Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
J Proteome Res ; 17(11): 3704-3718, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30239205

ABSTRACT

Trichomonas vaginalis is a sexually transmitted anaerobic parasite that infects humans causing trichomoniasis, a common and ubiquitous sexually transmitted disease. The life cycle of this parasite possesses a trophozoite form without a cystic stage. However, the presence of nonproliferative and nonmotile, yet viable and reversible spherical forms with internalized flagella, denominated pseudocysts, has been commonly observed for this parasite. To understand the mechanisms involved in the formation of pseudocysts, we performed a mass spectrometry-based high-throughput quantitative proteomics study using a label-free approach and functional assays by biochemical and flow cytometric methods. We observed that the morphological transformation of trophozoite to pseudocysts is coupled to (i) a metabolic shift toward a less glycolytic phenotype; (ii) alterations in the abundance of hydrogenosomal iron-sulfur cluster (ISC) assembly machinery; (iii) increased abundance of regulatory particles of the ubiquitin-proteasome system; (iv) significant alterations in proteins involved in adhesion and cytoskeleton reorganization; and (v) arrest in G2/M phase associated with alterations in the abundance of regulatory proteins of the cell cycle. These data demonstrate that pseudocysts experience important physiological and structural alterations for survival under unfavorable environmental conditions.


Subject(s)
Iron-Sulfur Proteins/chemistry , Life Cycle Stages/genetics , Proteomics/methods , Protozoan Proteins/chemistry , Trichomonas vaginalis/chemistry , Trophozoites/chemistry , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Flagella/chemistry , Flagella/metabolism , Flagella/ultrastructure , G2 Phase Cell Cycle Checkpoints , Gene Ontology , Iron/metabolism , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/isolation & purification , Mass Spectrometry , Molecular Sequence Annotation , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Protozoan Proteins/classification , Protozoan Proteins/isolation & purification , Trichomonas vaginalis/genetics , Trichomonas vaginalis/growth & development , Trichomonas vaginalis/metabolism , Trophozoites/genetics , Trophozoites/growth & development , Trophozoites/metabolism , Ubiquitin/chemistry , Ubiquitin/isolation & purification
2.
Inorg Chem ; 57(17): 10935-10944, 2018 Sep 04.
Article in English | MEDLINE | ID: mdl-30106575

ABSTRACT

The active site of the [FeFe] hydrogenase (HydA1), the H-cluster, is a 6-Fe cofactor that contains CO and CN- ligands. It undergoes several different oxidation and protonation state changes in its catalytic cycle to metabolize H2. Among them, the well-known Hox state and the recently identified Hhyd state are thought to be directly involved in H2 activation and evolution, and they are both EPR active with net spin S = 1/2. Herein, we report the pulse electronic paramagnetic spectroscopic investigation of these two catalytic states in Chlamydomonas reinhardtii HydA1 ( CrHydA1). Using an in vitro biosynthetic maturation approach, we site-specifically installed 13C into the CO or CN- ligands and 57Fe into the [2Fe]H subcluster of the H-cluster in order to measure the hyperfine couplings to these magnetic nuclei. For Hox, we measured 13C hyperfine couplings (13CO aiso of 25.5, 5.8, and 4.5 MHz) corresponding to all three CO ligands in the H-cluster. We also observed two 57Fe hyperfine couplings (57Fe aiso of ∼17 and 5.7 MHz) arising from the two Fe atoms in the [2Fe]H subcluster. For Hhyd, we only observed two distinct 13CO hyperfine interactions (13CO aiso of 0.16 and 0.08 MHz) and only one for 13CN- (13CN aiso of 0.16 MHz); the couplings to the 13CO/13CN- on the distal Fe of [2Fe]H may be too small to detect. We also observed a very small (<0.3 MHz) 57Fe HFI from the labeled [2Fe]H subcluster and four 57Fe HFI from the labeled [4Fe-4S]H subcluster (57Fe aiso of 7.2, 16.6, 28.2, and 35.3 MHz). These hyperfine coupling constants are consistent with the previously proposed electronic structure of the H-cluster at both Hox and Hhyd states and provide a basis for more detailed analysis.


Subject(s)
Chlamydomonas reinhardtii/enzymology , Electron Spin Resonance Spectroscopy , Hydrogenase/chemistry , Chlamydomonas reinhardtii/chemistry , Hydrogenase/classification , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/classification , Ligands , Molecular Structure , Oxidation-Reduction
3.
Emerg Microbes Infect ; 7(1): 49, 2018 Mar 29.
Article in English | MEDLINE | ID: mdl-29593238

ABSTRACT

Type VI secretion systems (T6SSs) contribute to the pathogenicity of avian pathogenic Escherichia coli (APEC), one of the leading causative agents of sepsis and meningitis in poultry. The Hcp protein is a core component of the T6SS tail tube and acts as an exported receptor and a chaperone of effectors. In this study, four distinct Hcp types (Ia, Ib, IIa, and IIb) were designated in Gram-negative bacteria, three of which were widely distributed in APEC. We detected divergence in transcription levels among three hcp clusters in 50% duck serum and demonstrated that hcp1 was upregulated by relieving Fur repression. Further analyses revealed that the host serum could activate the hcp2B operon by H-NS derepression to transcribe the downstream xmtU/xmtV pair for inter-bacterial antagonism. Notably, in a structural analysis based on the genetic classification, Hcp proteins exhibited significant differences in the extended loop regions, suggesting that these regions were related to their functional properties. Indeed, the variant region Vs2 (Loop L2, 3) in Hcp1 and Hcp2B was essential for the delivery of antibacterial effectors and the inhibition of macrophage phagocytosis. Further analyses using a duck model indicated that these Hcps play different roles in the pathogenic processes of APEC and immunoprotection. These results indicated that the functional differentiation of Hcp homologs was driven by differences in transcriptional regulation, extended loop regions, and effector delivery.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Sequence Homology, Amino Acid , Animals , Anti-Bacterial Agents/pharmacology , Birds , Ducks/virology , Escherichia coli/drug effects , Escherichia coli/immunology , Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/blood , Escherichia coli Proteins/classification , Humans , Iron-Sulfur Proteins/blood , Iron-Sulfur Proteins/classification , Macrophages/immunology , Phagocytosis , Transcription, Genetic , Type VI Secretion Systems/chemistry , Type VI Secretion Systems/genetics , Type VI Secretion Systems/metabolism , Virulence/genetics , Virulence/immunology
4.
Biochemistry ; 55(51): 7047-7064, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27966889

ABSTRACT

The ferredoxin (Fd) protein family is a structurally diverse group of iron-sulfur proteins that function as electron carriers, linking biochemical pathways important for energy transduction, nutrient assimilation, and primary metabolism. While considerable biochemical information about individual Fd protein electron carriers and their reactions has been acquired, we cannot yet anticipate the proportion of electrons shuttled between different Fd-partner proteins within cells using biochemical parameters that govern electron flow, such as holo-Fd concentration, midpoint potential (driving force), molecular interactions (affinity and kinetics), conformational changes (allostery), and off-pathway electron leakage (chemical oxidation). Herein, we describe functional and structural gaps in our Fd knowledge within the context of a sequence similarity network and phylogenetic tree, and we propose a strategy for improving our understanding of Fd sequence-function relationships. We suggest comparing the functions of divergent Fds within cells whose growth, or other measurable output, requires electron transfer between defined electron donor and acceptor proteins. By comparing Fd-mediated electron transfer with biochemical parameters that govern electron flow, we posit that models that anticipate energy flow across Fd interactomes can be built. This approach is expected to transform our ability to anticipate Fd control over electron flow in cellular settings, an obstacle to the construction of synthetic electron transfer pathways and rational optimization of existing energy-conserving pathways.


Subject(s)
Bacteria/metabolism , Electrons , Ferredoxins/metabolism , Metabolic Networks and Pathways , Amino Acid Sequence , Bacteria/cytology , Electron Transport , Ferredoxins/chemistry , Ferredoxins/genetics , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Kinetics , Models, Molecular , Mutation , Oxidation-Reduction , Phylogeny , Protein Conformation , Sequence Homology, Amino Acid
5.
Sci China Life Sci ; 59(12): 1324-1331, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27614751

ABSTRACT

Magnetoreception is essential for magnetic orientation in animal migration. The molecular basis for magnetoreception has recently been elucidated in fruitfly as complexes between the magnetic receptor magnetoreceptor (MagR) and its ligand cryptochrome (Cry). MagR and Cry are present in the animal kingdom. However, it is unknown whether they perform a conserved role in diverse animals. Here we report the identification and expression of zebrafish MagR and Cry homologs towards understanding their roles in lower vertebrates. A single magr gene and 7 cry genes are present in the zebrafish genome. Zebrafish has four cry1 genes (cry1aa, cry1ab, cry1ba and cry1bb) homologous to human CRY1 and a single ortholog of human CRY2 as well as 2 cry-like genes (cry4 and cry5). By RT-PCR, magr exhibited a high level of ubiquitous RNA expression in embryos and adult organs, whereas cry genes displayed differential embryonic and adult expression. Importantly, magr depletion did not produce apparent abnormalities in organogenesis. Taken together, magr and cry2 exist as a single copy gene, whereas cry1 exists as multiple gene duplicates in zebrafish. Our result suggests that magr may play a dispensable role in organogenesis and predicts a possibility to generate magr mutants for analyzing its role in zebrafish.


Subject(s)
Cryptochromes/genetics , Iron-Sulfur Proteins/genetics , Mechanoreceptors/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Cryptochromes/classification , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Iron-Sulfur Proteins/classification , Magnetics , Mechanoreceptors/classification , Mechanotransduction, Cellular/genetics , Organogenesis/genetics , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Zebrafish/embryology , Zebrafish/growth & development
6.
J Proteome Res ; 15(4): 1308-22, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26889782

ABSTRACT

Organisms from all kingdoms of life use iron-sulfur proteins (FeS-Ps) in a multitude of functional processes. We applied a bioinformatics approach to investigate the human portfolio of FeS-Ps. Sixty-one percent of human FeS-Ps bind Fe4S4 clusters, whereas 39% bind Fe2S2 clusters. However, this relative ratio varies significantly depending on the specific cellular compartment. We compared the portfolio of human FeS-Ps to 12 other eukaryotes and to about 700 prokaryotes. The comparative analysis of the organization of the prokaryotic homologues of human FeS-Ps within operons allowed us to reconstruct the human functional networks involving the conserved FeS-Ps common to prokaryotes and eukaryotes. These functional networks have been maintained during evolution and thus presumably represent fundamental cellular processes. The respiratory chain and the ISC machinery for FeS-P biogenesis are the two conserved processes that involve the majority of human FeS-Ps. Purine metabolism is another process including several FeS-Ps, in which BOLA proteins possibly have a regulatory role. The analysis of the co-occurrence of human FeS-Ps with other proteins highlighted numerous links between the iron-sulfur cluster machinery and the response mechanisms to cell damage, from repair to apoptosis. This relationship probably relates to the production of reactive oxygen species within the biogenesis and degradation of FeS-Ps.


Subject(s)
Bacteria/metabolism , Bacterial Proteins/chemistry , Electron Transport/genetics , Iron-Sulfur Proteins/chemistry , Operon , Apoptosis/genetics , Arabidopsis/genetics , Arabidopsis/metabolism , Archaea/genetics , Archaea/metabolism , Bacteria/genetics , Bacterial Proteins/classification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Evolution , Cell Nucleus/metabolism , Computational Biology , Cytoplasm/metabolism , Gene Expression , Humans , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Mitochondria/metabolism , Protein Structure, Secondary , Reactive Oxygen Species/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Species Specificity , Structural Homology, Protein , User-Computer Interface
7.
J Inherit Metab Dis ; 38(3): 391-403, 2015 May.
Article in English | MEDLINE | ID: mdl-25526709

ABSTRACT

Pyruvate oxidation defects (PODs) are among the most frequent causes of deficiencies in the mitochondrial energy metabolism and represent a substantial subset of classical mitochondrial diseases. PODs are not only caused by deficiency of subunits of the pyruvate dehydrogenase complex (PDHC) but also by various disorders recently described in the whole pyruvate oxidation route including cofactors, regulation of PDHC and the mitochondrial pyruvate carrier. Our own patients from 2000 to July 2014 and patients identified by a systematic survey of the literature from 1970 to July 2014 with a pyruvate oxidation disorder and a genetically proven defect were included in the study (n=628). Of these defects 74.2% (n=466) belong to PDHC subunits, 24.5% (n=154) to cofactors, 0.5% (n=3) to PDHC regulation and 0.8% (n=5) to mitochondrial pyruvate import. PODs are underestimated in the field of mitochondrial diseases because not all diagnostic centres include biochemical investigations of PDHC in their routine analysis. Cofactor and transport defects can be missed, if pyruvate oxidation is not measured in intact mitochondria routinely. Furthermore deficiency of the X-chromosomal PDHA1 can be biochemically missed depending on the X-inactivation pattern. This is reflected by an increasing number of patients diagnosed recently by genetic high throughput screening approaches. PDHC deficiency including regulation and import affect mainly the glucose dependent central and peripheral nervous system and skeletal muscle. PODs with combined enzyme defects affect also other organs like heart, lung and liver. The spectrum of clinical presentation of PODs is still expanding. PODs are a therapeutically interesting group of mitochondrial diseases since some can be bypassed by ketogenic diet or treated by cofactor supplementation. PDHC kinase inhibition, chaperone therapy and PGC1α stimulation is still a matter of further investigations.


Subject(s)
Iron-Sulfur Proteins/metabolism , Pyruvate Dehydrogenase Complex Deficiency Disease/diagnosis , Pyruvate Dehydrogenase Complex/metabolism , Thiamine Pyrophosphate/metabolism , Thioctic Acid/metabolism , Energy Metabolism , Female , Humans , Iron-Sulfur Proteins/classification , Male , Oxidation-Reduction , Pyruvate Dehydrogenase Complex/classification , Pyruvate Dehydrogenase Complex Deficiency Disease/drug therapy , Pyruvate Dehydrogenase Complex Deficiency Disease/genetics , Thiamine Pyrophosphate/classification , Thioctic Acid/classification
8.
Met Ions Life Sci ; 14: 279-313, 2014.
Article in English | MEDLINE | ID: mdl-25416398

ABSTRACT

Dimethylsulfide (DMS) is a naturally occurring chemical that is part of the biogeochemical sulfur cycle and has been implicated in climate-relevant atmospheric processes. In addition, DMS occurs in soil environments as well as in food stuff as a flavor compound and it can also be associated with disease states such as halitosis. A major environmental source of DMS is the marine algal osmoprotectant dimethylsulfoniopropionate (DMSP). A variety of bacterial enzyme systems lead either to the production of DMS from DMSP or dimethylsulfoxide (DMSO) or its oxidation to, e.g., DMSO. The interconversion of DMS and DMSO is catalyzed by molybdenum-containing metalloenzymes that have been very well studied, and recently another enzyme system, an NADH-dependent, flavin-containing monooxygenase, that produces formaldehyde and methanethiol from DMS has also been described.DMS conversions are not limited to a specialized group of bacteria - evidence for DMS-based metabolism exists for heterotrophic, autotrophic and phototrophic bacteria and there is also evidence for the occurrence of this type of sulfur compound conversion in Archaea.


Subject(s)
Bacteria, Aerobic/enzymology , Bacteria, Anaerobic/enzymology , Iron-Sulfur Proteins/metabolism , Oxidoreductases/metabolism , Sulfides/metabolism , Aerobiosis , Anaerobiosis , Biotransformation , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/classification , Oxidation-Reduction , Oxidoreductases/chemistry , Oxidoreductases/classification , Phylogeny
9.
J Bacteriol ; 195(20): 4726-34, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23955005

ABSTRACT

Iron is essential for pathogen survival, virulence, and colonization. Feo is suggested to function as the ferrous iron (Fe(2+)) transporter. The enterobacterial Feo system is composed of 3 proteins: FeoB is the indispensable component and is a large membrane protein likely to function as a permease; FeoA is a small Src homology 3 (SH3) domain protein that interacts with FeoB; FeoC is a winged-helix protein containing 4 conserved Cys residues in a sequence suitable for harboring a putative iron-sulfur (Fe-S) cluster. The presence of an iron-sulfur cluster on FeoC has never been shown experimentally. We report that under anaerobic conditions, the recombinant Klebsiella pneumoniae FeoC (KpFeoC) exhibited hyperfine-shifted nuclear magnetic resonance (NMR) and a UV-visible (UV-Vis) absorbance spectrum characteristic of a paramagnetic center. The electron paramagnetic resonance (EPR) and extended X-ray absorption fine structure (EXAFS) results were consistent only with the [4Fe-4S] clusters. Substituting the cysteinyl sulfur with oxygen resulted in significantly reduced cluster stability, establishing the roles of these cysteines as the ligands for the Fe-S cluster. When exposed to oxygen, the [4Fe-4S] cluster degraded to [3Fe-4S] and eventually disappeared. We propose that KpFeoC may regulate the function of the Feo transporter through the oxygen- or iron-sensitive coordination of the Fe-S cluster.


Subject(s)
Bacterial Proteins/metabolism , Iron-Sulfur Proteins/metabolism , Klebsiella pneumoniae/metabolism , Absorptiometry, Photon , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial/physiology , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/genetics , Klebsiella pneumoniae/genetics , Magnetic Resonance Spectroscopy , Oxidation-Reduction
10.
PLoS One ; 7(10): e47232, 2012.
Article in English | MEDLINE | ID: mdl-23056615

ABSTRACT

The Cas4 protein is one of the core CRISPR-associated (Cas) proteins implicated in the prokaryotic CRISPR system for antiviral defence. Cas4 is thought to play a role in the capture of new viral DNA sequences for incorporation into the host genome. No biochemical activity has been reported for Cas4, but it is predicted to include a RecB nuclease domain. We show here that Cas4 family proteins from the archaeon Sulfolobus solfataricus utilise four conserved cysteine residues to bind an iron-sulfur cluster in an arrangement reminiscent of the AddB nuclease of Bacillus subtilis. The Cas4 family protein Sso0001 is a 5' to 3' single stranded DNA exonuclease in vitro that is stalled by extrahelical DNA adducts. A role for Cas4 in DNA duplex strand resectioning to generate recombinogenic 3' single stranded DNA overhangs is proposed. Comparison of the AddB structure with that of a related bacterial nuclease from Eubacterium rectales reveals that the iron-sulfur cluster can be replaced by a zinc ion without disrupting the protein structure, with implications for the evolution of iron-sulfur binding proteins.


Subject(s)
Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/classification , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Chromatography, Gel , Exodeoxyribonucleases/classification , Exodeoxyribonucleases/genetics , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/genetics , Phylogeny , Sulfolobus solfataricus/enzymology
11.
Lett Appl Microbiol ; 53(4): 473-80, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21838748

ABSTRACT

AIMS: The aim of this study was to explore and characterize the genetic diversity of [FeFe] hydrogenases in a representative set of strains from Clostridium sp. and to reveal the existence of neither yet detected nor characterized [FeFe] hydrogenases in hydrogen-producing strains. METHODS AND RESULTS: The genomes of 57 Clostridium strains (34 different genotypic species), representing six phylogenetic clusters based on their 16S rRNA sequence analysis (cluster I, III, XIa, XIb, XIV and XVIII), were screened for different [FeFe] hydrogenases. Based on the obtained alignments, ten pairs of [FeFe] hydrogenase cluster-specific degenerate primers were newly designed. Ten Clostridium strains were screened by PCRs to assess the specificity of the primers designed and to examine the genetic diversity of [FeFe] hydrogenases. Using this approach, a diversity of hydrogenase genes was discovered in several species previously shown to produce hydrogen in bioreactors: Clostridium sartagoforme, Clostridium felsineum, Clostridium roseum and Clostridium pasteurianum. CONCLUSIONS: The newly designed [FeFe] hydrogenase cluster-specific primers, targeting the cluster-conserved regions, allow for a direct amplification of a specific hydrogenase gene from the species of interest. SIGNIFICANCE AND IMPACT OF THE STUDY: Using this strategy for a screening of different Clostridium ssp. will provide new insights into the diversity of hydrogenase genes and should be a first step to study a complex hydrogen metabolism of this genus.


Subject(s)
Clostridium/genetics , DNA Primers/chemistry , Genetic Variation , Hydrogenase/genetics , Iron-Sulfur Proteins/genetics , Clostridium/classification , Clostridium/enzymology , Humans , Hydrogenase/classification , Iron-Sulfur Proteins/classification , Phylogeny , Polymerase Chain Reaction
12.
Biochem J ; 420(3): 463-72, 2009 May 27.
Article in English | MEDLINE | ID: mdl-19309314

ABSTRACT

IscA/SufA paralogues are the members of the iron-sulfur cluster assembly machinery in Escherichia coli. Whereas deletion of either IscA or SufA has only a mild effect on cell growth, deletion of both IscA and SufA results in a null-growth phenotype in minimal medium under aerobic growth conditions. Here we report that cell growth of the iscA/sufA double mutant (E. coli strain in which both iscA and sufA had been in-frame-deleted) can be partially restored by supplementing with BCAAs (branched-chain amino acids) and thiamin. We further demonstrate that deletion of IscA/SufA paralogues blocks the [4Fe-4S] cluster assembly in IlvD (dihydroxyacid dehydratase) of the BCAA biosynthetic pathway in E. coli cells under aerobic conditions and that addition of the iron-bound IscA/SufA efficiently promotes the [4Fe-4S] cluster assembly in IlvD and restores the enzyme activity in vitro, suggesting that IscA/SufA may act as an iron donor for the [4Fe-4S] cluster assembly under aerobic conditions. Additional studies reveal that IscA/SufA are also required for the [4Fe-4S] cluster assembly in enzyme ThiC of the thiamin-biosynthetic pathway, aconitase B of the citrate acid cycle and endonuclease III of the DNA-base-excision-repair pathway in E. coli under aerobic conditions. Nevertheless, deletion of IscA/SufA does not significantly affect the [2Fe-2S] cluster assembly in the redox transcription factor SoxR, ferredoxin and the siderophore-iron reductase FhuF. The results suggest that the biogenesis of the [4Fe-4S] clusters and the [2Fe-2S] clusters may have distinct pathways and that IscA/SufA paralogues are essential for the [4Fe-4S] cluster assembly, but are dispensable for the [2Fe-2S] cluster assembly in E. coli under aerobic conditions.


Subject(s)
Carrier Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Hydro-Lyases/metabolism , Iron-Sulfur Proteins/metabolism , Aerobiosis , Amino Acids, Branched-Chain/biosynthesis , Amino Acids, Branched-Chain/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/genetics , Cell Division/drug effects , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Hydro-Lyases/genetics , Iron/metabolism , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/genetics , Mutation , Thiamine/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
13.
BMC Biochem ; 10: 3, 2009 Feb 02.
Article in English | MEDLINE | ID: mdl-19187533

ABSTRACT

BACKGROUND: Iron-sulfur clusters are ubiquitous and evolutionarily ancient inorganic prosthetic groups, the biosynthesis of which depends on complex protein machineries. Three distinct assembly systems involved in the maturation of cellular Fe-S proteins have been determined, designated the NIF, ISC and SUF systems. Although well described in several organisms, these machineries are poorly understood in Gram-positive bacteria. Within the Firmicutes phylum, the Enterococcus spp. genus have recently assumed importance in clinical microbiology being considered as emerging pathogens for humans, wherein Enterococcus faecalis represents the major species associated with nosocomial infections. The aim of this study was to carry out a phylogenetic analysis in Enterococcus faecalis V583 and a structural and conformational characterisation of it SufU protein. RESULTS: BLAST searches of the Enterococcus genome revealed a series of genes with sequence similarity to the Escherichia coli SUF machinery of [Fe-S] cluster biosynthesis, namely sufB, sufC, sufD and SufS. In addition, the E. coli IscU ortholog SufU was found to be the scaffold protein of Enterococcus spp., containing all features considered essential for its biological activity, including conserved amino acid residues involved in substrate and/or co-factor binding (Cys50,76,138 and Asp52) and, phylogenetic analyses showed a close relationship with orthologues from other Gram-positive bacteria. Molecular dynamics for structural determinations and molecular modeling using E. faecalis SufU primary sequence protein over the PDB:1su0 crystallographic model from Streptococcus pyogenes were carried out with a subsequent 50 ns molecular dynamic trajectory. This presented a stable model, showing secondary structure modifications near the active site and conserved cysteine residues. Molecular modeling using Haemophilus influenzae IscU primary sequence over the PDB:1su0 crystal followed by a MD trajectory was performed to analyse differences in the C-terminus region of Gram-positive SufU and Gram-negative orthologous proteins, in which several modifications in secondary structure were observed. CONCLUSION: The data describe the identification of the SUF machinery for [Fe-S] cluster biosynthesis present in the Firmicutes genome, showing conserved sufB, sufC, sufD and sufS genes and the presence of the sufU gene coding for scaffold protein, instead of sufA; neither sufE nor sufR are present. Primary sequences and structural analysis of the SufU protein demonstrated its structural-like pattern to the scaffold protein IscU nearby on the ISC machinery. E. faecalis SufU molecular modeling showed high flexibility over the active site regions, and demonstrated the existence of a specific region in Firmicutes denoting the Gram positive region (GPR), suggested as a possible candidate for interaction with other factors and/or regulators.


Subject(s)
Enterococcus faecalis/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Databases, Protein , Enterococcus faecalis/classification , Genome, Bacterial/genetics , Gram-Positive Bacteria/metabolism , Iron-Sulfur Proteins/classification , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Structure, Secondary , Sequence Homology, Amino Acid
14.
Cell Res ; 18(5): 566-76, 2008 May.
Article in English | MEDLINE | ID: mdl-18332905

ABSTRACT

Thiamine (vitamin B(1)) is an essential compound for organisms. It contains a pyrimidine ring structure and a thiazole ring structure. These two moieties of thiamine are synthesized independently and then coupled together. Here we report the molecular characterization of AtTHIC, which is involved in thiamine biosynthesis in Arabidopsis. AtTHIC is similar to Escherichia coli ThiC, which is involved in pyrimidine biosynthesis in prokaryotes. Heterologous expression of AtTHIC could functionally complement the thiC knock-out mutant of E. coli. Downregulation of AtTHIC expression by T-DNA insertion at its promoter region resulted in a drastic reduction of thiamine content in plants and the knock-down mutant thic1 showed albino (white leaves) and lethal phenotypes under the normal culture conditions. The thic1 mutant could be rescued by supplementation of thiamine and its defect functions could be complemented by expression of AtTHIC cDNA. Transient expression analysis revealed that the AtTHIC protein targets plastids and chloroplasts. AtTHIC was strongly expressed in leaves, flowers and siliques and the transcription of AtTHIC was downregulated by extrinsic thiamine. In conclusion, AtTHIC is a gene involved in pyrimidine synthesis in the thiamine biosynthesis pathway of Arabidopsis, and our results provide some new clues for elucidating the pathway of thiamine biosynthesis in plants.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Iron-Sulfur Proteins/metabolism , Thiamine/biosynthesis , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis Proteins/classification , Arabidopsis Proteins/genetics , Blotting, Northern , Diphosphates/chemistry , Diphosphates/metabolism , Escherichia coli/genetics , Genetic Complementation Test , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/genetics , Models, Biological , Molecular Sequence Data , Molecular Structure , Mutation , Phenotype , Phylogeny , Plants, Genetically Modified , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Thiamine/chemistry
15.
Biometals ; 21(4): 459-67, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18286376

ABSTRACT

Three genes within the genome of E. coli K12 are predicted to encode proteins containing the typical Rieske iron-sulfur cluster-binding motifs. Two of these, hcaC and yeaW, were overexpressed in E. coli BL21 and Tuner (DE3) pLacI. The recombinant proteins were purified and analyzed by UV/Vis- and EPR-spectroscopy. HcaC and YeaW display the typical redox-dependent UV/Vis-spectra of iron-sulfur proteins. The EPR spectrum of reduced HcaC shows characteristic g-values of a Rieske cluster whereas the g-values for YeaW are close to the upper limit for this type of iron-sulfur cluster. Both iron-sulfur clusters could be reduced by dithionite, but not by ascorbate, confirming their classification as low-potential Rieske proteins as derived from the amino acid sequences. A phylogenetic analysis of the two proteins reveals that HcaC clearly segregates with the Rieske ferredoxins of class IIB oxygenases whereas the classification of YeaW remains doubtful.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Amino Acid Sequence , Escherichia coli/metabolism , Escherichia coli Proteins/classification , Escherichia coli Proteins/genetics , Ferredoxins/chemistry , Ferredoxins/classification , Ferredoxins/genetics , Ferredoxins/metabolism , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/genetics , Models, Molecular , Molecular Sequence Data , Oxidoreductases/chemistry , Oxidoreductases/classification , Oxidoreductases/genetics , Oxidoreductases/metabolism , Phylogeny , Protein Structure, Tertiary , Recombinant Proteins/classification , Recombinant Proteins/genetics , Sequence Alignment , Sequence Analysis, Protein
16.
Biochim Biophys Acta ; 1778(9): 1897-929, 2008 Sep.
Article in English | MEDLINE | ID: mdl-17964535

ABSTRACT

Bacterial genomes encode an extensive range of respiratory enzymes that enable respiratory metabolism with a diverse group of reducing and oxidizing substrates under both aerobic and anaerobic growth conditions. An important class of enzymes that contributes to this broad diversity is the complex iron-sulfur molybdoenzyme (CISM) family. The architecture of this class comprises the following subunits. (i) A molybdo-bis(pyranopterin guanine dinucleotide) (Mo-bisPGD) cofactor-containing catalytic subunit that also contains a cubane [Fe-S] cluster (FS0). (ii) A four-cluster protein (FCP) subunit that contains 4 cubane [Fe-S] clusters (FS1-FS4). (iii) A membrane anchor protein (MAP) subunit which anchors the catalytic and FCP subunits to the cytoplasmic membrane. In this review, we define the CISM family of enzymes on the basis of emerging structural and bioinformatic data, and show that the catalytic and FCP subunit architectures appear in a wide range of bacterial redox enzymes. We evaluate evolutionary events involving genes encoding the CISM catalytic subunit that resulted in the emergence of the complex I (NADH:ubiquinone oxidoreductase) Nqo3/NuoG subunit architecture. We also trace a series of evolutionary events leading from a primordial Cys-containing peptide to the FCP architecture. Finally, many of the CISM archetypes and related enzymes rely on the tat translocon to transport fully folded monomeric or dimeric subunits across the cytoplasmic membrane. We have used genome sequence data to establish that there is a bias against the presence of soluble periplasmic molybdoenzymes in bacteria lacking an outer membrane.


Subject(s)
Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Molybdenum/chemistry , Prokaryotic Cells/enzymology , Amino Acid Sequence , Biological Evolution , Catalytic Domain/physiology , Iron-Sulfur Proteins/classification , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Biological , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction , Periplasm/chemistry , Periplasm/enzymology , Periplasm/metabolism , Phylogeny , Prokaryotic Cells/chemistry , Protein Subunits/chemistry , Protein Subunits/physiology , Sequence Homology, Amino Acid
17.
Proc Natl Acad Sci U S A ; 104(34): 13626-31, 2007 Aug 21.
Article in English | MEDLINE | ID: mdl-17698959

ABSTRACT

Understanding the biogenesis of iron-sulfur (Fe-S) proteins is relevant to many fields, including bioenergetics, gene regulation, and cancer research. Several multiprotein complexes assisting Fe-S assembly have been identified in both prokaryotes and eukaryotes. Here, we identify in Escherichia coli an A-type Fe-S protein that we named ErpA. Remarkably, erpA was found essential for growth of E. coli in the presence of oxygen or alternative electron acceptors. It was concluded that isoprenoid biosynthesis was impaired by the erpA mutation. First, the eukaryotic mevalonate-dependent pathway for biosynthesis of isopentenyl diphosphate restored the respiratory defects of an erpA mutant. Second, the erpA mutant contained a greatly reduced amount of ubiquinone and menaquinone. Third, ErpA bound Fe-S clusters and transferred them to apo-IspG, a protein catalyzing isopentenyl diphosphate biosynthesis in E. coli. Surprisingly, the erpA gene maps at a distance from any other Fe-S biogenesis-related gene. ErpA is an A-type Fe-S protein that is characterized by an essential role in cellular metabolism.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Iron-Sulfur Proteins/metabolism , Aerobiosis , Anaerobiosis , Benzoquinones/metabolism , Cell Respiration , Escherichia coli/cytology , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/classification , Escherichia coli Proteins/genetics , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/genetics , Mevalonic Acid/pharmacology , Microbial Viability , Multigene Family , Mutation/genetics , Phenotype , Protein Binding
18.
FEBS Lett ; 579(5): 1020-6, 2005 Feb 14.
Article in English | MEDLINE | ID: mdl-15710385

ABSTRACT

A new subtype of archaeal Rieske ferredoxin (RFd) has been identified in the genome of the thermoacidophilic archaeon Acidianus ambivalens. The gene is inserted in an atypical genomic context in a gene cluster encoding a NiFe hydrogenase. Sequence and phyletic analysis showed that the protein is related to bacterial RFd but not to any of the known archaeal Rieske proteins. The recombinant 14 kDa protein isolated from Escherichia coli behaved as a dimer in solution. It contained approximately 2 Fe/mol and all visible and EPR spectroscopic features typical of Rieske centre-containing proteins. However, its redox potential (+170 mV) was significantly higher than those of canonical RFd. This difference is rationalized in terms of the protein structure environment, as discrete amino acid substitutions in key positions around the metal centre account for the higher potential.


Subject(s)
Acidianus/chemistry , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Electron Transport Complex III/classification , Electron Transport Complex III/metabolism , Ferredoxins/classification , Ferredoxins/metabolism , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/metabolism , Acidianus/classification , Amino Acid Sequence , Archaeal Proteins/classification , Archaeal Proteins/genetics , Electron Transport Complex III/chemistry , Electron Transport Complex III/genetics , Ferredoxins/chemistry , Ferredoxins/genetics , Gene Expression , Genome, Archaeal , Hydrogen-Ion Concentration , Iron/pharmacology , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Kinetics , Molecular Biology , Oxidation-Reduction , Phylogeny , Protein Conformation , Protein Denaturation , Sequence Alignment , Spectrum Analysis , Temperature
19.
Int J Parasitol ; 32(10): 1207-17, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12204220

ABSTRACT

During the last 4 years there has been an enormous interest in the question how iron-sulphur ([Fe-S]) clusters, which are essential building blocks for life, are synthesised and assembled into apo-proteins, both in prokaryotes and in eukaryotes. The emerging picture is that the basic mechanism of this pathway has been well conserved during evolution. In yeast and probably all other eukaryotes the mitochondrion is the place where [Fe-S] clusters are synthesised, even for extramitochondrial [Fe-S] cluster-containing proteins, and a number of proteins have been functionally characterised to a certain extent within this pathway. However, almost nothing is known about this aspect in parasitic protists, although recent studies of amitochondriate protists and on the plastid-like organelle of apicomplexan parasites, the apicoplast, have started to change this. In this article I will summarise the current view of [Fe-S] cluster biogenesis in eukaryotes and discuss its implications for amitochondriate protists and for the plastid-like organelle of apicomplexan parasites.


Subject(s)
Apicomplexa/metabolism , Eukaryota/metabolism , Iron-Sulfur Proteins/biosynthesis , Animals , Apicomplexa/cytology , Apicomplexa/genetics , Bacterial Physiological Phenomena , Eukaryota/cytology , Eukaryotic Cells/physiology , Iron-Sulfur Proteins/classification , Mitochondria/metabolism , Models, Biological , Yeasts/physiology
20.
FEMS Microbiol Rev ; 25(4): 455-501, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11524134

ABSTRACT

Hydrogenases (H2ases) catalyze the reversible oxidation of molecular hydrogen and play a central role in microbial energy metabolism. Most of these enzymes are found in Archaea and Bacteria, but a few are present in Eucarya as well. They can be distributed into three classes: the [Fe]-H2ases, the [NiFe]-H2ases, and the metal-free H2ases. The vast majority of known H2ases belong to the first two classes, and over 100 of these enzymes have been characterized genetically and/or biochemically. Compelling evidence from sequences and structures indicates that the [NiFe]- and [Fe]-H2ases are phylogenetically distinct classes of proteins. The catalytic core of the [NiFe]-H2ases is a heterodimeric protein, although additional subunits are present in many of these enzymes. Functional classes of [NiFe]-H2ases have been defined, and they are consistent with categories defined by sequence similarity of the catalytic subunits. The catalytic core of the [Fe]-H2ases is a ca. 350-residue domain that accommodates the active site (H-cluster). A few monomeric [Fe]-H2ases are barely larger than the H-cluster domain. Many others are monomeric as well, but possess additional domains that contain redox centers, mostly iron-sulfur. Some [Fe]-H2ases are oligomeric. The modular structure of H2ases is strikingly illustrated in recently unveiled sequences and structures. It is also remarkable that most of the accessory domains and subunits of H2ases have counterparts in other redox complexes, in particular NADH-ubiquinone oxidoreductase (Complex I) of respiratory chains. Microbial genome sequences are bringing forth a significant body of additional H2ase sequence data and contribute to the understanding of H2ase distribution and evolution. Altogether, the available data suggest that [Fe]-H2ases are restricted to Bacteria and Eucarya, while [NiFe]-H2ases, with one possible exception, seem to be present only in Archaea and Bacteria. H2ase processing and maturation involve the products of several genes which have been identified and are currently being characterized in the case of the [NiFe]-H2ases. In contrast, near to nothing is known regarding the maturation of the [Fe]-H2ases. Inspection of the currently available genome sequences suggests that the [NiFe]-H2ase maturation proteins have no similar counterparts in the genomes of organisms possessing [Fe]-H2ases only. This observation, if confirmed, would be consistent with the phylogenetic distinctiveness of the two classes of H2ases. Sequence alignments of catalytic subunits of H2ases have been implemented to construct phylogenetic trees that were found to be consistent, in the main, with trees derived from other data. On the basis of the comparisons performed and discussed here, proposals are made to simplify and rationalize the nomenclature of H2ase-encoding genes.


Subject(s)
Bacteria/enzymology , Evolution, Molecular , Hydrogenase/chemistry , Hydrogenase/classification , Phylogeny , Amino Acid Sequence , Bacteria/genetics , Binding Sites , Hydrogenase/genetics , Hydrogenase/metabolism , Iron/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/classification , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Nickel/metabolism , Protein Subunits
SELECTION OF CITATIONS
SEARCH DETAIL