Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Inorg Chem ; 63(19): 8730-8738, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38687645

RESUMO

Iron-sulfur (Fe-S) clusters are essential inorganic cofactors dedicated to a wide range of biological functions, including electron transfer and catalysis. Specialized multiprotein machineries present in all types of organisms support their biosynthesis. These machineries encompass a scaffold protein, on which Fe-S clusters are assembled before being transferred to cellular targets. Here, we describe the first characterization of the native Fe-S cluster of the anaerobically purified SufBC2D scaffold from Escherichia coli by XAS and Mössbauer, UV-visible absorption, and EPR spectroscopies. Interestingly, we propose that SufBC2D harbors two iron-sulfur-containing species, a [2Fe-2S] cluster and an as-yet unidentified species. Mutagenesis and biochemistry were used to propose amino acid ligands for the [2Fe-2S] cluster, supporting the hypothesis that both SufB and SufD are involved in the Fe-S cluster ligation. The [2Fe-2S] cluster can be transferred to ferredoxin in agreement with the SufBC2D scaffold function. These results are discussed in the context of Fe-S cluster biogenesis.


Assuntos
Proteínas de Escherichia coli , Escherichia coli , Proteínas Ferro-Enxofre , Proteínas Ferro-Enxofre/química , Proteínas Ferro-Enxofre/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Espectroscopia de Mossbauer , Espectroscopia por Absorção de Raios X , Proteínas de Transporte
2.
J Biol Chem ; 293(20): 7689-7702, 2018 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-29626095

RESUMO

Fe-S cluster-containing proteins occur in most organisms, wherein they assist in myriad processes from metabolism to DNA repair via gene expression and bioenergetic processes. Here, we used both in vitro and in vivo methods to investigate the capacity of the four Fe-S carriers, NfuA, SufA, ErpA, and IscA, to fulfill their targeting role under oxidative stress. Likewise, Fe-S clusters exhibited varying half-lives, depending on the carriers they were bound to; an NfuA-bound Fe-S cluster was more stable (t½ = 100 min) than those bound to SufA (t½ = 55 min), ErpA (t½ = 54 min), or IscA (t½ = 45 min). Surprisingly, the presence of NfuA further enhanced stability of the ErpA-bound cluster to t½ = 90 min. Using genetic and plasmon surface resonance analyses, we showed that NfuA and ErpA interacted directly with client proteins, whereas IscA or SufA did not. Moreover, NfuA and ErpA interacted with one another. Given all of these observations, we propose an architecture of the Fe-S delivery network in which ErpA is the last factor that delivers cluster directly to most if not all client proteins. NfuA is proposed to assist ErpA under severely unfavorable conditions. A comparison with the strategy employed in yeast and eukaryotes is discussed.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Redes e Vias Metabólicas , Estresse Oxidativo , Oxigênio/metabolismo , Regulação Bacteriana da Expressão Gênica , Oxirredução
3.
J Biol Inorg Chem ; 23(4): 581-596, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29280002

RESUMO

Iron-sulfur clusters (Fe-S) are amongst the most ancient and versatile inorganic cofactors in nature which are used by proteins for fundamental biological processes. Multiprotein machineries (NIF, ISC, SUF) exist for Fe-S cluster biogenesis which are mainly conserved from bacteria to human. SUF system (sufABCDSE operon) plays a general role in many bacteria under conditions of iron limitation or oxidative stress. In this mini-review, we will summarize the current understanding of the molecular mechanism of Fe-S biogenesis by SUF. The advances in our understanding of the molecular aspects of SUF originate from biochemical, biophysical and recent structural studies. Combined with recent in vivo experiments, the understanding of the Fe-S biogenesis mechanism considerably moved forward.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Ferro-Enxofre/biossíntese , Proteínas de Bactérias/genética , Humanos , Óperon/genética
4.
J Biol Inorg Chem ; 23(4): 597, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29869185

RESUMO

The article "Iron-sulfur clusters biogenesis by the SUF machinery: close to the molecular mechanism understanding", written by J. Pérard, Sandrine Ollagnier de Choudens was originally published electronically on the publisher's internet portal (currently SpringerLink) 26 December, 2017 without open access.

