculosis (TB). MTb colonizes the human lung, often entering a non-replicating state before progressing to life-threatening active infections. Transcriptional reprogramming is essential for TB pathogenesis. In vitro, Cmr (a member of the CRP/FNR super-family of transcription regulators) bound at a single DNA site to act as a dual regulator of cmr transcription and an activator of the divergent rv1676 gene. Transcriptional profiling and DNA-binding assays suggested that Cmr directly represses dosR expression. The DosR regulon is thought to be involved in establishing latent tuberculosis infections in response to hypoxia and nitric oxide. Accordingly, DNA-binding by Cmr was severely impaired by nitrosation. A cmr mutant was better able to survive a nitrosative stress challenge but was attenuated in a mouse aerosol infection model. The complemented mutant exhibited a ∼2-fold increase in cmr expression, which led to increased sensitivity to nitrosative stress. This, and the inability to restore wild-type behaviour in the infection model, suggests that precise regulation of the cmr locus, which is associated with Region of Difference 150 in hypervirulent Beijing strains of Mtb, is important for TB pathogenesis.


Asunto(s)
Proteínas Bacterianas/genética , Mycobacterium tuberculosis/metabolismo , Proteínas Quinasas/genética , Factores de Transcripción/fisiología , Tuberculosis/microbiología , Animales , Proteínas Bacterianas/metabolismo , Células Cultivadas , Proteínas de Unión al ADN , Escherichia coli , Femenino , Regulación Bacteriana de la Expresión Génica , Macrófagos/microbiología , Ratones Endogámicos BALB C , Mycobacterium smegmatis , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Oxidación-Reducción , Unión Proteica , Proteínas Quinasas/metabolismo , Transcripción Genética , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
18.
Plant J ; 89(3): 590-600, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27801963

RESUMEN

Proteins of the cytosolic pathway for iron-sulphur (FeS) cluster assembly are conserved, except that plants lack a gene for CFD1 (Cytosolic FeS cluster Deficient 1). This poses the question of how NBP35 (Nucleotide-Binding Protein 35 kDa), the heteromeric partner of CFD1 in metazoa, functions on its own in plants. Firstly, we created viable mutant alleles of NBP35 in Arabidopsis to overcome embryo lethality of previously reported knockout mutations. RNAi knockdown lines with less than 30% NBP35 protein surprisingly showed no developmental or biochemical differences to wild-type. Substitution of Cys14 to Ala, which destabilized the N-terminal Fe4 S4 cluster in vitro, caused mild growth defects and a significant decrease in the activity of cytosolic FeS enzymes such as aconitase and aldehyde oxidases. The DNA glycosylase ROS1 was only partially decreased in activity and xanthine dehydrogenase not at all. Plants with strongly depleted NBP35 protein in combination with Cys14 to Ala substitution had distorted leaf development and decreased FeS enzyme activities. To find protein interaction partners of NBP35, a yeast-two-hybrid screen was carried out that identified NBP35 and DRE2 (Derepressed for Ribosomal protein S14 Expression). NBP35 is known to form a dimer, and DRE2 acts upstream in the cytosolic FeS protein assembly pathway. The NBP35-DRE2 interaction was not disrupted by Cys14 to Ala substitution. Our results show that NBP35 has a function in the maturation of FeS proteins that is conserved in plants, and is closely allied to the function of DRE2.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Proteínas Portadoras/metabolismo , Citosol/metabolismo , Proteínas Hierro-Azufre/metabolismo , Secuencia de Aminoácidos , Arabidopsis/genética , Arabidopsis/crecimiento & desarrollo , Proteínas de Arabidopsis/genética , Proteínas Portadoras/genética , Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Proteínas Hierro-Azufre/genética , Mutación Missense , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Hojas de la Planta/genética , Hojas de la Planta/crecimiento & desarrollo , Hojas de la Planta/metabolismo , Plantas Modificadas Genéticamente , Unión Proteica , Interferencia de ARN , Homología de Secuencia de Aminoácido
19.
Biochim Biophys Acta Proteins Proteom ; 1866(2): 275-282, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29146226

RESUMEN

Copper-transporting P-type ATPases, which play important roles in trafficking Cu(I) across membranes for the biogenesis of copper proteins or for copper detoxification, contain a variable number of soluble metal-binding domains at their N-termini. It is increasingly apparent that these play an important role in regulating copper transport in a Cu(I)-responsive manner, but how they do this is unknown. CopA, a Cu(I)-transporter from Bacillus subtilis, contains two N-terminal soluble domains that are closely packed, with inter-domain interactions at two principal regions. Here, we sought to determine the extent to which the domains interact in the absence of their inter-domain covalent linker, and how their Cu(I)-binding properties are affected. Studies of a 1:1 mixture of separate CopAa and CopAb domains showed that the domains do not form a stable complex, with only indirect evidence of a weak interaction between them. Their Cu(I)-binding behaviour was distinct from that of the two domain protein and consistent with a lack of interaction between the domains. Cu(I)-mediated protein association was observed, but this occurred only between domains of the same type. Thus, the inter-domain covalent link between CopAa and CopAb is essential for inter-domain interactions and for Cu(I)-binding behaviour.


Asunto(s)
Bacillus subtilis/química , Proteínas Bacterianas/química , Cobre/química , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cobre/metabolismo , Dominios Proteicos
20.
J Biol Chem ; 291(16): 8663-72, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26887943

RESUMEN

NsrR is an iron-sulfur cluster protein that regulates the nitric oxide (NO) stress response of many bacteria. NsrR from Streptomyces coelicolor regulates its own expression and that of only two other genes, hmpA1 and hmpA2, which encode HmpA enzymes predicted to detoxify NO. NsrR binds promoter DNA with high affinity only when coordinating a [4Fe-4S] cluster. Here we show that reaction of [4Fe-4S] NsrR with NO affects DNA binding differently depending on the gene promoter. Binding to the hmpA2 promoter was abolished at ∼2 NO per cluster, although for the hmpA1 and nsrR promoters, ∼4 and ∼8 NO molecules, respectively, were required to abolish DNA binding. Spectroscopic and kinetic studies of the NO reaction revealed a rapid, multi-phase, non-concerted process involving up to 8-10 NO molecules per cluster, leading to the formation of several iron-nitrosyl species. A distinct intermediate was observed at ∼2 NO per cluster, along with two further intermediates at ∼4 and ∼6 NO. The NsrR nitrosylation reaction was not significantly affected by DNA binding. These results show that NsrR regulates different promoters in response to different concentrations of NO. Spectroscopic evidence indicates that this is achieved by different NO-FeS complexes.


Asunto(s)
Proteínas Bacterianas/metabolismo , ADN Bacteriano/metabolismo , Proteínas Hierro-Azufre/metabolismo , Óxido Nítrico/metabolismo , Regiones Promotoras Genéticas/fisiología , Streptomyces coelicolor/metabolismo , Factores de Transcripción/metabolismo , Proteínas Bacterianas/genética , ADN Bacteriano/genética , Proteínas Hierro-Azufre/genética , Streptomyces coelicolor/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA