Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Microb Cell Fact ; 23(1): 151, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789996

ABSTRACT

BACKGROUND: Xylans are polysaccharides that are naturally abundant in agricultural by-products, such as cereal brans and straws. Microbial degradation of arabinoxylan is facilitated by extracellular esterases that remove acetyl, feruloyl, and p-coumaroyl decorations. The bacterium Ruminiclostridium cellulolyticum possesses the Xua (xylan utilization associated) system, which is responsible for importing and intracellularly degrading arabinoxylodextrins. This system includes an arabinoxylodextrins importer, four intracellular glycosyl hydrolases, and two intracellular esterases, XuaH and XuaJ which are encoded at the end of the gene cluster. RESULTS: Genetic studies demonstrate that the genes xuaH and xuaJ are part of the xua operon, which covers xuaABCDD'EFGHIJ. This operon forms a functional unit regulated by the two-component system XuaSR. The esterases encoded at the end of the cluster have been further characterized: XuaJ is an acetyl esterase active on model substrates, while XuaH is a xylan feruloyl- and p-coumaryl-esterase. This latter is active on oligosaccharides derived from wheat bran and wheat straw. Modelling studies indicate that XuaH has the potential to interact with arabinoxylobiose acylated with mono- or diferulate. The intracellular esterases XuaH and XuaJ are believed to allow the cell to fully utilize the complex acylated arabinoxylo-dextrins imported into the cytoplasm during growth on wheat bran or straw. CONCLUSIONS: This study reports for the first time that a cytosolic feruloyl esterase is part of an intracellular arabinoxylo-dextrin import and degradation system, completing its cytosolic enzymatic arsenal. This system represents a new pathway for processing highly-decorated arabinoxylo-dextrins, which could provide a competitive advantage to the cell and may have interesting biotechnological applications.


Subject(s)
Lignin , Xylans , Xylans/metabolism , Lignin/metabolism , Biomass , Coumaric Acids/metabolism , Oligosaccharides/metabolism , Clostridiales/metabolism , Operon , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Multigene Family , Acetylesterase/metabolism , Acetylesterase/genetics , Carboxylic Ester Hydrolases
2.
Inorg Chem ; 62(17): 6617-6628, 2023 May 01.
Article in English | MEDLINE | ID: mdl-37057906

ABSTRACT

A family of bacterial copper storage proteins (the Csps) possess thiolate-lined four-helix bundles whose cores can be filled with Cu(I) ions. The majority of Csps are cytosolic (Csp3s), and in vitro studies carried out to date indicate that the Csp3s from Methylosinus trichosporium OB3b (MtCsp3), Bacillus subtilis (BsCsp3), and Streptomyces lividans (SlCsp3) are alike. Bioinformatics have highlighted homologues with potentially different Cu(I)-binding properties from these characterized "classical" Csp3s. Determination herein of the crystal structure of the protein (RkCsp3) from the methanotroph Methylocystis sp. strain Rockwell with Cu(I) bound identifies this as the first studied example of a new subgroup of Csp3s. The most significant structural difference from classical Csp3s is the presence of only two Cu(I) sites at the mouth of the bundle via which Cu(I) ions enter and leave. This is due to the absence of three Cys residues and a His-containing motif, which allow classical Csp3s to bind five to six Cu(I) ions in this region. Regardless, RkCsp3 exhibits rapid Cu(I) binding and the fastest measured Cu(I) removal rate for a Csp3 when using high-affinity ligands as surrogate partners. New experiments on classical Csp3s demonstrate that their His-containing motif is not essential for fast Cu(I) uptake and removal. Other structural features that could be important for these functionally relevant in vitro properties are discussed.


Subject(s)
Bacterial Proteins , Methylosinus trichosporium , Bacterial Proteins/chemistry , Copper/chemistry , Methylosinus trichosporium/chemistry , Methylosinus trichosporium/metabolism
4.
mBio ; 12(6): e0220621, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34749527