5.
PLoS Genet ; 11(5): e1005134, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25996492

RESUMO

Fe-S bound proteins are ubiquitous and contribute to most basic cellular processes. A defect in the ISC components catalyzing Fe-S cluster biogenesis leads to drastic phenotypes in both eukaryotes and prokaryotes. In this context, the Frataxin protein (FXN) stands out as an exception. In eukaryotes, a defect in FXN results in severe defects in Fe-S cluster biogenesis, and in humans, this is associated with Friedreich's ataxia, a neurodegenerative disease. In contrast, prokaryotes deficient in the FXN homolog CyaY are fully viable, despite the clear involvement of CyaY in ISC-catalyzed Fe-S cluster formation. The molecular basis of the differing importance in the contribution of FXN remains enigmatic. Here, we have demonstrated that a single mutation in the scaffold protein IscU rendered E. coli viability strictly dependent upon a functional CyaY. Remarkably, this mutation changed an Ile residue, conserved in prokaryotes at position 108, into a Met residue, conserved in eukaryotes. We found that in the double mutant IscUIM ΔcyaY, the ISC pathway was completely abolished, becoming equivalent to the ΔiscU deletion strain and recapitulating the drastic phenotype caused by FXN deletion in eukaryotes. Biochemical analyses of the "eukaryotic-like" IscUIM scaffold revealed that it exhibited a reduced capacity to form Fe-S clusters. Finally, bioinformatic studies of prokaryotic IscU proteins allowed us to trace back the source of FXN-dependency as it occurs in present-day eukaryotes. We propose an evolutionary scenario in which the current mitochondrial Isu proteins originated from the IscUIM version present in the ancestor of the Rickettsiae. Subsequent acquisition of SUF, the second Fe-S cluster biogenesis system, in bacteria, was accompanied by diminished contribution of CyaY in prokaryotic Fe-S cluster biogenesis, and increased tolerance to change in the amino acid present at the 108th position of the scaffold.


Assuntos
Proteínas de Escherichia coli/genética , Escherichia coli/genética , Proteínas de Ligação ao Ferro/metabolismo , Proteínas Ferro-Enxofre/genética , Biologia Computacional , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Deleção de Genes , Proteínas de Ligação ao Ferro/genética , Proteínas Ferro-Enxofre/metabolismo , Viabilidade Microbiana , Mutação , Filogenia , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Frataxina
6.
J Am Chem Soc ; 138(36): 11802-9, 2016 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-27545412

RESUMO

The enzyme NadA catalyzes the synthesis of quinolinic acid (QA), the precursor of the universal nicotinamide adenine dinucleotide (NAD) cofactor. Here, we report the crystal structures of complexes between the Thermotoga maritima (Tm) NadA K219R/Y107F variant and (i) the first intermediate (W) resulting from the condensation of dihydroxyacetone phosphate (DHAP) with iminoaspartate and (ii) the DHAP analogue and triose-phosphate isomerase inhibitor phosphoglycolohydroxamate (PGH). In addition, using the TmNadA K219R/Y21F variant, we have reacted substrates and obtained a crystalline complex between this protein and the QA product. We also show that citrate can bind to both TmNadA K219R and its Y21F variant. The W structure indicates that condensation causes dephosphorylation. We propose that catalysis by the K219R/Y107F variant is arrested at the W intermediate because the mutated protein is unable to catalyze its aldo-keto isomerization and/or cyclization that ultimately lead to QA formation. Intriguingly, PGH binds to NadA with its phosphate group at the site where the carboxylate groups of W also bind. Our results shed significant light on the mechanism of the reaction catalyzed by NadA.


Assuntos
Alquil e Aril Transferases/química , Alquil e Aril Transferases/metabolismo , Ácido Quinolínico/metabolismo , Alquil e Aril Transferases/genética , Cristalografia por Raios X , Fosfato de Di-Hidroxiacetona/metabolismo , Simulação de Acoplamento Molecular , Mutação , Conformação Proteica , Thermotoga maritima/enzimologia
7.
Biochemistry ; 54(42): 6443-6, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26455817

RESUMO

Quinolinate synthase (NadA) is an Fe4S4 cluster-containing dehydrating enzyme involved in the synthesis of quinolinic acid (QA), the universal precursor of the essential coenzyme nicotinamide adenine dinucleotide. The reaction catalyzed by NadA is not well understood, and two mechanisms have been proposed in the literature that differ in the nature of the molecule (DHAP or G-3P) that condenses with iminoaspartate (IA) to form QA. In this article, using biochemical approaches, we demonstrate that DHAP is the triose that condenses with IA to form QA. The capacity of NadA to use G-3P is due to its previously unknown triose phosphate isomerase activity.


Assuntos
Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Triose-Fosfato Isomerase/química , Triose-Fosfato Isomerase/metabolismo , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Fosfato de Di-Hidroxiacetona/metabolismo , Redes e Vias Metabólicas , Modelos Químicos , NAD/biossíntese , Ácido Quinolínico/metabolismo , Thermotoga maritima/enzimologia
8.
Proc Natl Acad Sci U S A ; 109(26): 10426-31, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22699510

RESUMO

Iron/sulfur cluster (ISC)-containing proteins are essential components of cells. In most eukaryotes, Fe/S clusters are synthesized by the mitochondrial ISC machinery, the cytosolic iron/sulfur assembly system, and, in photosynthetic species, a plastid sulfur-mobilization (SUF) system. Here we show that the anaerobic human protozoan parasite Blastocystis, in addition to possessing ISC and iron/sulfur assembly systems, expresses a fused version of the SufC and SufB proteins of prokaryotes that it has acquired by lateral transfer from an archaeon related to the Methanomicrobiales, an important lineage represented in the human gastrointestinal tract microbiome. Although components of the Blastocystis ISC system function within its anaerobic mitochondrion-related organelles and can functionally replace homologues in Trypanosoma brucei, its SufCB protein has similar biochemical properties to its prokaryotic homologues, functions within the parasite's cytosol, and is up-regulated under oxygen stress. Blastocystis is unique among eukaryotic pathogens in having adapted to its parasitic lifestyle by acquiring a SUF system from nonpathogenic Archaea to synthesize Fe/S clusters under oxygen stress.


Assuntos
Evolução Biológica , Blastocystis/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Anaerobiose , Animais , Dados de Sequência Molecular , Filogenia
9.
J Bacteriol ; 196(2): 300-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24187084

RESUMO

The cell envelope of Gram-negative bacteria is an essential organelle that is important for cell shape and protection from toxic compounds. Proteins involved in envelope biogenesis are therefore attractive targets for the design of new antibacterial agents. In a search for new envelope assembly factors, we screened a collection of Escherichia coli deletion mutants for sensitivity to detergents and hydrophobic antibiotics, a phenotype indicative of defects in the cell envelope. Strains lacking yciM were among the most sensitive strains of the mutant collection. Further characterization of yciM mutants revealed that they display a thermosensitive growth defect on low-osmolarity medium and that they have a significantly altered cell morphology. At elevated temperatures, yciM mutants form bulges containing cytoplasmic material and subsequently lyse. We also discovered that yciM genetically interacts with envC, a gene encoding a regulator of the activity of peptidoglycan amidases. Altogether, these results indicate that YciM is required for envelope integrity. Biochemical characterization of the protein showed that YciM is anchored to the inner membrane via its N terminus, the rest of the protein being exposed to the cytoplasm. Two CXXC motifs are present at the C terminus of YciM and serve to coordinate a redox-sensitive iron center of the rubredoxin type. Both the N-terminal membrane anchor and the C-terminal iron center of YciM are important for function.


Assuntos
Membrana Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/fisiologia , Proteínas de Membrana/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Bacteriólise , Meios de Cultura/química , Endopeptidases/metabolismo , Escherichia coli/citologia , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/efeitos da radiação , Proteínas de Escherichia coli/genética , Deleção de Genes , Temperatura Alta , Ferro/metabolismo , Proteínas de Membrana/genética , Microscopia , Dados de Sequência Molecular , Pressão Osmótica , Ligação Proteica , Mapeamento de Interação de Proteínas , Alinhamento de Sequência
10.
Biochemistry ; 53(50): 7867-9, 2014 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-25485887

RESUMO

Fe/S biosynthesis is controlled in Escherichia coli by two machineries, the housekeeping ISC machinery and the SUF system that is functional under stress conditions. Despite many in vivo studies showing that SUF is more adapted for Fe/S assembly under stress, no molecular data supporting this concept have been provided so far. This work focuses on molecular studies of key actors in Fe/S assembly, the SufB and IscU scaffolds under oxidative stress and iron limitation. We show that the IscU Fe2S2 cluster is less stable than the SufB Fe2S2 cluster in the presence of hydrogen peroxide, oxygen, and an iron chelator.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Estresse Fisiológico/fisiologia , Proteínas de Transporte/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Peróxido de Hidrogênio/metabolismo , Proteínas Ferro-Enxofre/genética , Oxigênio/metabolismo
11.
J Am Chem Soc ; 136(14): 5253-6, 2014 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-24650327

RESUMO

Quinolinate synthase (NadA) is a Fe4S4 cluster-containing dehydrating enzyme involved in the synthesis of quinolinic acid (QA), the universal precursor of the essential nicotinamide adenine dinucleotide (NAD) coenzyme. A previously determined apo NadA crystal structure revealed the binding of one substrate analog, providing partial mechanistic information. Here, we report on the holo X-ray structure of NadA. The presence of the Fe4S4 cluster generates an internal tunnel and a cavity in which we have docked the last precursor to be dehydrated to form QA. We find that the only suitably placed residue to initiate this process is the conserved Tyr21. Furthermore, Tyr21 is close to a conserved Thr-His-Glu triad reminiscent of those found in proteases and other hydrolases. Our mutagenesis data show that all of these residues are essential for activity and strongly suggest that Tyr21 deprotonation, to form the reactive nucleophilic phenoxide anion, is mediated by the triad. NadA displays a dehydration mechanism significantly different from the one found in archetypical dehydratases such as aconitase, which use a serine residue deprotonated by an oxyanion hole. The X-ray structure of NadA will help us unveil its catalytic mechanism, the last step in the understanding of NAD biosynthesis.


Assuntos
Hidrolases/química , Complexos Multienzimáticos/química , Tirosina/química , Cristalografia por Raios X , Desidratação , Hidrolases/metabolismo , Modelos Moleculares , Estrutura Molecular , Complexos Multienzimáticos/metabolismo , Tirosina/metabolismo
12.
Mol Microbiol ; 87(3): 493-508, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23320508

RESUMO

The multi-proteins Isc and Suf systems catalyse the biogenesis of [Fe-S] proteins. Here we investigate how NsrR and IscR, transcriptional regulators that sense NO and [Fe-S] homeostasis, acquire their [Fe-S] clusters under both normal and iron limitation conditions. Clusters directed at the apo-NsrR and apo-IscR proteins are built on either of the two scaffolds, IscU or SufB. However, differences arise in [Fe-S] delivery steps. In the case of NsrR, scaffolds deliver clusters to either one of the two ATCs, IscA and SufA, and, subsequently, to the 'non-Isc non-Suf' ATC, ErpA. Nevertheless, a high level of SufA can bypass the requirement for ErpA. In the case of IscR, several routes occur. One does not include assistance of any ATC. Others implicate ATCs IscA or ErpA, but, surprisingly, SufA was totally absent from any IscR maturation pathways. Both IscR and NsrR have the intrinsic capacity to sense iron limitation. However, NsrR appeared to be efficiently matured by Isc and Suf, thereby preventing NsrR to act as a physiologically relevant iron sensor. This work emphasizes that different maturation pathways arise as a function of the apo-target considered, possibly in relation with the type of cluster, [2Fe-2S] versus [4Fe-4S], it binds.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Ferro/metabolismo , Enxofre/metabolismo , Fatores de Transcrição/metabolismo , Processamento de Proteína Pós-Traducional
13.
J Am Chem Soc ; 135(2): 733-40, 2013 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-23265191

RESUMO

Iron-sulfur (Fe-S) cluster-containing proteins are essential components of cells. In eukaryotes, Fe-S clusters are synthesized by the mitochondrial iron-sulfur cluster (ISC) machinery and the cytosolic iron-sulfur assembly (CIA) system. In the mammalian ISC machinery, preassembly of the Fe-S cluster on the scaffold protein (ISCU) involves a cysteine desulfurase complex (NFS1/ISD11) and frataxin (FXN), the protein deficient in Friedreich's ataxia. Here, by comparing the biochemical and spectroscopic properties of quaternary (ISCU/NFS1/ISD11/FXN) and ternary (ISCU/NFS1/ISD11) complexes, we show that FXN stabilizes the quaternary complex and controls iron entry to the complex through activation of cysteine desulfurization. Furthermore, we show for the first time that in the presence of iron and L-cysteine, an [Fe(4)S(4)] cluster is formed within the quaternary complex that can be transferred to mammalian aconitase (mACO2) to generate an active enzyme. In the absence of FXN, although the ternary complex can assemble an Fe-S cluster, the cluster is inefficiently transferred to ACO2. Taken together, these data help to unravel further the Fe-S cluster assembly process and the molecular basis of Friedreich's ataxia.


Assuntos
Proteínas de Ligação ao Ferro/fisiologia , Proteínas Ferro-Enxofre/química , Ferro/metabolismo , Enxofre/metabolismo , Animais , Complexos de Coordenação/química , Humanos , Modelos Moleculares , Frataxina
14.
Mol Microbiol ; 86(1): 155-71, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22966982

RESUMO

Biosynthesis of iron-sulphur (Fe-S) proteins is catalysed by multi-protein systems, ISC and SUF. However, 'non-ISC, non-SUF' Fe-S biosynthesis factors have been described, both in prokaryotes and eukaryotes. Here we report in vitro and in vivo investigations of such a 'non-ISC, non SUF' component, the Nfu proteins. Phylogenomic analysis allowed us to define four subfamilies. Escherichia coli NfuA is within subfamily II. Most members of this subfamily have a Nfu domain fused to a 'degenerate' A-type carrier domain (ATC*) lacking Fe-S cluster co-ordinating Cys ligands. The Nfu domain binds a [4Fe-4S] cluster while the ATC* domain interacts with NuoG (a complex I subunit) and aconitase B (AcnB). In vitro, holo-NfuA promotes maturation of AcnB. In vivo, NfuA is necessary for full activity of complex I under aerobic growth conditions, and of AcnB in the presence of superoxide. NfuA receives Fe-S clusters from IscU/HscBA and SufBCD scaffolds and eventually transfers them to the ATCs IscA and SufA. This study provides significant information on one of the Fe-S biogenesis factors that has been often used as a building block by ISC and/or SUF synthesizing organisms, including bacteria, plants and animals.


Assuntos
Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Proteínas Ferro-Enxofre/genética , Proteínas Ferro-Enxofre/metabolismo , Aconitato Hidratase/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Filogenia , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
15.
Biomolecules ; 13(5)2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-37238602

RESUMO

Iron-sulfur (Fe-S) clusters are inorganic prosthetic groups in proteins composed exclusively of iron and inorganic sulfide. These cofactors are required in a wide range of critical cellular pathways. Iron-sulfur clusters do not form spontaneously in vivo; several proteins are required to mobilize sulfur and iron, assemble and traffic-nascent clusters. Bacteria have developed several Fe-S assembly systems, such as the ISC, NIF, and SUF systems. Interestingly, in Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), the SUF machinery is the primary Fe-S biogenesis system. This operon is essential for the viability of Mtb under normal growth conditions, and the genes it contains are known to be vulnerable, revealing the Mtb SUF system as an interesting target in the fight against tuberculosis. In the present study, two proteins of the Mtb SUF system were characterized for the first time: Rv1464(sufS) and Rv1465(sufU). The results presented reveal how these two proteins work together and thus provide insights into Fe-S biogenesis/metabolism by this pathogen. Combining biochemistry and structural approaches, we showed that Rv1464 is a type II cysteine-desulfurase enzyme and that Rv1465 is a zinc-dependent protein interacting with Rv1464. Endowed with a sulfurtransferase activity, Rv1465 significantly enhances the cysteine-desulfurase activity of Rv1464 by transferring the sulfur atom from persulfide on Rv1464 to its conserved Cys40 residue. The zinc ion is important for the sulfur transfer reaction between SufS and SufU, and His354 in SufS plays an essential role in this reaction. Finally, we showed that Mtb SufS-SufU is more resistant to oxidative stress than E. coli SufS-SufE and that the presence of zinc in SufU is likely responsible for this improved resistance. This study on Rv1464 and Rv1465 will help guide the design of future anti-tuberculosis agents.


Assuntos
Escherichia coli , Mycobacterium tuberculosis , Escherichia coli/metabolismo , Mycobacterium tuberculosis/metabolismo , Cisteína/metabolismo , Zinco/metabolismo , Liases de Carbono-Enxofre/química , Liases de Carbono-Enxofre/genética , Liases de Carbono-Enxofre/metabolismo , Enxofre/metabolismo , Ferro/metabolismo
16.
Angew Chem Int Ed Engl ; 51(31): 7711-4, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22715136

RESUMO

Stop for NadA! A [4Fe-4S] enzyme, NadA, catalyzes the formation of quinolinic acid in de novo nicotinamide adenine dinucleotide (NAD) biosynthesis. A structural analogue of an intermediate, 4,5-dithiohydroxyphthalic acid (DTHPA), has an in vivo NAD biosynthesis inhibiting activity in E. coli. The inhibitory effect can be explained by the coordination of DTHPA thiolate groups to a unique Fe site of the NadA [4Fe-4S] cluster.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Fosfato de Di-Hidroxiacetona/farmacologia , Inibidores Enzimáticos/farmacologia , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas Ferro-Enxofre/antagonistas & inibidores , Alquil e Aril Transferases/metabolismo , Sítios de Ligação/efeitos dos fármacos , Fosfato de Di-Hidroxiacetona/química , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Proteínas de Escherichia coli/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
17.
Nat Ecol Evol ; 6(10): 1564-1572, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36109654

RESUMO

Iron-sulfur (Fe-S) clusters are ubiquitous cofactors essential for life. It is largely thought that the emergence of oxygenic photosynthesis and progressive oxygenation of the atmosphere led to the origin of multiprotein machineries (ISC, NIF and SUF) assisting Fe-S cluster synthesis in the presence of oxidative stress and shortage of bioavailable iron. However, previous analyses have left unclear the origin and evolution of these systems. Here, we combine exhaustive homology searches with genomic context analysis and phylogeny to precisely identify Fe-S cluster biogenesis systems in over 10,000 archaeal and bacterial genomes. We highlight the existence of two additional and clearly distinct 'minimal' Fe-S cluster assembly machineries, MIS (minimal iron-sulfur) and SMS (SUF-like minimal system), which we infer in the last universal common ancestor (LUCA) and we experimentally validate SMS as a bona fide Fe-S cluster biogenesis system. These ancestral systems were kept in archaea whereas they went through stepwise complexification in bacteria to incorporate additional functions for higher Fe-S cluster synthesis efficiency leading to SUF, ISC and NIF. Horizontal gene transfers and losses then shaped the current distribution of these systems, driving ecological adaptations such as the emergence of aerobic lifestyles in archaea. Our results show that dedicated machineries were in place early in evolution to assist Fe-S cluster biogenesis and that their origin is not directly linked to Earth oxygenation.


Assuntos
Proteínas Ferro-Enxofre , Genoma Bacteriano , Ferro , Proteínas Ferro-Enxofre/genética , Filogenia , Enxofre/metabolismo
18.
Elife ; 112022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35244541

RESUMO

Iron-sulfur (Fe-S) clusters are ancient and ubiquitous protein cofactors and play irreplaceable roles in many metabolic and regulatory processes. Fe-S clusters are built and distributed to Fe-S enzymes by dedicated protein networks. The core components of these networks are widely conserved and highly versatile. However, Fe-S proteins and enzymes are often inactive outside their native host species. We sought to systematically investigate the compatibility of Fe-S networks with non-native Fe-S enzymes. By using collections of Fe-S enzyme orthologs representative of the entire range of prokaryotic diversity, we uncovered a striking correlation between phylogenetic distance and probability of functional expression. Moreover, coexpression of a heterologous Fe-S biogenesis pathway increases the phylogenetic range of orthologs that can be supported by the foreign host. We also find that Fe-S enzymes that require specific electron carrier proteins are rarely functionally expressed unless their taxon-specific reducing partners are identified and co-expressed. We demonstrate how these principles can be applied to improve the activity of a radical S-adenosyl methionine(rSAM) enzyme from a Streptomyces antibiotic biosynthesis pathway in Escherichia coli. Our results clarify how oxygen sensitivity and incompatibilities with foreign Fe-S and electron transfer networks each impede heterologous activity. In particular, identifying compatible electron transfer proteins and heterologous Fe-S biogenesis pathways may prove essential for engineering functional Fe-S enzyme-dependent pathways.


Assuntos
Proteínas de Escherichia coli , Proteínas Ferro-Enxofre , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Ferro/metabolismo , Proteínas Ferro-Enxofre/genética , Proteínas Ferro-Enxofre/metabolismo , Filogenia , Enxofre/metabolismo
19.
J Biol Chem ; 285(30): 23331-41, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20460376

RESUMO

Assembly of iron-sulfur (Fe-S) clusters and maturation of Fe-S proteins in vivo require complex machineries. In Escherichia coli, under adverse stress conditions, this process is achieved by the SUF system that contains six proteins as follows: SufA, SufB, SufC, SufD, SufS, and SufE. Here, we provide a detailed characterization of the SufBCD complex whose function was so far unknown. Using biochemical and spectroscopic analyses, we demonstrate the following: (i) the complex as isolated exists mainly in a 1:2:1 (B:C:D) stoichiometry; (ii) the complex can assemble a [4Fe-4S] cluster in vitro and transfer it to target proteins; and (iii) the complex binds one molecule of flavin adenine nucleotide per SufBC(2)D complex, only in its reduced form (FADH(2)), which has the ability to reduce ferric iron. These results suggest that the SufBC(2)D complex functions as a novel type of scaffold protein that assembles an Fe-S cluster through the mobilization of sulfur from the SufSE cysteine desulfurase and the FADH(2)-dependent reductive mobilization of iron.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Flavina-Adenina Dinucleotídeo/análogos & derivados , Proteínas Ferro-Enxofre/química , Proteínas Ferro-Enxofre/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Anaerobiose , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Escherichia coli/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Dados de Sequência Molecular , Oxidantes/metabolismo , Oxirredução
20.
ACS Chem Biol ; 16(11): 2423-2433, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34609124

RESUMO

Quinolinate synthase, also called NadA, is a [4Fe-4S]-containing enzyme that uses what is probably the oldest pathway to generate quinolinic acid (QA), the universal precursor of the biologically essential cofactor nicotinamide adenine dinucleotide (NAD). Its synthesis comprises the condensation of dihydroxyacetone phosphate (DHAP) and iminoaspartate (IA), which involves dephosphorylation, isomerization, cyclization, and two dehydration steps. The convergence of the three homologous domains of NadA defines a narrow active site that contains a catalytically essential [4Fe-4S] cluster. A tunnel, which can be opened or closed depending on the nature (or absence) of the bound ligand, connects this cofactor to the protein surface. One outstanding riddle has been the observation that the so far characterized active site is too small to bind IA and DHAP simultaneously. Here, we have used site-directed mutagenesis, X-ray crystallography, functional analyses, and molecular dynamics simulations to propose a condensation mechanism that involves the transient formation of a second active site cavity to which one of the substrates can migrate before this reaction takes place.


Assuntos
Complexos Multienzimáticos/química , Ácido Quinolínico/química , Catálise , Domínio Catalítico , Cristalografia por Raios X , Fosfato de Di-Hidroxiacetona/química , Modelos Moleculares , Complexos Multienzimáticos/metabolismo , Conformação Proteica , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa