Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 280
Filtrar
1.
J Biol Chem ; 298(7): 102094, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35654137

RESUMEN

The cytosolic iron-sulfur (Fe-S) cluster assembly (CIA) pathway delivers Fe-S clusters to nuclear and cytosolic Fe-S proteins involved in essential cellular functions. Although the delivery process is regulated by the availability of iron and oxygen, it remains unclear how CIA components orchestrate the cluster transfer under varying cellular environments. Here, we utilized a targeted proteomics assay for monitoring CIA factors and substrates to characterize the CIA machinery. We find that nucleotide-binding protein 1 (NUBP1/NBP35), cytosolic iron-sulfur assembly component 3 (CIAO3/NARFL), and CIA substrates associate with nucleotide-binding protein 2 (NUBP2/CFD1), a component of the CIA scaffold complex. NUBP2 also weakly associates with the CIA targeting complex (MMS19, CIAO1, and CIAO2B) indicating the possible existence of a higher order complex. Interactions between CIAO3 and the CIA scaffold complex are strengthened upon iron supplementation or low oxygen tension, while iron chelation and reactive oxygen species weaken CIAO3 interactions with CIA components. We further demonstrate that CIAO3 mutants defective in Fe-S cluster binding fail to integrate into the higher order complexes. However, these mutants exhibit stronger associations with CIA substrates under conditions in which the association with the CIA targeting complex is reduced suggesting that CIAO3 and CIA substrates may associate in complexes independently of the CIA targeting complex. Together, our data suggest that CIA components potentially form a metabolon whose assembly is regulated by environmental cues and requires Fe-S cluster incorporation in CIAO3. These findings provide additional evidence that the CIA pathway adapts to changes in cellular environment through complex reorganization.


Asunto(s)
Proteínas Hierro-Azufre , Hierro , Citosol/metabolismo , Proteínas de Unión al GTP/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hierro/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Proteínas Hierro-Azufre/metabolismo , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Azufre/metabolismo
2.
mBio ; 12(6): e0242521, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34781750

RESUMEN

Building iron-sulfur (Fe-S) clusters and assembling Fe-S proteins are essential actions for life on Earth. The three processes that sustain life, photosynthesis, nitrogen fixation, and respiration, require Fe-S proteins. Genes coding for Fe-S proteins can be found in nearly every sequenced genome. Fe-S proteins have a wide variety of functions, and therefore, defective assembly of Fe-S proteins results in cell death or global metabolic defects. Compared to alternative essential cellular processes, there is less known about Fe-S cluster synthesis and Fe-S protein maturation. Moreover, new factors involved in Fe-S protein assembly continue to be discovered. These facts highlight the growing need to develop a deeper biological understanding of Fe-S cluster synthesis, holo-protein maturation, and Fe-S cluster repair. Here, we outline bacterial strategies used to assemble Fe-S proteins and the genetic regulation of these processes. We focus on recent and relevant findings and discuss future directions, including the proposal of using Fe-S protein assembly as an antipathogen target.


Asunto(s)
Bacterias/metabolismo , Proteínas Bacterianas/biosíntesis , Proteínas Hierro-Azufre/biosíntesis , Bacterias/genética , Proteínas Bacterianas/genética , Hierro/metabolismo , Proteínas Hierro-Azufre/genética , Azufre/metabolismo
3.
Mol Microbiol ; 116(2): 606-623, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34032321

RESUMEN

The malaria parasite harbors two [Fe-S] biogenesis pathways of prokaryotic origin-the SUF and ISC systems in the apicoplast and mitochondrion, respectively. While the SUF machinery has been delineated, there is little experimental evidence on the ISC pathway. We confirmed mitochondrial targeting of Plasmodium falciparum ISC proteins followed by analyses of cysteine desulfurase, scaffold, and [Fe-S]-carrier components. PfIscU functioned as the scaffold in complex with the PfIscS-PfIsd11 cysteine desulfurase and could directly assemble [4Fe-4S] without prior [2Fe-2S] formation seen in other homologs. Small angle X-ray scattering and spectral studies showed that PfIscU, a trimer, bound one [4Fe-4S]. In a deviation from reported complexes from other organisms, the P. falciparum desulfurase-scaffold complex assembled around a PfIscS tetramer instead of a dimer, resulting in a symmetric hetero-hexamer [2× (2PfIscS-2PfIsd11-2PfIscU)]. PfIscU directly transferred [4Fe-4S] to the apo-protein aconitase B thus abrogating the requirement of intermediary proteins for conversion of [2Fe-2S] to [4Fe-4S] before transfer to [4Fe-4S]-recipients. Among the putative cluster-carriers, PfIscA2 was more efficient than PfNifU-like protein; PfIscA1 primarily bound iron, suggesting its potential role as a Fe2+ carrier/donor. Our results identify the core P. falciparum ISC machinery and reveal unique features compared with those in bacteria or yeast and human mitochondria.


Asunto(s)
Liasas de Carbono-Azufre/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Mitocondrias/metabolismo , Plasmodium falciparum/metabolismo , Aconitato Hidratasa/metabolismo , Proteínas Portadoras/metabolismo , Humanos , Malaria Falciparum/patología , Multimerización de Proteína
4.
Int J Mol Sci ; 21(18)2020 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-32957566

RESUMEN

Several biological activities depend on iron-sulfur clusters ([Fe-S]). Even though they are well-known in several organisms their function and metabolic pathway were poorly understood in the majority of the organisms. We propose to use the amoeba Dictyostelium discoideum, as a biological model to study the biosynthesis of [Fe-S] at the molecular, cellular and organism levels. First, we have explored the D. discoideum genome looking for genes corresponding to the subunits that constitute the molecular machinery for Fe-S cluster assembly and, based on the structure of the mammalian supercomplex and amino acid conservation profiles, we inferred the full functionality of the amoeba machinery. After that, we expressed the recombinant mature form of D. discoideum frataxin protein (DdFXN), the kinetic activator of this pathway. We characterized the protein and its conformational stability. DdFXN is monomeric and compact. The analysis of the secondary structure content, calculated using the far-UV CD spectra, was compatible with the data expected for the FXN fold, and near-UV CD spectra were compatible with the data corresponding to a folded protein. In addition, Tryptophan fluorescence indicated that the emission occurs from an apolar environment. However, the conformation of DdFXN is significantly less stable than that of the human FXN, (4.0 vs. 9.0 kcal mol-1, respectively). Based on a sequence analysis and structural models of DdFXN, we investigated key residues involved in the interaction of DdFXN with the supercomplex and the effect of point mutations on the energetics of the DdFXN tertiary structure. More than 10 residues involved in Friedreich's Ataxia are conserved between the human and DdFXN forms, and a good correlation between mutational effect on the energetics of both proteins were found, suggesting the existence of similar sequence/function/stability relationships. Finally, we integrated this information in an evolutionary context which highlights particular variation patterns between amoeba and humans that may reflect a functional importance of specific protein positions. Moreover, the complete pathway obtained forms a piece of evidence in favor of the hypothesis of a shared and highly conserved [Fe-S] assembly machinery between Human and D. discoideum.


Asunto(s)
Dictyostelium/metabolismo , Ataxia de Friedreich/genética , Proteínas de Unión a Hierro/química , Proteínas de Unión a Hierro/metabolismo , Proteínas Hierro-Azufre/metabolismo , Secuencia de Aminoácidos/genética , Cromatografía Líquida de Alta Presión , Dicroismo Circular , Biología Computacional , Cristalografía , Dictyostelium/genética , Humanos , Proteínas de Unión a Hierro/genética , Proteínas Hierro-Azufre/biosíntesis , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Cinética , Simulación de Dinámica Molecular , Filogenia , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes , Alineación de Secuencia , Espectrometría de Fluorescencia , Espectrofotometría Ultravioleta , Frataxina
5.
Biotechnol Appl Biochem ; 67(4): 574-585, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32770861

RESUMEN

We explore the capacity of the de novo protein, S824, to incorporate a multinuclear iron-sulfur cluster within the core of a single-chain four-helix bundle. This topology has a high intrinsic designability because sequences are constrained largely by the pattern of hydrophobic and hydrophilic amino acids, thereby allowing for the extensive substitution of individual side chains. Libraries of novel proteins based on these constraints have surprising functional potential and have been shown to complement the deletion of essential genes in E. coli. Our structure-based design of four first-shell cysteine ligands, one per helix, in S824 resulted in successful incorporation of a cubane Fe4 S4 cluster into the protein core. A number of challenges were encountered during the design and characterization process, including nonspecific metal-induced aggregation and the presence of competing metal-cluster stoichiometries. The introduction of buried iron-sulfur clusters into the helical bundle is an initial step toward converting libraries of designed structures into functional de novo proteins with catalytic or electron-transfer functionalities.