ABSTRACT

Xyloglucan utilization by Ruminiclostridium cellulolyticum was formerly shown to imply the uptake of large xylogluco-oligosaccharides, followed by cytosolic depolymerization into glucose, galactose, xylose, and cellobiose. This raises the question of how the anaerobic bacterium manages the simultaneous presence of multiple sugars. Using genetic and biochemical approaches targeting the corresponding metabolic pathways, we observed that, surprisingly, all sugars are catabolized, collectively, but glucose consumption is prioritized. Most selected enzymes display unusual features, especially the GTP-dependent hexokinase of glycolysis, which appeared reversible and crucial for xyloglucan utilization. In contrast, mutant strains lacking either galactokinase, cellobiose-phosphorylase, or xylulokinase still catabolize xyloglucan but display variably altered growth. Furthermore, the xylogluco-oligosaccharide depolymerization process appeared connected to the downstream pathways through an intricate network of competitive and noncompetitive inhibitions. Altogether, our data indicate that xyloglucan utilization by R. cellulolyticum relies on an energy-saving central carbon metabolism deviating from current bacterial models, which efficiently prevents carbon overflow. IMPORTANCE The study of the decomposition of recalcitrant plant biomass is of great interest as the limiting step of terrestrial carbon cycle and to produce plant-derived valuable chemicals and energy. While extracellular cellulose degradation and catabolism have been studied in detail, few publications describe the complete metabolism of hemicelluloses and, to date, the published models are limited to the extracellular degradation and sequential entry of simple sugars. Here, we describe how the model anaerobic bacterium Ruminiclostridium cellulolyticum deals with the synchronous intracellular release of glucose, galactose, xylose, and cellobiose upon cytosolic depolymerization of imported xyloglucan oligosaccharides. The described novel metabolic strategy involves the simultaneous activity of different metabolic pathways coupled to a network of inhibitions controlling the carbon flux and is distinct from the ubiquitously observed sequential uptake and metabolism of carbohydrates known as the diauxic shift. Our results highlight the diversity of cellular responses related to a complex environment.


Subject(s)
Firmicutes/metabolism , Glucans/metabolism , Xylans/metabolism , Anaerobiosis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cellobiose/metabolism , Firmicutes/genetics , Firmicutes/growth & development , Glucose/metabolism , Hexokinase/genetics , Hexokinase/metabolism , Metabolic Networks and Pathways , Polysaccharides/metabolism
5.
Metab Eng Commun ; 12: e00157, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33457204

ABSTRACT

The necessity to decrease our fossil energy dependence requests bioprocesses based on biomass degradation. Cellobiose is the main product released by cellulases when acting on the major plant cell wall polysaccharide constituent, the cellulose. Escherichia coli, one of the most common model organisms for the academy and the industry, is unable to metabolize this disaccharide. In this context, the remodeling of E. coli to catabolize cellobiose should thus constitute an important progress for the design of such applications. Here, we developed a robust E. coli strain able to metabolize cellobiose by integration of a small set of modifications in its genome. Contrary to previous studies that use adaptative evolution to achieve some growth on this sugar by reactivating E. coli cryptic operons coding for cellobiose metabolism, we identified easily insertable modifications impacting the cellobiose import (expression of a gene coding a truncated variant of the maltoporin LamB, modification of the expression of lacY encoding the lactose permease) and its intracellular degradation (genomic insertion of a gene encoding either a cytosolic ß-glucosidase or a cellobiose phosphorylase). Taken together, our results provide an easily transferable set of mutations that confers to E. coli an efficient growth phenotype on cellobiose (doubling time of 2.2 â€‹h in aerobiosis) without any prior adaptation.

6.
Biotechnol Bioeng ; 117(3): 626-636, 2020 03.
Article in English | MEDLINE | ID: mdl-31814100

ABSTRACT

Cellulosomes are large plant cell wall degrading complexes secreted by some anaerobic bacteria. They are typically composed of a major scaffolding protein containing multiple receptors called cohesins, which tightly anchor a small complementary module termed dockerin harbored by the cellulosomal enzymes. In the present study, we have successfully cell surface exposed in Escherichia coli a hybrid scaffoldin, Scaf6, fused to the curli protein CsgA, the latter is known to polymerize at the surface of E. coli to form extracellular fibers under stressful environmental conditions. The C-terminal part of the chimera encompasses the hybrid scaffoldin composed of three cohesins from different bacterial origins and a carbohydrate-binding module targeting insoluble cellulose. Using three cellulases hosting the complementary dockerin modules and labeled with different fluorophores, we have shown that the hybrid scaffoldin merged to CsgA is massively exposed at the cell surface of E. coli and that each cohesin module is fully operational. Altogether these data open a new route for a series of biotechnological applications exploiting the cell-surface exposure of CsgA-Scaf6 in various industrial sectors such as vaccines, biocatalysts or bioremediation, simply by grafting the small dockerin module to the desired proteins before incubation with the engineered E. coli.


Subject(s)
Escherichia coli Proteins , Membrane Proteins , Cell Cycle Proteins , Cellulase/genetics , Cellulosomes/chemistry , Cellulosomes/genetics , Cellulosomes/metabolism , Chromosomal Proteins, Non-Histone , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Cohesins
7.
FEBS J ; 287(12): 2544-2559, 2020 06.
Article in English | MEDLINE | ID: mdl-31769922

ABSTRACT

Cellulosomes are complex nanomachines produced by cellulolytic anaerobic bacteria such as Ruminiclostridium cellulolyticum (formerly known as Clostridium cellulolyticum). Cellulosomes are composed of a scaffoldin protein displaying several cohesin modules on which enzymatic components can bind to through their dockerin module. Although cellulosomes have been studied for decades, very little is known about the dynamics of complex assembly. We have investigated the ability of some dockerin-bearing enzymes to chase the catalytic subunits already bound onto a miniscaffoldin displaying a single cohesin. The stability of the preassembled enzyme-scaffoldin complex appears to depend on the nature of the dockerin, and we have identified a key position in the dockerin sequence that is involved in the stability of the complex with the cohesin. Depending on the residue occupying this position, the dockerin can establish with the cohesin partner either a nearly irreversible or a reversible interaction, independently of the catalytic domain associated with the dockerin. Site-directed mutagenesis of this residue can convert a dockerin able to form a highly stable complex with the miniscaffoldin into a reversible complex forming one and vice versa. We also show that refunctionalization can occur with natural purified cellulosomes. Altogether, our results shed light on the dynamics of cellulosomes, especially their capacity to be remodeled even after their assembly is 'achieved', suggesting an unforeseen adaptability of their enzymatic composition over time.


Subject(s)
Cellulosomes/metabolism , Clostridium cellulolyticum/chemistry , Multienzyme Complexes/metabolism , Biocatalysis , Catalytic Domain , Clostridium cellulolyticum/metabolism
8.
Biotechnol Biofuels ; 12: 208, 2019.
Article in English | MEDLINE | ID: mdl-31497068

ABSTRACT

BACKGROUND: In anaerobic cellulolytic micro-organisms, cellulolysis results in the action of several cellulases gathered in extracellular multi-enzyme complexes called cellulosomes. Their action releases cellobiose and longer cellodextrins which are imported and further degraded in the cytosol to fuel the cells. In Ruminiclostridium cellulolyticum, an anaerobic and cellulolytic mesophilic bacteria, three cellodextrin phosphorylases named CdpA, CdpB, and CdpC, were identified in addition to the cellobiose phosphorylase (CbpA) previously characterized. The present study aimed at characterizing them, exploring their implication during growth on cellulose to better understand the life-style of cellulolytic bacteria on such substrate. RESULTS: The three cellodextrin phosphorylases from R. cellulolyticum displayed marked different enzymatic characteristics. They are specific for cellodextrins of different lengths and present different k cat values. CdpC is the most active enzyme before CdpA, and CdpB is weakly active. Modeling studies revealed that a mutation of a conserved histidine residue in the phosphate ion-binding pocket in CdpB and CdpC might explain their activity-level differences. The genes encoding these enzymes are scattered over the chromosome of R. cellulolyticum and only the expression of the gene encoding the cellobiose phosphorylase and the gene cdpA is induced during cellulose growth. Characterization of four independent mutants constructed in R. cellulolyticum for each of the cellobiose and cellodextrin phosphorylases encoding genes indicated that only the cellobiose phosphorylase is essential for growth on cellulose. CONCLUSIONS: Unexpectedly, the cellobiose phosphorylase but not the cellodextrin phosphorylases is essential for the growth of the model bacterium on cellulose. This suggests that the bacterium adopts a "short" dextrin strategy to grow on cellulose, even though the use of long cellodextrins might be more energy-saving. Our results suggest marked differences in the cellulose catabolism developed among cellulolytic bacteria, which is a result that might impact the design of future engineered strains for biomass-to-biofuel conversion.

9.
FEBS J ; 286(17): 3359-3373, 2019 09.
Article in English | MEDLINE | ID: mdl-31004451

ABSTRACT

Ruminiclostridium cellulolyticum and Lachnoclostridium phytofermentans are cellulolytic clostridia either producing extracellular multienzymatic complexes termed cellulosomes or secreting free cellulases respectively. In the free state, the cellulase Cel9A secreted by L. phytofermentans is much more active on crystalline cellulose than any cellulosomal family-9 enzyme produced by R. cellulolyticum. Nevertheless, the incorporation of Cel9A in vitro in hybrid cellulosomes was formerly shown to generate artificial complexes with altered activity, whereas its incorporation in vivo in native R. cellulolyticum cellulosomes resulted in a strain displaying a weakened cellulolytic phenotype. In this study, we investigated why Cel9A is so potent in the free state but functions poorly as a cellulosomal component, in contrast to the most similar enzyme synthesized by R. cellulolyticum, Cel9G, weakly active in the free state but whose activity on crystalline cellulose is drastically increased in cellulosomes. We show that the removal of the C-terminal moiety of Cel9A encompassing the two X2 modules and the family-3b carbohydrate binding module (CBM3b), reduces its activity on crystalline cellulose. Grafting a dockerin module further diminishes the activity, but this truncated cellulosomal form of Cel9A displays important synergies in hybrid cellulosomes with the pivotal family-48 cellulosomal enzyme of R. cellulolyticum. The exact inverse approach was applied to the cellulosomal Cel9G. Grafting the two X2 modules and the CBM3b of Cel9A to Cel9G strongly increases its activity on crystalline cellulose, to reach Cel9A activity levels. Altogether these data emphasize the specific features required to generate an efficient free or cellulosomal family-9 cellulase.


Subject(s)
Bacterial Proteins/metabolism , Cellulases/metabolism , Cellulosomes/metabolism , Clostridiales/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Cellulases/chemistry , Cellulases/genetics , Cellulose/metabolism , Clostridiales/genetics , Protein Binding
10.
FEBS Lett ; 592(2): 190-198, 2018 01.
Article in English | MEDLINE | ID: mdl-29282732

ABSTRACT

Ruminiclostridium cellulolyticum produces extracellular cellulosomes which contain interalia numerous family-9 glycoside hydrolases, including the inactive Cel9V. The latter shares the same organization and 79% sequence identity with the active cellulase Cel9E. Nevertheless, two aromatic residues and a four-residue stretch putatively critical for the activity are missing in Cel9V. Introduction of one Trytophan and the four-residue stretch restored some weak activity in Cel9V, whereas the replacement of its catalytic domain by that of Cel9E generated a fully active cellulase. Altogether our data indicate that a series of mutations in the catalytic domain of Cel9V lead to an essentially inactive cellulase.


Subject(s)
Cellulase/genetics , Cellulase/metabolism , Clostridium cellulolyticum/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cellulase/chemistry , Enzyme Activation , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Sequence Homology, Amino Acid , Tryptophan/metabolism
11.
Sci Rep ; 6: 39065, 2016 12 19.
Article in English | MEDLINE | ID: mdl-27991525

ABSTRACT

Bacteria are thought to avoid using the essential metal ion copper in their cytosol due to its toxicity. Herein we characterize Csp3, the cytosolic member of a new family of bacterial copper storage proteins from Methylosinus trichosporium OB3b and Bacillus subtilis. These tetrameric proteins possess a large number of Cys residues that point into the cores of their four-helix bundle monomers. The Csp3 tetramers can bind a maximum of approximately 80 Cu(I) ions, mainly via thiolate groups, with average affinities in the (1-2) × 1017 M-1 range. Cu(I) removal from these Csp3s by higher affinity potential physiological partners and small-molecule ligands is very slow, which is unexpected for a metal-storage protein. In vivo data demonstrate that Csp3s prevent toxicity caused by the presence of excess copper. Furthermore, bacteria expressing Csp3 accumulate copper and are able to safely maintain large quantities of this metal ion in their cytosol. This suggests a requirement for storing copper in this compartment of Csp3-producing bacteria.


Subject(s)
Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Copper/metabolism , Methylosinus trichosporium/metabolism , Bacillus subtilis/drug effects , Binding Sites , Copper/toxicity , Crystallography, X-Ray , Cytosol/chemistry , Cytosol/metabolism , Gene Expression , Methylosinus trichosporium/drug effects , Models, Molecular , Protective Agents/chemistry , Protective Agents/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Secondary
12.
Nature ; 525(7567): 140-3, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26308900

ABSTRACT

Methane-oxidizing bacteria (methanotrophs) require large quantities of copper for the membrane-bound (particulate) methane monooxygenase. Certain methanotrophs are also able to switch to using the iron-containing soluble methane monooxygenase to catalyse methane oxidation, with this switchover regulated by copper. Methane monooxygenases are nature's primary biological mechanism for suppressing atmospheric levels of methane, a potent greenhouse gas. Furthermore, methanotrophs and methane monooxygenases have enormous potential in bioremediation and for biotransformations producing bulk and fine chemicals, and in bioenergy, particularly considering increased methane availability from renewable sources and hydraulic fracturing of shale rock. Here we discover and characterize a novel copper storage protein (Csp1) from the methanotroph Methylosinus trichosporium OB3b that is exported from the cytosol, and stores copper for particulate methane monooxygenase. Csp1 is a tetramer of four-helix bundles with each monomer binding up to 13 Cu(I) ions in a previously unseen manner via mainly Cys residues that point into the core of the bundle. Csp1 is the first example of a protein that stores a metal within an established protein-folding motif. This work provides a detailed insight into how methanotrophs accumulate copper for the oxidation of methane. Understanding this process is essential if the wide-ranging biotechnological applications of methanotrophs are to be realized. Cytosolic homologues of Csp1 are present in diverse bacteria, thus challenging the dogma that such organisms do not use copper in this location.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Copper/metabolism , Methane/metabolism , Methylosinus trichosporium/chemistry , Amino Acid Motifs , Crystallography, X-Ray , Cytosol/metabolism , Methane/chemistry , Methylosinus trichosporium/enzymology , Models, Molecular , Oxidation-Reduction , Oxygenases/metabolism , Protein Folding , Protein Structure, Secondary
13.
Front Microbiol ; 6: 606, 2015.
Article in English | MEDLINE | ID: mdl-26167158

ABSTRACT

The ability to respire sulfate linked to lactate oxidation is a key metabolic signature of the Desulfovibrio genus. Lactate oxidation by these incomplete oxidizers generates reductants through lactate dehydrogenase (LDH) and pyruvate-ferredoxin oxidoreductase (PFOR), with the latter catalyzing pyruvate conversion into acetyl-CoA. Acetyl-CoA is the source of substrate-level phosphorylation through the production of ATP. Here, we show that these crucial steps are performed by enzymes encoded by a nonacistronic transcriptional unit named now as operon luo (for lactate utilization operon). Using a combination of genetic and biochemical techniques, we assigned a physiological role to the operon genes DVU3027-28 and DVU3032-33. The growth of mutant Δ26-28 was highly disrupted on D-lactate, whereas the growth of mutant Δ32-33 was slower on L-lactate, which could be related to a decrease in the activity of D-lactate or L-lactate oxidase in the corresponding mutants. The DVU3027-28 and DVU3032-33 genes thus encode functional D-LDH and L-LDH enzymes, respectively. Scanning of the genome for lactate utilization revealed several lactate permease and dehydrogenase homologs. However, transcriptional compensation was not observed in any of the mutants except for lactate permease. Although there is a high degree of redundancy for lactate oxidase, it is not functionally efficient in LDH mutants. This result could be related to the identification of several operon enzymes, including LDHs, in the PFOR activity bands, suggesting the occurrence of a lactate-oxidizing supermolecular structure that can optimize the performance of lactate utilization in Desulfovibrio species.

14.
Anal Chem ; 85(5): 2891-8, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23360365

ABSTRACT

New information on a protein's structure, intra- and intermolecular hydrogen bonds, or metal-ligand bond properties can be unraveled in the far-infrared (far-IR)-terahertz-domain (600-3 cm(-1) or 18-0.1 THz). In this study, we compare the performances of thermal sources with synchrotron far-IR to record reaction-induced Fourier transform infrared (FT-IR) difference signals with proteins in solution. Using the model protein Cu-azurin placed in a short path length electrochemical cell adapted for transmission spectroscopy in vacuum-purged optics, we show that minute spectral shifts induced by metal isotope labeling or temperature changes are detected using the far-IR beamline AILES of the synchrotron SOLEIL. On one hand, these data allow us to identify modes involving Cu-ligand vibrations and pave the way for the analysis of metal sites or metal redox states of proteins not amenable to resonance Raman spectroscopy. On another hand, small band shifts or changes in band intensity upon temperature modifications show that far-IR difference spectroscopy allows one to extract from a complex background hydrogen-bonding signatures directly relevant to the protein function. For Cu-azurin, a temperature-sensitive IR mode involving Cu(II)-His vibrations points to the role of a hydrogen bond between a Cu histidine ligand and the water solvent in tuning the Cu(II)-histidine bond properties. Furthermore, these experimental data support the possible role of a His117-water interaction in electron-transfer activity of Cu-azurin proposed by theoretical studies.


Subject(s)
Metalloproteins/chemistry , Spectroscopy, Fourier Transform Infrared/instrumentation , Synchrotrons , Electrochemistry , Hydrogen Bonding , Ligands , Models, Molecular , Protein Conformation , Signal-To-Noise Ratio , Temperature , Vacuum
15.
J Biol Chem ; 287(3): 1688-97, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22128175

ABSTRACT

Cytoplasmic desulfothioredoxin (Dtrx) from the anaerobe Desulfovibrio vulgaris Hildenborough has been identified as a new member of the thiol disulfide oxidoreductase family. The active site of Dtrx contains a particular consensus sequence, CPHC, never seen in the cytoplasmic thioredoxins and generally found in periplasmic oxidases. Unlike canonical thioredoxins (Trx), Dtrx does not present any disulfide reductase activity, but it presents instead an unusual disulfide isomerase activity. We have used NMR spectroscopy to gain insights into the structure and the catalytic mechanism of this unusual Dtrx. The redox potential of Dtrx (-181 mV) is significantly less reducing than that of canonical Trx. A pH dependence study allowed the determination of the pK(a) of all protonable residues, including the cysteine and histidine residues. Thus, the pK(a) values for the thiol group of Cys(31) and Cys(34) are 4.8 and 11.3, respectively. The His(33) pK(a) value, experimentally determined for the first time, differs notably as a function of the redox states, 7.2 for the reduced state and 4.6 for the oxidized state. These data suggest an important role for His(33) in the molecular mechanism of Dtrx catalysis that is confirmed by the properties of mutant DtrxH33G protein. The NMR structure of Dtrx shows a different charge repartition compared with canonical Trx. The results presented are likely indicative of the involvement of this protein in the catalysis of substrates specific of the anaerobe cytoplasm of DvH. The study of Dtrx is an important step toward revealing the molecular details of the thiol-disulfide oxidoreductase catalytic mechanism.


Subject(s)
Bacterial Proteins/chemistry , Desulfovibrio/enzymology , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Protein Disulfide-Isomerases/chemistry , Amino Acid Substitution , Bacterial Proteins/genetics , Catalysis , Desulfovibrio/genetics , Mutation, Missense , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/genetics , Protein Disulfide-Isomerases/genetics , Protein Structure, Tertiary , Structure-Activity Relationship
16.
Angew Chem Int Ed Engl ; 50(35): 8062-6, 2011 Aug 22.
Article in English | MEDLINE | ID: mdl-21751310

ABSTRACT

Vibrations of the metal active site of the Cu,Zn-superoxide dismutase enzyme were analyzed by far-infrared difference spectroscopy and theoretical normal mode calculation. Both electrochemically triggered Cu(I) and Cu(II) redox states show well-defined infrared vibrational modes, notably modes of the histidine ligands, the Cu(II)-His(61)-Zn(II) bridge and of the water pseudo-ligand.


Subject(s)
Superoxide Dismutase/chemistry , Catalytic Domain , Copper/chemistry , Histidine/chemistry , Ligands , Oxidation-Reduction , Spectroscopy, Fourier Transform Infrared , Superoxide Dismutase/metabolism , Water/chemistry
17.
J Biol Chem ; 286(10): 7812-7821, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21199874

ABSTRACT

Sulfate reducers have developed a multifaceted adaptative strategy to survive against oxidative stresses. Along with this oxidative stress response, we recently characterized an elegant reversible disulfide bond-dependent protective mechanism in the pyruvate:ferredoxin oxidoreductase (PFOR) of various Desulfovibrio species. Here, we searched for thiol redox systems involved in this mechanism. Using thiol fluorescent labeling, we show that glutathione is not the major thiol/disulfide balance-controlling compound in four different Desulfovibrio species and that no other plentiful low molecular weight thiol can be detected. Enzymatic analyses of two thioredoxins (Trxs) and three thioredoxin reductases allow us to propose the existence of two independent Trx systems in Desulfovibrio vulgaris Hildenborough (DvH). The TR1/Trx1 system corresponds to the typical bacterial Trx system. We measured a TR1 apparent K(m) value for Trx1 of 8.9 µM. Moreover, our results showed that activity of TR1 was NADPH-dependent. The second system named TR3/Trx3 corresponds to an unconventional Trx system as TR3 used preferentially NADH (K(m) for NADPH, 743 µM; K(m) for NADH, 5.6 µM), and Trx3 was unable to reduce insulin. The K(m) value of TR3 for Trx3 was 1.12 µM. In vitro experiments demonstrated that the TR1/Trx1 system was the only one able to reactivate the oxygen-protected form of Desulfovibrio africanus PFOR. Moreover, ex vivo pulldown assays using the mutant Trx1(C33S) as bait allowed us to capture PFOR from the DvH extract. Altogether, these data demonstrate that PFOR is a new target for Trx1, which is probably involved in the protective switch mechanism of the enzyme.


Subject(s)
Bacterial Proteins/metabolism , Desulfovibrio vulgaris/metabolism , Pyruvate Synthase/metabolism , Thioredoxins/metabolism , Anaerobiosis/physiology , Bacterial Proteins/genetics , Desulfovibrio vulgaris/genetics , Disulfides/metabolism , Mutation , NAD/genetics , NAD/metabolism , Oxidation-Reduction , Pyruvate Synthase/genetics , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism , Thioredoxins/genetics
18.
Biochemistry ; 47(3): 957-64, 2008 Jan 22.
Article in English | MEDLINE | ID: mdl-18161989

ABSTRACT

Oxidative decarboxylation of pyruvate forming acetyl-coenzyme A is a crucial step in many metabolic pathways. In most anaerobes, this reaction is carried out by pyruvate-ferredoxin oxidoreductase (PFOR), an enzyme normally oxygen sensitive except in Desulfovibrio africanus (Da), where it shows an abnormally high oxygen stability. Using site-directed mutagenesis, we have specified a disulfide bond-dependent protective mechanism against oxidative conditions in Da PFOR. Our data demonstrated that the two cysteine residues forming the only disulfide bond in the as-isolated PFOR are crucial for the stability of the enzyme in oxidative conditions. A methionine residue located in the environment of the proximal [4Fe-4S] cluster was also found to be essential for this protective mechanism. In vivo analysis demonstrated unambiguously that PFOR in Da cells as well as two other Desulfovibrio species was efficiently protected against oxidative stress. Importantly, a less active but stable Da PFOR in oxidized cells rapidly reactivated when returned to anaerobic medium. Our work demonstrates the existence of an elegant disulfide bond-dependent reversible mechanism, found in the Desulfovibrio species to protect one of the key enzymes implicated in the central metabolism of these strict anaerobes. This new mechanism could be considered as an adaptation strategy used by sulfate-reducing bacteria to cope with temporary oxidative conditions and to maintain an active dormancy.


Subject(s)
Desulfovibrio/enzymology , Disulfides/metabolism , Oxidative Stress/physiology , Pyruvate Synthase/metabolism , Adaptation, Physiological , Amino Acid Sequence , Amino Acid Substitution , Anaerobiosis , Catalysis/drug effects , Clostridium acetobutylicum/drug effects , Clostridium acetobutylicum/enzymology , Culture Media, Conditioned/chemistry , Cysteine/genetics , Cysteine/metabolism , Cystine/metabolism , Desulfovibrio/drug effects , Desulfovibrio/genetics , Desulfovibrio africanus/drug effects , Desulfovibrio africanus/enzymology , Desulfovibrio africanus/genetics , Desulfovibrio desulfuricans/drug effects , Desulfovibrio desulfuricans/enzymology , Desulfovibrio vulgaris/drug effects , Desulfovibrio vulgaris/enzymology , Dithioerythritol/pharmacology , Enzyme Activation/drug effects , Enzyme Stability , Hydrogen Peroxide/pharmacology , Molecular Sequence Data , Oxidation-Reduction/drug effects , Oxygen/analysis , Oxygen/pharmacology , Pyruvate Synthase/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...