Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 340
Filter
1.
Biosci Rep ; 40(7)2020 07 31.
Article in English | MEDLINE | ID: mdl-32614053

ABSTRACT

Dihydropyrimidine dehydrogenase (PydA) catalyzes the first step of the reductive pyrimidine degradation (Pyd) pathway in bacteria and eukaryotes, enabling pyrimidines to be utilized as substrates for growth. PydA homologs studied to date catalyze the reduction of uracil to dihydrouracil, coupled to the oxidation of NAD(P)H. Uracil reduction occurs at a flavin mononucleotide (FMN) site, and NAD(P)H oxidation occurs at a flavin adenine dinucleotide (FAD) site, with two ferredoxin domains thought to mediate inter-site electron transfer. Here, we report the biochemical characterization of a Clostridial PydA homolog (PydAc) from a Pyd gene cluster in the strict anaerobic bacterium Clostridium chromiireducens. PydAc lacks the FAD domain, and instead is able to catalyze uracil reduction using reduced methyl viologen or reduced ferredoxin as the electron source. Homologs of PydAc are present in Pyd gene clusters in many strict anaerobic bacteria, which use reduced ferredoxin as an intermediate in their energy metabolism.


Subject(s)
Clostridium/enzymology , Dihydrouracil Dehydrogenase (NADP)/metabolism , Ferredoxins/metabolism , Cloning, Molecular , Dihydrouracil Dehydrogenase (NADP)/genetics , Dihydrouracil Dehydrogenase (NADP)/isolation & purification , Electron Transport , Enzyme Assays , Ferredoxins/genetics , Ferredoxins/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Uracil/metabolism
2.
Nat Commun ; 10(1): 3566, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31395877

ABSTRACT

Iron-sulfur (Fe-S) clusters are essential protein cofactors whose biosynthetic defects lead to severe diseases among which is Friedreich's ataxia caused by impaired expression of frataxin (FXN). Fe-S clusters are biosynthesized on the scaffold protein ISCU, with cysteine desulfurase NFS1 providing sulfur as persulfide and ferredoxin FDX2 supplying electrons, in a process stimulated by FXN but not clearly understood. Here, we report the breakdown of this process, made possible by removing a zinc ion in ISCU that hinders iron insertion and promotes non-physiological Fe-S cluster synthesis from free sulfide in vitro. By binding zinc-free ISCU, iron drives persulfide uptake from NFS1 and allows persulfide reduction into sulfide by FDX2, thereby coordinating sulfide production with its availability to generate Fe-S clusters. FXN stimulates the whole process by accelerating persulfide transfer. We propose that this reconstitution recapitulates physiological conditions which provides a model for Fe-S cluster biosynthesis, clarifies the roles of FDX2 and FXN and may help develop Friedreich's ataxia therapies.


Subject(s)
Ferredoxins/metabolism , Iron-Binding Proteins/metabolism , Iron-Sulfur Proteins/metabolism , Sulfides/metabolism , Carbon-Sulfur Lyases/metabolism , Ferredoxins/isolation & purification , Friedreich Ataxia/pathology , Iron/metabolism , Iron-Binding Proteins/isolation & purification , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Proton Magnetic Resonance Spectroscopy , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Zinc/metabolism , Frataxin
3.
Biotechnol Bioeng ; 116(9): 2316-2329, 2019 09.
Article in English | MEDLINE | ID: mdl-31161644

ABSTRACT

The Rnf complex is a membrane-bound ferredoxin(Fd):NAD(P)+ oxidoreductase (Fno) that couples Fd oxidation to vectorial H+ /Na+ transport across the cytoplasmic membrane. Here, we produced two putative Rnf-complexes from Clostridioides difficile (Cd-Rnf) and Clostridium ljungdahlii (Cl-Rnf) for the first time in Escherichia coli. A redox-responsive low-expression system enabled Rnf assembly in the membranes of E. coli as confirmed by in vitro activity measurements. To study the physiological effects of Rnf on the metabolism of E. coli, we assembled additional Fd-dependent enzymes by plasmid-based multigene expression: (a) an Fd-linked butyrate pathway (But) from C. difficile, (b) an [FeFe]-hydrogenase (Hyd) to modulate the redox state of Fd, and (c) heterologous ferredoxins as electron carriers. The hydrogenase efficiently modulated butyrate formation by H2 -mediated Fd reoxidation under nitrogen. In its functionally assembled state, Rnf severely impaired cell growth. Including Hyd in the But/Rnf background, in turn, restored normal growth. Our findings suggest that Rnf mediates reverse electron flow from NADH to Fd, which requires E. coli's F-type ATPase to function in its reverse, ATP hydrolyzing direction. The reduced Fd is then reoxidized by endogenous Fd:NAD(P)H oxidoreductase (Fpr), which regenerates NADH and, thereby, initiates a futile cycle fueled by ATP hydrolysis. The introduction of hydrogenase interrupts this futile cycle under N2 by providing an efficient NAD(P)+ -independent Fd reoxidation route, whereas under H2 , Hyd outcompetes Rnf for Fd reduction. This is the first report of an Rnf complex being functionally produced and physiologically investigated in E. coli.


Subject(s)
Bacterial Proteins/isolation & purification , Clostridioides difficile/enzymology , Clostridium/enzymology , Ferredoxin-NADP Reductase/chemistry , Ferredoxins/isolation & purification , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Clostridioides difficile/genetics , Clostridium/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Ferredoxin-NADP Reductase/genetics , Ferredoxins/chemistry , Ferredoxins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
4.
Protein Expr Purif ; 121: 1-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26748213

ABSTRACT

Ferredoxins are small, acidic proteins containing iron-sulfur clusters that are widespread in living organisms. They play key roles as electron carriers in various metabolic processes, including respiration, photosynthesis, fermentation, nitrogen fixation, carbon dioxide fixation, and hydrogen production. However, only several kinds of ferredoxins are commercially available now, greatly limiting the investigation of ferredoxin-related enzymes and metabolic processes. Here we describe the heterologous overproduction of 2[4Fe4S]- and [2Fe2S]-type clostridial ferredoxins and [2Fe2S]-type agrobacterial ferredoxin. Adding extra iron and sulfur sources to the medium in combination with using Escherichia coli C41(DE3) harboring pCodonplus and pRKISC plasmids as host greatly enhanced iron-sulfur cluster synthesis in the three ferredoxins. After induction for 12 h in terrific broth and purification by affinity chromatography and anion exchange chromatography, approximately 3.4 mg of streptavidin (Strep)-tagged and 3.7 mg of polyhistidine (His)-tagged clostridial 2[4Fe4S] ferredoxins were obtained from 1 l of culture. Excitingly, after induction for 24 h in terrific broth, around 40 mg of His-tagged clostridial [2Fe2S] and 23 mg of His-tagged agrobacterial [2Fe2S] ferredoxins were purified from 1 l of culture. The recombinant ferredoxins apparently exhibited identical properties and physiological function to native ferredoxins. No negative impact of two different affinity tags on ferredoxin activity was found. In conclusion, we successfully developed a convenient method for heterologous overproduction of the three kinds of ferredoxins with satisfactory yields and activities, which would be very helpful for the ferredoxin-related researches.


Subject(s)
Ferredoxins/biosynthesis , Iron-Sulfur Proteins/biosynthesis , Recombinant Proteins/biosynthesis , Agrobacterium/genetics , Chromatography, Affinity , Clostridium/genetics , Electron Spin Resonance Spectroscopy , Escherichia coli/genetics , Ferredoxins/chemistry , Ferredoxins/isolation & purification , Iron/chemistry , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/isolation & purification , Plasmids/biosynthesis , Plasmids/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
5.
BMC Microbiol ; 15: 16, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25648224

ABSTRACT

BACKGROUND: Legumes establish with rhizobial bacteria a nitrogen-fixing symbiosis which is of the utmost importance for both plant nutrition and a sustainable agriculture. Calcium is known to act as a key intracellular messenger in the perception of symbiotic signals by both the host plant and the microbial partner. Regulation of intracellular free Ca(2+) concentration, which is a fundamental prerequisite for any Ca(2+)-based signalling system, is accomplished by complex mechanisms including Ca(2+) binding proteins acting as Ca(2+) buffers. In this work we investigated the occurrence of Ca(2+) binding proteins in Mesorhizobium loti, the specific symbiotic partner of the model legume Lotus japonicus. RESULTS: A soluble, low molecular weight protein was found to share several biochemical features with the eukaryotic Ca(2+)-binding proteins calsequestrin and calreticulin, such as Stains-all blue staining on SDS-PAGE, an acidic isoelectric point and a Ca(2+)-dependent shift of electrophoretic mobility. The protein was purified to homogeneity by an ammonium sulfate precipitation procedure followed by anion-exchange chromatography on DEAE-Cellulose and electroendosmotic preparative electrophoresis. The Ca(2+) binding ability of the M. loti protein was demonstrated by (45)Ca(2+)-overlay assays. ESI-Q-TOF MS/MS analyses of the peptides generated after digestion with either trypsin or endoproteinase AspN identified the rhizobial protein as ferredoxin II and confirmed the presence of Ca(2+) adducts. CONCLUSIONS: The present data indicate that ferredoxin II is a major Ca(2+) binding protein in M. loti that may participate in Ca(2+) homeostasis and suggest an evolutionarily ancient origin for protein-based Ca(2+) regulatory systems.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Ferredoxins/metabolism , Mesorhizobium/enzymology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/isolation & purification , Chemical Precipitation , Chromatography, Ion Exchange , Electrophoresis , Ferredoxins/chemistry , Ferredoxins/isolation & purification , Isoelectric Point , Nitrogen Fixation , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
6.
Appl Environ Microbiol ; 81(6): 2182-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25595756

ABSTRACT

Acetochlor [2-chloro-N-(ethoxymethyl)-N-(2-ethyl-6-methylphenyl)-acetamide] is a widely applied herbicide with potential carcinogenic properties. N-Deethoxymethylation is the key step in acetochlor biodegradation. N-Deethoxymethylase is a multicomponent enzyme that catalyzes the conversion of acetochlor to 2'-methyl-6'-ethyl-2-chloroacetanilide (CMEPA). Fast detection of CMEPA by a two-enzyme (N-deethoxymethylase-amide hydrolase) system was established in this research. Based on the fast detection method, a three-component enzyme was purified from Rhodococcus sp. strain T3-1 using ammonium sulfate precipitation and hydrophobic interaction chromatography. The molecular masses of the components of the purified enzyme were estimated to be 45, 43, and 11 kDa by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Based on the results of peptide mass fingerprint analysis, acetochlor N-deethoxymethylase was identified as a cytochrome P450 system, composed of a cytochrome P450 oxygenase (43-kDa component; EthB), a ferredoxin (45 kDa; EthA), and a reductase (11 kDa; EthD), that is involved in the degradation of methyl tert-butyl ether. The gene cluster ethABCD was cloned by PCR amplification and expressed in Escherichia coli BL21(DE3). Resting cells of a recombinant E. coli strain showed deethoxymethylation activity against acetochlor. Subcloning of ethABCD showed that ethABD expressed in E. coli BL21(DE3) has the activity of acetochlor N-deethoxymethylase and is capable of converting acetochlor to CMEPA.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Herbicides/metabolism , Rhodococcus/enzymology , Rhodococcus/metabolism , Toluidines/metabolism , Biotransformation , Chemical Fractionation , Chromatography, Liquid , Cloning, Molecular , Cluster Analysis , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/isolation & purification , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Escherichia coli/metabolism , Ferredoxins/chemistry , Ferredoxins/isolation & purification , Ferredoxins/metabolism , Gene Expression , Molecular Sequence Data , Molecular Weight , Oxidoreductases/chemistry , Oxidoreductases/isolation & purification , Oxidoreductases/metabolism , Phylogeny , Sequence Analysis, DNA , Sequence Homology
7.
J Bacteriol ; 194(5): 932-40, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22178968

ABSTRACT

Although successful iron acquisition by pathogens within a host is a prerequisite for the establishment of infection, surprisingly little is known about the intracellular distribution of iron within bacterial pathogens. We have used a combination of anaerobic native liquid chromatography, inductively coupled plasma mass spectrometry, principal-component analysis, and peptide mass fingerprinting to investigate the cytosolic iron distribution in the pathogen Bacillus anthracis. Our studies identified three of the major iron pools as being associated with the electron transfer protein ferredoxin, the miniferritin Dps2, and the superoxide dismutase (SOD) enzymes SodA1 and SodA2. Although both SOD isozymes were predicted to utilize manganese cofactors, quantification of the metal ions associated with SodA1 and SodA2 in cell extracts established that SodA1 is associated with both manganese and iron, whereas SodA2 is bound exclusively to iron in vivo. These data were confirmed by in vitro assays using recombinant protein preparations, showing that SodA2 is active with an iron cofactor, while SodA1 is cambialistic, i.e., active with manganese or iron. Furthermore, we observe that B. anthracis cells exposed to superoxide stress increase their total iron content more than 2-fold over 60 min, while the manganese and zinc contents are unaffected. Notably, the acquired iron is not localized to the three identified cytosolic iron pools.


Subject(s)
Bacillus anthracis/chemistry , Cytosol/chemistry , Iron/analysis , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Chromatography, Liquid , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Ferredoxins/isolation & purification , Ferredoxins/metabolism , Mass Spectrometry , Peptide Mapping , Protein Binding , Superoxide Dismutase/isolation & purification , Superoxide Dismutase/metabolism
8.
J Mol Biol ; 413(5): 940-51, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21945528

ABSTRACT

Ferredoxins are ubiquitous proteins with electron transfer activity involved in a variety of biological processes. In this work, we investigated the characteristics and function of Fdx1 from Sorangium cellulosum So ce56 by using a combination of bioinformatics and of biochemical/biophysical approaches. We were able to experimentally confirm a role of Fdx1 in the iron-sulfur cluster biosynthesis by in vitro reduction studies with cluster-loaded So ce56 IscU and by transfer studies of the cluster from the latter protein to apo-aconitase A. Moreover, we found that Fdx1 can replace mammalian adrenodoxin in supporting the activity of bovine CYP11A1. This makes S. cellulosum Fdx1 the first prokaryotic ferredoxin reported to functionally interact with this mammalian enzyme. Although the interaction with CYP11A1 is non-physiological, this is-to the best of our knowledge-the first study to experimentally prove the activity of a postulated ISC-type ferredoxin in both the ISC assembly and a cytochrome P450 system. This proves that a single ferredoxin can be structurally able to provide electrons to both cytochromes P450 and IscU and thus support different biochemical processes. Combining this finding with phylogenetic and evolutionary trace analyses led us to propose the evolution of eukaryotic mitochondrial P450-type ferredoxins and ISC-type ferredoxins from a common prokaryotic ISC-type ancestor.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cholesterol Side-Chain Cleavage Enzyme/chemistry , Evolution, Molecular , Ferredoxins/chemistry , Ferredoxins/metabolism , Iron-Sulfur Proteins/chemistry , Mitochondria/enzymology , Aconitate Hydratase/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/isolation & purification , Cattle , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Circular Dichroism , Cloning, Molecular , Computational Biology , Ferredoxins/isolation & purification , Iron/analysis , Iron-Sulfur Proteins/metabolism , Molecular Sequence Data , Myxococcales/genetics , Myxococcales/metabolism , NADP , Oxidation-Reduction , Phylogeny , Sequence Homology, Amino Acid , Sulfite Reductase (Ferredoxin)/metabolism
9.
Bioelectrochemistry ; 82(1): 22-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21600857

ABSTRACT

The redox behaviour of a ferredoxin (Fd) from Desulfovibrio alaskensis was characterized by electrochemistry. The protein was isolated and purified, and showed to be a tetramer containing one [3Fe-4S] and one [4Fe-4S] centre. This ferredoxin has high homology with FdI from Desulfovibrio vulgaris Miyazaki and Hildenborough and FdIII from Desulfovibrio africanus. From differential pulse voltammetry the following signals were identified: [3Fe-4S](+1/0) (E(0')=-158±5mV); [4Fe-4S](+2/+1) (E(0')=-474±5mV) and [3Fe-4S](0/-2) (E(0')=-660±5mV). The effect of pH on these signals showed that the reduced [3Fe-4S](0) cluster has a pK'(red)(')=5.1±0.1, the [4Fe-4S](+2/+1) centre is pH independent, and the [3Fe-4S](0/-2) reduction is accompanied by the binding of two protons. The ability of the [3Fe-4S](0) cluster to be converted into a new [4Fe-4S] cluster was proven. The redox potential of the original [4Fe-4S] centre showed to be dependent on the formation of the new [4Fe-4S] centre, which results in a positive shift (ca. 70mV) of the redox potential of the original centre. Being most [Fe-S] proteins involved in electron transport processes, the electrochemical characterization of their clusters is essential to understand their biological function. Complementary EPR studies were performed.


Subject(s)
Bacterial Proteins/chemistry , Desulfovibrio/chemistry , Ferredoxins/chemistry , Bacterial Proteins/isolation & purification , Electrochemical Techniques , Electron Spin Resonance Spectroscopy , Electron Transport , Ferredoxins/isolation & purification , Oxidation-Reduction
10.
J Biol Inorg Chem ; 16(5): 763-75, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21484348

ABSTRACT

The structure of the all-cysteinyl-coordinated D14C variant of [4Fe-4S] ferredoxin from the hyperthermophilic archaeon Pyrococcus furiosus has been determined to 1.7 Å resolution from a crystal belonging to space group C222(1) with two types of molecules, A and B, in the asymmetric unit. A and B molecules have different crystal packing and intramolecular disulfide bond conformation. The crystal packing reveals a ß-sheet interaction between A molecules in adjacent asymmetric units, whereas B molecules are packed as monomers in a less rigid position next to the A-A extended ß-sheet dimers. The A molecules contain an intramolecular disulfide bond in a double conformation with 60% occupancy left-handed and 40% occupancy right-handed spiral conformation, whereas B molecules have an intramolecular disulfide bond in a right-handed spiral conformation. The cluster in D14C [4Fe-4S] P. furiosus ferredoxin was chemically oxidized at pH 5.8 to [3Fe-4S]. For purification at pH 8.0, two forms of the protein are obtained. Mass spectrometric analysis shows that the two forms are the D14C [3Fe-4S] P. furiosus ferredoxin monomer and a disulfide-bonded dimer of D14C [3Fe-4S] P. furiosus ferredoxin. When oxidization and purification are carried out at pH 5.8, only the monomer is obtained. The crystal structure of D14C [3Fe-4S] P. furiosus ferredoxin monomer was determined to 2.8 Å resolution from a crystal belonging to space group P2(1)2(1)2(1) with two molecules in the asymmetric unit. The molecules resemble molecule A of D14C [4Fe-4S] P. furiosus ferredoxin and electron density clearly shows the presence of a [3Fe-4S] cluster.


Subject(s)
Ferredoxins/chemistry , Ferredoxins/genetics , Mutation , Pyrococcus furiosus/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Cysteine/genetics , Ferredoxins/isolation & purification , Models, Molecular , Molecular Sequence Data , Protein Conformation , Pyrococcus furiosus/genetics , Sequence Alignment
11.
Dalton Trans ; 39(12): 3115-23, 2010 Mar 28.
Article in English | MEDLINE | ID: mdl-20221546

ABSTRACT

Rational design of the active site of cytochrome P450cam has been carried out to catalyse oxygenation of various potentially important chemical reactions. The modeling studies showed that the distal pocket of the heme consisting of the Y96, T101, F87 and L244 residues could be suitably mutated to change the substrate specificity of the enzyme. We found that the mutant enzymes could catalyse oxygenation of indole to produce indigo. While Y96F was found to be several times better as a catalyst for conversion of indole to indigo, the double mutant Y96F/L244A showed the highest NADH oxidation rate as well as yield of indigo. The oxidative catalysis using H(2)O(2) as the oxygen source was found to produce a higher purity of indigo, and lesser or no formation of indirubin was detected. The enzymatic oxygenation of aromatic hydrocarbons such as coumarin and analogues was also found to be enhanced on mutation of Y96 and L244 residues in the enzyme. The studies also showed that mutation of suitable residues can alter the regio-selectivity of hydroxylation of the aromatic hydrocarbons.


Subject(s)
Camphor 5-Monooxygenase , Catalytic Domain , Protein Engineering/methods , Camphor 5-Monooxygenase/chemistry , Camphor 5-Monooxygenase/genetics , Camphor 5-Monooxygenase/metabolism , Coloring Agents/chemical synthesis , Coloring Agents/chemistry , Ferredoxins/genetics , Ferredoxins/isolation & purification , Ferredoxins/metabolism , Hydrogen Peroxide/chemistry , Indigo Carmine , Indoles/chemical synthesis , Indoles/chemistry , Molecular Structure , Mutagenesis, Site-Directed , NAD/chemistry , NAD/metabolism , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/isolation & purification , NADH, NADPH Oxidoreductases/metabolism , Oxidants/chemistry , Oxidation-Reduction , Substrate Specificity
12.
J Biol Inorg Chem ; 15(2): 271-81, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19862563

ABSTRACT

Rieske proteins and Rieske ferredoxins are present in the three domains of life and are involved in a variety of cellular processes. Despite their functional diversity, these small Fe-S proteins contain a highly conserved all-beta fold, which harbors a [2Fe-2S] Rieske center. We have identified a novel subtype of Rieske ferredoxins present in hyperthermophilic archaea, in which a two-cysteine conserved SKTPCX((2-3))C motif is found at the C-terminus. We establish that in the Acidianus ambivalens representative, Rieske ferredoxin 2 (RFd2), these cysteines form a novel disulfide bond within the Rieske fold, which can be selectively broken under mild reducing conditions insufficient to reduce the [2Fe-2S] cluster or affect the secondary structure of the protein, as shown by visible circular dichroism, absorption, and attenuated total reflection Fourier transform IR spectroscopies. RFd2 presents all the EPR, visible absorption, and visible circular dichroism spectroscopic features of the [2Fe-2S] Rieske center. The cluster has a redox potential of +48 mV (25 degrees C and pH 7) and a pK (a) of 10.1 +/- 0.2. These shift to +77 mV and 8.9 +/- 0.3, respectively, upon reduction of the disulfide. RFd2 has a melting temperature near the boiling point of water (T(m) = 99 degrees C, pH 7.0), but it becomes destabilized upon disulfide reduction (DeltaT(m) = -9 degrees C, DeltaC(m) = -0.7 M guanidinium hydrochloride). This example illustrates how the incorporation of an additional structural element such as a disulfide bond in a highly conserved fold such as that of the Rieske domain may fine-tune the protein for a particular function or for increased stability.


Subject(s)
Disulfides/chemistry , Ferredoxins/chemistry , Acidianus/chemistry , Amino Acid Sequence , Cloning, Molecular , Ferredoxins/genetics , Ferredoxins/isolation & purification , Molecular Sequence Data , Oxidation-Reduction , Protein Conformation , Protein Folding , Sequence Alignment , Solubility , Temperature
13.
J Mol Biol ; 392(2): 436-51, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19616558

ABSTRACT

Carbazole 1,9a-dioxygenase (CARDO) consists of terminal oxygenase (Oxy), ferredoxin (Fd), and ferredoxin reductase (Red) components and is a member of the Rieske nonheme iron oxygenases. Rieske nonheme iron oxygenases are divided into five subclasses (IA, IB, IIA, IIB, and III) based on the number of constituents and the nature of their redox centers. Each component of a class IIB CARDO from Nocardioides aromaticivorans IC177 was purified, and the interchangeability of the electron transfer reactions with each component from the class III CARDOs was investigated. Despite the fact that the Fds of both classes are Rieske-type, strict specificities between the Oxy and Fd components were observed. On the other hand, the Fd and Red components were interchangeable, even though the Red components differ in cofactor composition; the class IIB Red contains flavin-adenine-dinucleotide (FAD)- and NADH-binding domains, whereas the class III Red has a chloroplast-type [2Fe-2S] cluster in addition to the FAD- and NADH-binding domains. The crystal structures of the class IIB Oxy and Fd components were compared to the previously reported Fd:Oxy complex structure of class III CARDO. This comparison suggested residues in common between class IIB and class III CARDOs that are important for interactions between Fd and Oxy. In the class IIB CARDOs, these included His75 and Glu71 in Fd and Lys20 and Glu357 in Oxy for electrostatic interactions, and Phe74 and Pro90 in Fd and Trp21, Leu359, and Val367 in Oxy for hydrophobic interactions. The residues that formed the interacting surface but were not conserved between classes were thought to be necessary to form the appropriate geometry and to determine electron transfer specificity between Fd and Oxy.


Subject(s)
Actinomycetales/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Dioxygenases/chemistry , Dioxygenases/metabolism , Protein Interaction Domains and Motifs , Protein Subunits/chemistry , Protein Subunits/metabolism , Bacterial Proteins/isolation & purification , Crystallography, X-Ray , Dioxygenases/isolation & purification , Ferredoxins/chemistry , Ferredoxins/isolation & purification , Ferredoxins/metabolism , Models, Biological , Models, Molecular , Oxygenases/chemistry , Oxygenases/isolation & purification , Oxygenases/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Subunits/isolation & purification
14.
Appl Environ Microbiol ; 75(4): 1021-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19124587

ABSTRACT

Clostridium acetobutylicum, an obligate anaerobe, grows normally under continuous-O(2)-flow culture conditions, where the cells consume O(2) proficiently. An O(2)-responsive NADH:rubredoxin oxidoreductase operon composed of three genes (nror, fprA2, and dsr), encoding NROR, functionally uncharacterized flavoprotein A2 (FprA2), and the predicted superoxide reductase desulfoferrodoxin (Dsr), has been proposed to participate in defense against O(2) stress. To functionally characterize these proteins, native NROR from C. acetobutylicum, recombinant NROR (rNROR), FprA2, Dsr, and rubredoxin (Rd) expressed in Escherichia coli were purified. Purified native NROR and rNROR both exhibited weak H(2)O(2)-forming NADH oxidase activity that was slightly activated by Rd. A mixture of NROR, Rd, and FprA2 functions as an efficient H(2)O-forming NADH oxidase with a high affinity for O(2) (the K(m) for O(2) is 2.9 +/- 0.4 microM). A mixture of NROR, Rd, and Dsr functions as an NADH-dependent O(2)(-) reductase. A mixture of NROR, Rd, and rubperoxin (Rpr, a rubrerythrin homologue) functions as an inefficient H(2)O-forming NADH oxidase but an efficient NADH peroxidase with a low affinity for O(2) and a high affinity for H(2)O(2) (the K(m)s for O(2) and H(2)O(2) are 303 +/- 39 microM and

Subject(s)
Bacterial Proteins/metabolism , Clostridium acetobutylicum/metabolism , Ferredoxins/metabolism , Flavoproteins/metabolism , NADH, NADPH Oxidoreductases/metabolism , Oxidoreductases/metabolism , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Rubredoxins/metabolism , Bacterial Proteins/isolation & purification , Clostridium acetobutylicum/enzymology , Ferredoxins/isolation & purification , Flavoproteins/isolation & purification , Gene Expression Regulation, Bacterial , Hydrogen Peroxide/metabolism , NADH, NADPH Oxidoreductases/isolation & purification , Oxidoreductases/isolation & purification , Rubredoxins/isolation & purification , Transcription, Genetic , Water/metabolism
15.
Eukaryot Cell ; 8(1): 47-55, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19011120

ABSTRACT

Trichomonas vaginalis is one of a few eukaryotes that have been found to encode several homologues of flavodiiron proteins (FDPs). Widespread among anaerobic prokaryotes, these proteins are believed to function as oxygen and/or nitric oxide reductases to provide protection against oxidative/nitrosative stresses and host immune responses. One of the T. vaginalis FDP homologues is equipped with a hydrogenosomal targeting sequence and is expressed in the hydrogenosomes, oxygen-sensitive organelles that participate in carbohydrate metabolism and assemble iron-sulfur clusters. The bacterial homologues characterized thus far have been dimers or tetramers; the trichomonad protein is a dimer of identical 45-kDa subunits, each noncovalently binding one flavin mononucleotide. The protein reduces dioxygen to water but is unable to utilize nitric oxide as a substrate, similarly to its closest homologue from another human parasite Giardia intestinalis and related archaebacterial proteins. T. vaginalis FDP is able to accept electrons derived from pyruvate or NADH via ferredoxin and is proposed to play a role in the protection of hydrogenosomes against oxygen.


Subject(s)
Ferredoxins/metabolism , Organelles/enzymology , Oxidoreductases/metabolism , Oxygen/metabolism , Protozoan Proteins/metabolism , Trichomonas vaginalis/enzymology , Amino Acid Sequence , Animals , Ferredoxins/chemistry , Ferredoxins/genetics , Ferredoxins/isolation & purification , Flavin Mononucleotide/metabolism , Molecular Sequence Data , Organelles/chemistry , Organelles/genetics , Oxidoreductases/chemistry , Oxidoreductases/genetics , Oxidoreductases/isolation & purification , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/isolation & purification , Substrate Specificity , Trichomonas vaginalis/chemistry , Trichomonas vaginalis/genetics
16.
FEBS Lett ; 581(29): 5605-10, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18005665

ABSTRACT

Desulfoferrodoxin (cac2450) of Clostridium acetobutylicum was purified after overexpression in E. coli. In an in vitro assay the enzyme exhibited superoxide reductase activity with rubredoxin (cac2778) of C. acetobutylicum as the proximal electron donor. Rubredoxin was reduced by ferredoxin:NADP(+) reductase from spinach and NADPH. The superoxide anions, generated from dissolved oxygen using Xanthine and Xanthine oxidase, were reduced to hydrogen peroxide. Thus, we assume that desulfoferrodoxin is the key factor in the superoxide reductase dependent part of an alternative pathway for detoxification of reactive oxygen species in this obligate anaerobic bacterium.


Subject(s)
Bacterial Proteins/physiology , Clostridium acetobutylicum/enzymology , Ferredoxins/physiology , Oxidoreductases/physiology , Cytochromes c/metabolism , Ferredoxins/isolation & purification , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Rubredoxins/isolation & purification , Superoxide Dismutase/metabolism , Superoxides/metabolism
17.
FEMS Microbiol Lett ; 277(1): 50-5, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17986084

ABSTRACT

Haloferax mediterranei is a halophilic archaeon that can grow using nitrate as the sole nitrogen source. A ferredoxin that serves as the physiological electron donor to the nitrate and nitrite reductases in this assimilatory process has been characterized. The ferredoxin was found to contain approximately two atoms of iron and two atoms of sulphur, indicative of the binding of a [2Fe-2S] cluster. The electron paramagnetic resonance spectrum of the reduced form of the protein displayed a rhombic signal, with g(x)=1.91, g(y)=1.98, g(z)=2.07, that shows considerable similarity to plant and algal [2Fe-2S] ferredoxins. UV-visible spectropotentiometric analysis determined a midpoint redox potential for the [2Fe-2S](2+/1+) transition of around -285 mV vs. SHE that was independent of salt concentration. UV-visible spectroscopy was also used to establish that the [2Fe-2S] cluster integrity of this protein was maintained over the pH range 5-11. Significantly, the Haloferax mediterranei ferredoxin was shown to be a highly thermostable protein. It was stable up to 60 degrees C in a low-salt (0.2 M) medium and this increased to 80 degrees C in a high-salt (4 M) medium. This thermostability at high salt concentration is an essential physiological characteristic because haloarchaea are mainly found in environments where high temperatures and concentrated salt water occur.


Subject(s)
Electron Spin Resonance Spectroscopy/methods , Ferredoxins/chemistry , Ferredoxins/metabolism , Haloferax mediterranei/enzymology , Hot Temperature , Nitrates/metabolism , Sodium Chloride/pharmacology , Spectrophotometry, Ultraviolet/methods , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Electron Spin Resonance Spectroscopy/instrumentation , Electron Transport , Enzyme Stability , Ferredoxins/isolation & purification , Haloferax mediterranei/growth & development , Hydrogen-Ion Concentration , Oxidation-Reduction , Spectrophotometry, Ultraviolet/instrumentation
18.
Curr Microbiol ; 55(6): 518-23, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17909890

ABSTRACT

The [2Fe-2S] cluster containing ferredoxin has attracted much attention in recent years. Genetic analyses show that it has an essential role in the maturation of various iron-sulfur (Fe-S) proteins and functions as a component of the complex machinery responsible for the biogenesis of Fe-S clusters. The gene of ferredoxin from A. ferrooxidans ATCC 23270 was cloned, successfully expressed in Escherichia coli, and purified by one-step affinity chromatography to homogeneity. The MALDI-TOF MS and spectra results of the recombinant protein confirmed that the iron-sulfur cluster was correctly inserted into the active site of the protein. Site-directed mutagenesis results revealed that Cys42, Cys48, Cys51, and Cys87 were ligating with the [Fe(2)S(2)] cluster of the protein.


Subject(s)
Acidithiobacillus thiooxidans/enzymology , Escherichia coli/enzymology , Ferredoxins , Iron-Sulfur Proteins , Acidithiobacillus thiooxidans/genetics , Acidithiobacillus thiooxidans/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Chromatography, Affinity , Cloning, Molecular , Escherichia coli/genetics , Ferredoxins/chemistry , Ferredoxins/genetics , Ferredoxins/isolation & purification , Ferredoxins/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/isolation & purification , Iron-Sulfur Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
FEMS Microbiol Lett ; 275(1): 113-21, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17681007

ABSTRACT

In Clostridium acetobutylicum, [FeFe]-hydrogenase is involved in hydrogen production in vivo by transferring electrons from physiological electron donors, ferredoxin and flavodoxin, to protons. In this report, by modifications of the purification procedure, the specific activity of the enzyme has been improved and its complete catalytic profile in hydrogen evolution, hydrogen uptake, proton/deuterium exchange and para-H2/ortho-H2 conversion has been determined. The major ferredoxin expressed in the solvent-producing C. acetobutylicum cells was purified and identified as encoded by ORF CAC0303. Clostridium acetobutylicum recombinant holoflavodoxin CAC0587 was also purified. The kinetic parameters of C. acetobutylicum [FeFe]-hydrogenase for both physiological partners, ferredoxin CAC0303 and flavodoxin CAC0587, are reported for hydrogen uptake and hydrogen evolution activities.


Subject(s)
Clostridium acetobutylicum/enzymology , Ferredoxins/chemistry , Flavodoxin/chemistry , Hydrogenase/chemistry , Escherichia coli/genetics , Ferredoxins/isolation & purification , Flavodoxin/biosynthesis , Flavodoxin/isolation & purification , Hydrogen/chemistry , Hydrogen/metabolism , Hydrogenase/isolation & purification , Kinetics , Oxidation-Reduction
20.
Biochem Biophys Res Commun ; 360(3): 666-72, 2007 Aug 31.
Article in English | MEDLINE | ID: mdl-17618912

ABSTRACT

Twelve of the fifteen potential P450 enzymes from the bacterium Novosphingobium aromaticivorans, which is known to degrade a wide range of aromatic hydrocarbons, have been produced via heterologous expression in Escherichia coli. The enzymes were tested for their ability to bind a range of substrates including polyaromatic hydrocarbons. While two of the enzymes were found to bind aromatic compounds (CYP108D1 and CYP203A2), the others show binding with a variety of compounds including linear alkanes (CYP153C1) and mono- and sesqui-terpenoid compounds (CYP101B1, CYP101C1, CYP101D1, CYP101D2, CYP111A1, and CYP219A1). A 2Fe-2S ferredoxin (Arx-A), which is associated with CYP101D2, was also produced. The activity of five of the P450 enzymes (CYP101B1, CYP101C1, CYP101D1, CYP101D2, and CYP111A2) was reconstituted with Arx-A and putidaredoxin reductase (of the P450cam system from Pseudomonas putida) in a Class I type electron transfer system. Preliminary characterisation of the majority of the substrate oxidation products is reported.


Subject(s)
Bacterial Proteins , Cytochrome P-450 Enzyme System , Sphingomonadaceae/enzymology , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Camphor/analogs & derivatives , Camphor/chemistry , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/isolation & purification , Ferredoxins/biosynthesis , Ferredoxins/genetics , Ferredoxins/isolation & purification , Spectrum Analysis , Sphingomonadaceae/genetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...