Asunto(s)
Escherichia coli , Proteínas Hierro-Azufre , Ingeniería de Proteínas , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Conformación Proteica en Hélice alfa
6.
Int J Mol Sci ; 21(10)2020 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-32423011

RESUMEN

Sulfur is an essential element required for plant growth. It can be found as a thiol group of proteins or non-protein molecules, and as various sulfur-containing small biomolecules, including iron-sulfur (Fe/S) clusters, molybdenum cofactor (Moco), and sulfur-modified nucleotides. Thiol-mediated redox regulation has been well investigated, whereas biosynthesis pathways of the sulfur-containing small biomolecules have not yet been clearly described. In order to understand overall sulfur transfer processes in plant cells, it is important to elucidate the relationships among various sulfur delivery pathways as well as to investigate their interactions. In this review, we summarize the information from recent studies on the biosynthesis pathways of several sulfur-containing small biomolecules and the proteins participating in these processes. In addition, we show characteristic features of gene expression in Arabidopsis at the early stage of sulfate depletion from the medium, and we provide insights into sulfur transfer processes in plant cells.


Asunto(s)
Liasas de Carbono-Azufre/biosíntesis , Proteínas Hierro-Azufre/biosíntesis , Azufre/metabolismo , Sulfurtransferasas/biosíntesis , Vías Biosintéticas/genética , Liasas de Carbono-Azufre/genética , Coenzimas , Proteínas Hierro-Azufre/genética , Metaloproteínas , Cofactores de Molibdeno , Plantas/metabolismo , Pteridinas , Compuestos de Sulfhidrilo/metabolismo , Sulfurtransferasas/genética
7.
Genome Biol Evol ; 12(3): 160-173, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32108236

RESUMEN

Iron-sulfur (Fe-S) clusters play important roles in electron transfer, metabolic and biosynthetic reactions, and the regulation of gene expression. Understanding the biogenesis of Fe-S clusters is therefore relevant to many fields. In the complex process of Fe-S protein formation, the A-type assembly protein (ATAP) family, which consists of several subfamilies, plays an essential role in Fe-S cluster formation and transfer and is highly conserved across the tree of life. However, the taxonomic distribution, motif compositions, and the evolutionary history of the ATAP subfamilies are not well understood. To address these problems, our study investigated the taxonomic distribution of 321 species from a broad cross-section of taxa. Then, we identified common and specific motifs in multiple ATAP subfamilies to explain the functional conservation and nonredundancy of the ATAPs, and a novel, essential motif was found in Eumetazoa IscA1, which has a newly found magnetic function. Finally, we used phylogenetic analytical methods to reconstruct the evolution history of this family. Our results show that two types of ErpA proteins (nonproteobacteria-type ErpA1 and proteobacteria-type ErpA2) exist in bacteria. The ATAP family, consisting of seven subfamilies, can be further classified into two types of ATAPs. Type-I ATAPs include IscA, SufA, HesB, ErpA1, and IscA1, with an ErpA1-like gene as their last common ancestor, whereas type-II ATAPs consist of ErpA2 and IscA2, duplicated from an ErpA2-like gene. During the mitochondrial endosymbiosis, IscA became IscA1 in eukaryotes and ErpA2 became IscA2 in eukaryotes, respectively.


Asunto(s)
Evolución Molecular , Duplicación de Gen , Proteínas Hierro-Azufre/biosíntesis , Secuencias de Aminoácidos/genética , Proteínas Bacterianas/genética , Filogenia
8.
Microbiology (Reading) ; 166(3): 296-305, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31860439

RESUMEN

Iron-sulphur (FeS) clusters are versatile cofactors required for a range of biological processes within cells. Due to the reactive nature of the constituent molecules, assembly and delivery of these cofactors requires a multi-protein machinery in vivo. In prokaryotes, SufT homologues are proposed to function in the maturation and transfer of FeS clusters to apo-proteins. This study used targeted gene deletion to investigate the role of SufT in the physiology of mycobacteria, using Mycobacterium smegmatis as a model organism. Deletion of the sufT gene in M. smegmatis had no impact on growth under standard culture conditions and did not significantly alter activity of the FeS cluster dependent enzymes succinate dehydrogenase (SDH) and aconitase (ACN). Furthermore, the ΔsufT mutant was no more sensitive than the wild-type strain to the redox cycler 2,3-dimethoxy-1,4-naphthoquinone (DMNQ), or the anti-tuberculosis drugs isoniazid, clofazimine or rifampicin. In contrast, the ΔsufT mutant displayed a growth defect under iron limiting conditions, and an increased requirement for iron during biofilm formation. This data suggests that SufT is an accessory factor in FeS cluster biogenesis in mycobacteria which is required under conditions of iron limitation.


Asunto(s)
Coenzimas/genética , Proteínas Hierro-Azufre/metabolismo , Hierro/metabolismo , Mycobacterium smegmatis , Aconitato Hidratasa/metabolismo , Proteínas Bacterianas/genética , Biopelículas , Eliminación de Gen , Proteínas Hierro-Azufre/biosíntesis , Proteínas Hierro-Azufre/genética , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/metabolismo , Succinato Deshidrogenasa/metabolismo
9.
Mol Omics ; 15(5): 340-347, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31429849

RESUMEN

Adrenodoxin reductase, a widely conserved mitochondrial P450 protein, catalyses essential steps in steroid hormone biosynthesis and is highly expressed in the adrenal cortex. The yeast adrenodoxin reductase homolog, Arh1p, is involved in cytoplasmic and mitochondrial iron homeostasis and is required for activity of enzymes containing an Fe-S cluster. In this paper, we investigated the response of yeast to the loss of a single copy of ARH1, an oxidoreductase of the mitochondrial inner membrane, which is among the few mitochondrial proteins that is essential for viability in yeast. The phenotypic, transcriptional, proteomic, and metabolic landscape indicated that Saccharomyces cerevisiae successfully adapted to this loss, displaying an apparently dosage-insensitive cellular response. However, a considered investigation of transcriptional regulation in ARH1-impaired yeast highlighted that a significant hierarchical reorganisation occurred, involving the iron assimilation and tyrosine biosynthetic processes. The interconnected roles of the iron and tyrosine pathways, coupled with oxidative processes, are of interest beyond yeast since they are involved in dopaminergic neurodegeneration associated with Parkinson's disease. The identification of similar responses in yeast, albeit preliminary, suggests that this simple eukaryote could have potential as a model system for investigating the regulatory mechanisms leading to the initiation and progression of early disease responses in humans.


Asunto(s)
Ferredoxina-NADP Reductasa/metabolismo , Haploinsuficiencia , Proteínas de la Membrana/metabolismo , Enfermedad de Parkinson/metabolismo , Ploidias , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Biología Computacional , Ferredoxina-NADP Reductasa/genética , Regulación Enzimológica de la Expresión Génica , Regulación Fúngica de la Expresión Génica , Humanos , Proteínas Hierro-Azufre/biosíntesis , Proteínas de la Membrana/genética , Mutación , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
10.
Biochim Biophys Acta Mol Cell Res ; 1866(2): 240-251, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30419257

RESUMEN

Iron­sulfur (Fe/S) clusters are versatile inorganic cofactors that play central roles in essential cellular functions, from respiration to genome stability. >30 proteins involved in Fe/S protein biogenesis in eukaryotes are known, many of which bind clusters via cysteine residues. This opens up the possibility that the thiol-reducing glutaredoxin and thioredoxin systems are required at both the Fe/S biogenesis and target protein level to counteract thiol oxidation. To address the possible interplay of thiol redox chemistry and Fe/S protein biogenesis, we have characterized the status of the mitochondrial (ISC) and cytosolic (CIA) Fe/S protein assembly machineries in Saccharomyces cerevisiae mutants in which the three partially redundant glutathione (Glr1) and thioredoxin (Trr1 and Trr2) oxidoreductases have been inactivated in either mitochondria, cytosol, or both compartments. Cells devoid of mitochondrial oxidoreductases maintained a functional mitochondrial ISC machinery and showed no altered iron homeostasis despite a non-functional complex II of the respiratory chain due to redox-specific defects. In cells that lack either cytosolic or total cellular thiol reducing capacity, both the ISC system and iron homeostasis were normal, yet cytosolic and nuclear Fe/S target proteins were not matured. This dysfunction could be attributed to a failure in the assembly of [4Fe­4S] clusters in the CIA factor Nar1, even though Nar1 maintained robust protein levels and stable interactions with later-acting CIA components. Overall, our analysis has uncovered a hitherto unknown thiol-dependence of the CIA machinery and has demonstrated the surprisingly varying sensitivity of Fe/S proteins to thiol oxidation.


Asunto(s)
Proteínas Hierro-Azufre/biosíntesis , Proteínas Hierro-Azufre/metabolismo , Núcleo Celular/metabolismo , Citosol/metabolismo , Inestabilidad Genómica , Glutarredoxinas/metabolismo , Homeostasis , Hierro/metabolismo , Proteínas Hierro-Azufre/genética , Mitocondrias/metabolismo , Oxidación-Reducción , Transporte de Proteínas , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Compuestos de Sulfhidrilo/metabolismo , Azufre/metabolismo , Tiorredoxinas/metabolismo
11.
Molecules ; 23(9)2018 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-30200358

RESUMEN

Iron-sulfur (Fe-S) clusters, the ubiquitous protein cofactors found in all kingdoms of life, perform a myriad of functions including nitrogen fixation, ribosome assembly, DNA repair, mitochondrial respiration, and metabolite catabolism. The biogenesis of Fe-S clusters is a multi-step process that involves the participation of many protein partners. Recent biophysical studies, involving X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy, mass spectrometry (MS), and small angle X-ray scattering (SAXS), have greatly improved our understanding of these steps. In this review, after describing the biological importance of iron sulfur proteins, we focus on the contributions of NMR spectroscopy has made to our understanding of the structures, dynamics, and interactions of proteins involved in the biosynthesis of Fe-S cluster proteins.


Asunto(s)
Citosol/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Espectroscopía de Resonancia Magnética/métodos , Mitocondrias/metabolismo , Animales , Humanos , Modelos Biológicos
12.
FEBS Lett ; 592(24): 4011-4019, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30194723

RESUMEN

Iron-sulfur cluster biogenesis is a complex process mediated by numerous proteins among which two from bacteria chaperones, called HscB and HscA in bacteria. They are highly conserved up to eukaryotes and homologous to DnaJ and DnaK, respectively, but with specific differences. As compared with other chaperones, HscB and HscA have escaped attention and relatively little is known about their functions. After briefly introducing the various chaperone families, we reviewed here the current structural and functional knowledge HscA and HscB and on their role in cluster formation. We critically evaluated the literature and highlighted the weak aspects which will require more attention in the future. We sincerely hope that this study will inspire new interest on this important and interesting system.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Hierro-Azufre/genética , Chaperonas Moleculares/genética , Familia de Multigenes , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Dominios Proteicos
13.
Genome Biol Evol ; 10(8): 2061-2071, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30085124

RESUMEN

Plastids surrounded by four membranes harbor a special compartment between the outer and inner plastid membrane pair, the so-called periplastidal compartment (PPC). This cellular structure is usually presumed to be the reduced cytoplasm of a eukaryotic phototrophic endosymbiont, which was integrated into a host cell and streamlined into a plastid with a complex membrane structure. Up to date, no mitochondrion or mitochondrion-related organelle has been identified in the PPC of any representative. However, two prominent groups, the cryptophytes and the chlorarachniophytes, still harbor a reduced cell nucleus of symbiont origin, the nucleomorph, in their PPCs. Generally, many cytoplasmic and nucleus-located eukaryotic proteins need an iron-sulfur cofactor for their functionality. Beside some exceptions, their synthesis is depending on a so-called iron-sulfur complex (ISC) assembly machinery located in the mitochondrion. This machinery provides the cytoplasm with a still unknown sulfur component, which is then converted into iron-sulfur clusters via a cytosolic iron-sulfur protein assembly (CIA) machinery. Here, we investigated if a CIA machinery is present in mitochondrion-lacking PPCs. By using bioinformatic screens and in vivo-localizations of candidate proteins, we show that the presence of a PPC-specific CIA machinery correlates with the presence of a nucleomorph. Phylogenetic analyses of PPC- and host specific CIA components additionally indicate a complex evolution of the CIA machineries in organisms having plastids surrounded by four membranes.


Asunto(s)
Criptófitas/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Plastidios/metabolismo , Compartimento Celular , Criptófitas/genética , Citosol/metabolismo , Diatomeas/genética , Genoma , Modelos Biológicos , Filogenia
14.
Chem Commun (Camb) ; 54(62): 8614-8617, 2018 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-30019036

RESUMEN

We demonstrate the synthesis and spectroscopic characterization of an unusual high spin (S = 7/2) [Cu-3Fe-4S] cluster in a member of the radical-SAM enzymes. This is the first step in using synthetic [Me-3Fe-4S] clusters for obtaining new insight into the mechanism of radical-SAM catalysis.


Asunto(s)
Cobre/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Proteínas/metabolismo , S-Adenosilmetionina/metabolismo , Cobre/química , Radicales Libres/química , Radicales Libres/metabolismo , Humanos , Proteínas Hierro-Azufre/química , Estructura Molecular , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Proteínas/química , S-Adenosilmetionina/química
15.
Metallomics ; 10(8): 1038-1052, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30019043

RESUMEN

Iron-sulphur (Fe-S) clusters are versatile cofactors, which are essential for key metabolic processes in cells, such as respiration and photosynthesis, and which may have also played a crucial role in establishing life on Earth. They can be found in almost all living organisms, from unicellular prokaryotes and archaea to multicellular animals and plants, and exist in diverse forms. This review focuses on the most ancient Fe-S cluster assembly system, the sulphur utilization factor (SUF) mechanism, which is crucial in bacteria for cell survival under stress conditions such as oxidation and iron starvation, and which is also present in the chloroplasts of green microalgae and plants, where it is responsible for plastidial Fe-S protein maturation. We explain the SUF Fe-S cluster assembly process, the proteins involved, their regulation and provide evolutionary insights. We specifically focus on examples from Fe-S cluster synthesis in the model organisms Escherichia coli and Arabidopsis thaliana and discuss in an in vivo context the assembly of the [FeFe]-hydrogenase H-cluster from Chlamydomonas reinhardtii.


Asunto(s)
Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Hierro/metabolismo , Azufre/metabolismo , Humanos
16.
Biochim Biophys Acta Gen Subj ; 1862(10): 2152-2161, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30025855

RESUMEN

In the eukaryotic model yeast Saccharomyces cerevisiae, arsenic (As) detoxification is regulated by two transcriptional factors, Yap8 and Yap1. Yap8 specifically controls As extrusion from the cell, whether Yap1 avoids arsenic-induced oxidative damages. Accordingly, cells lacking both Yap1 and Yap8 are more sensitive to arsenate than cells lacking each regulator individually. Strikingly enough, the same sensitivity pattern was observed under anoxia, suggesting that Yap1 role in As detoxification might not be restricted to the regulation of the oxidative stress response. This finding prompted us to study the transcriptomic profile of wild-type and yap1 mutant cells exposed to arsenate. Interestingly, we found that, under such conditions, several genes involved in the biogenesis of FeS proteins were upregulated in a Yap1-dependent way. In line with this observation, arsenate treatment decreases the activity of the mitochondrial aconitase, Aco1, an FeS cluster-containing enzyme, this effect being even more pronounced in the yap1 mutant. Reinforcing the relevance of FeS cluster biogenesis in arsenate detoxification, the overexpression of several ISC and CIA machinery genes alleviates the deleterious effect of arsenate caused by the absence of Yap1 and Yap8. Altogether our data suggest that the upregulation of FeS biogenesis genes regulated by Yap1 might work as a cellular shield against arsenate toxicity.


Asunto(s)
Arseniatos/toxicidad , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Proteínas Hierro-Azufre/biosíntesis , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos , Proteínas Hierro-Azufre/efectos de los fármacos , Proteínas Hierro-Azufre/genética , Estrés Oxidativo/efectos de los fármacos , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/genética
17.
J Eukaryot Microbiol ; 65(6): 913-922, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29932290

RESUMEN

Cryptosporidium is a protozoan, apicomplexan, parasite that poses significant risk to humans and animals, as a common cause of potentially fatal diarrhea in immunodeficient hosts. The parasites have evolved a number of unique biological features that allow them to thrive in a highly specialized parasitic lifestyle. For example, the genome of Cryptosporidium parvum is highly reduced, encoding only 3,805 proteins, which is also reflected in its reduced cellular and organellar content and functions. As such, its remnant mitochondrion, dubbed a mitosome, is one of the smallest mitochondria yet found. While numerous studies have attempted to discover the function(s) of the C. parvum mitosome, most of them have been focused on in silico predictions. Here, we have localized components of a biochemical pathway in the C. parvum mitosome, in our investigations into the functions of this peculiar mitochondrial organelle. We have shown that three proteins involved in the mitochondrial iron-sulfur cluster biosynthetic pathway are localized in the organelle, and one of them can functionally replace its yeast homolog. Thus, it seems that the C. parvum mitosome is involved in iron-sulfur cluster biosynthesis, supporting the organellar and cytosolic apoproteins. These results spearhead further research on elucidating the functions of the mitosome and broaden our understanding in the minimalistic adaptations of these organelles.


Asunto(s)
Cryptosporidium parvum/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Orgánulos/metabolismo , Línea Celular , Cryptosporidium parvum/genética , Cryptosporidium parvum/patogenicidad , ADN Recombinante , Genes Protozoarios/genética , Humanos , Proteínas Hierro-Azufre/genética , Mitocondrias/metabolismo , Proteínas Protozoarias/genética
18.
Int J Biol Macromol ; 116: 1128-1145, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29782976

RESUMEN

The pathogenicity of protozoan parasites is frequently attributed to their ability to circumvent the deleterious effects of ROS and Fe-S clusters are among their susceptible targets with paramount importance for parasite survival. The biogenesis of Fe-S clusters is orchestrated by ISC system; the sulfur donor IscS and scaffold protein IscU being its core components. However, among protozoan parasites including Leishmania, no information is available regarding biochemical aspect of IscU, its interaction partners and regulation. Here, we show that Leishmania donovani IscU homolog, LdIscU, readily assembles [2Fe-2S] clusters and, interestingly, follows Michaelis-Menten enzyme kinetics. It is localized in the mitochondria of the parasite and interacts with LdIscS to form a stable complex. Additionally, LdIscU and Fe-S proteins activity is significantly upregulated in resistant isolates and during stationary growth stage indicating an association between them. The differential expression of LdIscU modulated by Fe-S proteins demand suggests its potential role in parasite survival and drug resistance. Thus, our study provides novel insight into the Fe-S scaffold protein of a protozoan parasite.


Asunto(s)
Resistencia a Medicamentos , Regulación de la Expresión Génica , Proteínas Hierro-Azufre/biosíntesis , Leishmania donovani/metabolismo , Proteínas Protozoarias/biosíntesis , Animales , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Leishmania donovani/química , Leishmania donovani/genética , Masculino , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Conejos
20.
J Biol Inorg Chem ; 23(4): 495-506, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29623423

RESUMEN

Iron-sulfur clusters are evolutionarily conserved biological structures which play an important role as cofactor for multiple enzymes in eukaryotic cells. The biosynthesis pathways of the iron-sulfur clusters are located in the mitochondria and in the cytosol. The mitochondrial iron-sulfur cluster biosynthesis pathway (ISC) can be divided into at least twenty enzymatic steps. Since the description of frataxin deficiency as the cause of Friedreich's ataxia, multiple other deficiencies in ISC biosynthesis pathway have been reported. In this paper, an overview is given of the clinical, biochemical and genetic aspects reported in humans affected by a defect in iron-sulfur cluster biosynthesis.


Asunto(s)
Proteínas Hierro-Azufre/biosíntesis , Mitocondrias/genética , Mitocondrias/metabolismo , Humanos , Proteínas Hierro-Azufre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...