Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.299
Filtrar
1.
New Phytol ; 242(6): 2787-2802, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38693568

RESUMEN

Root-knot nematodes (RKN; Meloidogyne species) are plant pathogens that introduce several effectors in their hosts to facilitate infection. The actual targets and functioning mechanism of these effectors largely remain unexplored. This study illuminates the role and interplay of the Meloidogyne javanica nematode effector ROS suppressor (Mj-NEROSs) within the host plant environment. Mj-NEROSs suppresses INF1-induced cell death as well as flg22-induced callose deposition and reactive oxygen species (ROS) production. A transcriptome analysis highlighted the downregulation of ROS-related genes upon Mj-NEROSs expression. NEROSs interacts with the plant Rieske's iron-sulfur protein (ISP) as shown by yeast-two-hybrid and bimolecular fluorescence complementation. Secreted from the subventral pharyngeal glands into giant cells, Mj-NEROSs localizes in the plastids where it interacts with ISP, subsequently altering electron transport rates and ROS production. Moreover, our results demonstrate that isp Arabidopsis thaliana mutants exhibit increased susceptibility to M. javanica, indicating ISP importance for plant immunity. The interaction of a nematode effector with a plastid protein highlights the possible role of root plastids in plant defense, prompting many questions on the details of this process.


Asunto(s)
Arabidopsis , Complejo III de Transporte de Electrones , Inmunidad de la Planta , Plastidios , Especies Reactivas de Oxígeno , Tylenchoidea , Especies Reactivas de Oxígeno/metabolismo , Arabidopsis/parasitología , Arabidopsis/inmunología , Arabidopsis/genética , Tylenchoidea/fisiología , Tylenchoidea/patogenicidad , Animales , Plastidios/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Enfermedades de las Plantas/parasitología , Enfermedades de las Plantas/inmunología , Proteínas del Helminto/metabolismo , Proteínas del Helminto/genética , Regulación de la Expresión Génica de las Plantas , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Unión Proteica , Mutación/genética , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/genética
2.
Metallomics ; 16(5)2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38744662

RESUMEN

Iron-sulfur (Fe-S) clusters are an essential and ubiquitous class of protein-bound prosthetic centers that are involved in a broad range of biological processes (e.g. respiration, photosynthesis, DNA replication and repair and gene regulation) performing a wide range of functions including electron transfer, enzyme catalysis, and sensing. In a general manner, Fe-S clusters can gain or lose electrons through redox reactions, and are highly sensitive to oxidation, notably by small molecules such as oxygen and nitric oxide. The [2Fe-2S] and [4Fe-4S] clusters, the most common Fe-S cofactors, are typically coordinated by four amino acid side chains from the protein, usually cysteine thiolates, but other residues (e.g. histidine, aspartic acid) can also be found. While diversity in cluster coordination ensures the functional variety of the Fe-S clusters, the lack of conserved motifs makes new Fe-S protein identification challenging especially when the Fe-S cluster is also shared between two proteins as observed in several dimeric transcriptional regulators and in the mitoribosome. Thanks to the recent development of in cellulo, in vitro, and in silico approaches, new Fe-S proteins are still regularly identified, highlighting the functional diversity of this class of proteins. In this review, we will present three main functions of the Fe-S clusters and explain the difficulties encountered to identify Fe-S proteins and methods that have been employed to overcome these issues.


Asunto(s)
Proteínas Hierro-Azufre , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Oxidación-Reducción
3.
Proc Natl Acad Sci U S A ; 121(21): e2400740121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38743629

RESUMEN

The biogenesis of iron-sulfur (Fe/S) proteins entails the synthesis and trafficking of Fe/S clusters, followed by their insertion into target apoproteins. In eukaryotes, the multiple steps of biogenesis are accomplished by complex protein machineries in both mitochondria and cytosol. The underlying biochemical pathways have been elucidated over the past decades, yet the mechanisms of cytosolic [2Fe-2S] protein assembly have remained ill-defined. Similarly, the precise site of glutathione (GSH) requirement in cytosolic and nuclear Fe/S protein biogenesis is unclear, as is the molecular role of the GSH-dependent cytosolic monothiol glutaredoxins (cGrxs). Here, we investigated these questions in human and yeast cells by various in vivo approaches. [2Fe-2S] cluster assembly of cytosolic target apoproteins required the mitochondrial ISC machinery, the mitochondrial transporter Atm1/ABCB7 and GSH, yet occurred independently of both the CIA system and cGrxs. This mechanism was strikingly different from the ISC-, Atm1/ABCB7-, GSH-, and CIA-dependent assembly of cytosolic-nuclear [4Fe-4S] proteins. One notable exception to this cytosolic [2Fe-2S] protein maturation pathway defined here was yeast Apd1 which used the CIA system via binding to the CIA targeting complex through its C-terminal tryptophan. cGrxs, although attributed as [2Fe-2S] cluster chaperones or trafficking proteins, were not essential in vivo for delivering [2Fe-2S] clusters to either CIA components or target apoproteins. Finally, the most critical GSH requirement was assigned to Atm1-dependent export, i.e. a step before GSH-dependent cGrxs function. Our findings extend the general model of eukaryotic Fe/S protein biogenesis by adding the molecular requirements for cytosolic [2Fe-2S] protein maturation.


Asunto(s)
Citosol , Glutarredoxinas , Glutatión , Proteínas Hierro-Azufre , Mitocondrias , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Citosol/metabolismo , Proteínas Hierro-Azufre/metabolismo , Humanos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Glutatión/metabolismo , Mitocondrias/metabolismo , Glutarredoxinas/metabolismo , Glutarredoxinas/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Mitocondriales/metabolismo
4.
Nat Commun ; 15(1): 4655, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38821922

RESUMEN

The human AAA-ATPase Bcs1L translocates the fully assembled Rieske iron-sulfur protein (ISP) precursor across the mitochondrial inner membrane, enabling respiratory Complex III assembly. Exactly how the folded substrate is bound to and released from Bcs1L has been unclear, and there has been ongoing debate as to whether subunits of Bcs1L act in sequence or in unison hydrolyzing ATP when moving the protein cargo. Here, we captured Bcs1L conformations by cryo-EM during active ATP hydrolysis in the presence or absence of ISP substrate. In contrast to the threading mechanism widely employed by AAA proteins in substrate translocation, subunits of Bcs1L alternate uniformly between ATP and ADP conformations without detectable intermediates that have different, co-existing nucleotide states, indicating that the subunits act in concert. We further show that the ISP can be trapped by Bcs1 when its subunits are all in the ADP-bound state, which we propose to be released in the apo form.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Difosfato , Adenosina Trifosfato , Microscopía por Crioelectrón , Complejo III de Transporte de Electrones , Adenosina Trifosfato/metabolismo , Hidrólisis , Complejo III de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/química , Humanos , Adenosina Difosfato/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/química , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/química , Conformación Proteica , Pliegue de Proteína , Modelos Moleculares , Transporte de Proteínas
5.
Nat Commun ; 15(1): 4244, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762605

RESUMEN

Cysteine metabolism occurs across cellular compartments to support diverse biological functions and prevent the induction of ferroptosis. Though the disruption of cytosolic cysteine metabolism is implicated in this form of cell death, it is unknown whether the substantial cysteine metabolism resident within the mitochondria is similarly pertinent to ferroptosis. Here, we show that despite the rapid depletion of intracellular cysteine upon loss of extracellular cystine, cysteine-dependent synthesis of Fe-S clusters persists in the mitochondria of lung cancer cells. This promotes a retention of respiratory function and a maintenance of the mitochondrial redox state. Under these limiting conditions, we find that glutathione catabolism by CHAC1 supports the mitochondrial cysteine pool to sustain the function of the Fe-S proteins critical to oxidative metabolism. We find that disrupting Fe-S cluster synthesis under cysteine restriction protects against the induction of ferroptosis, suggesting that the preservation of mitochondrial function is antagonistic to survival under starved conditions. Overall, our findings implicate mitochondrial cysteine metabolism in the induction of ferroptosis and reveal a mechanism of mitochondrial resilience in response to nutrient stress.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Cisteína , Ferroptosis , Glutatión , Neoplasias Pulmonares , Mitocondrias , Humanos , Cisteína/metabolismo , Mitocondrias/metabolismo , Glutatión/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Línea Celular Tumoral , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas Hierro-Azufre/metabolismo , Oxidación-Reducción , Ratones
6.
Nat Commun ; 15(1): 3269, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38627381

RESUMEN

Maturation of iron-sulfur proteins in eukaryotes is initiated in mitochondria by the core iron-sulfur cluster assembly (ISC) complex, consisting of the cysteine desulfurase sub-complex NFS1-ISD11-ACP1, the scaffold protein ISCU2, the electron donor ferredoxin FDX2, and frataxin, a protein dysfunctional in Friedreich's ataxia. The core ISC complex synthesizes [2Fe-2S] clusters de novo from Fe and a persulfide (SSH) bound at conserved cluster assembly site residues. Here, we elucidate the poorly understood Fe-dependent mechanism of persulfide transfer from cysteine desulfurase NFS1 to ISCU2. High-resolution cryo-EM structures obtained from anaerobically prepared samples provide snapshots that both visualize different stages of persulfide transfer from Cys381NFS1 to Cys138ISCU2 and clarify the molecular role of frataxin in optimally positioning assembly site residues for fast sulfur transfer. Biochemical analyses assign ISCU2 residues essential for sulfur transfer, and reveal that Cys138ISCU2 rapidly receives the persulfide without a detectable intermediate. Mössbauer spectroscopy assessing the Fe coordination of various sulfur transfer intermediates shows a dynamic equilibrium between pre- and post-sulfur-transfer states shifted by frataxin. Collectively, our study defines crucial mechanistic stages of physiological [2Fe-2S] cluster assembly and clarifies frataxin's molecular role in this fundamental process.


Asunto(s)
Frataxina , Proteínas Hierro-Azufre , Proteínas Hierro-Azufre/metabolismo , Sulfuros/metabolismo , Azufre/metabolismo , Liasas de Carbono-Azufre/metabolismo , Proteínas de Unión a Hierro/metabolismo
7.
Adv Biol (Weinh) ; 8(5): e2300545, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38574244

RESUMEN

HapX and SreA are transcription factors that regulate the response of the fungus Aspergillus fumigatus to the availability of iron. During iron starvation, HapX represses genes involved in iron consuming pathways and upon a shift to iron excess, HapX activates these same genes. SreA blocks the expression of genes needed for iron uptake during periods of iron availability. Both proteins possess cysteine-rich regions (CRR) that are hypothesized to be necessary for the sensing of iron levels. However, the contribution of each of these domains to the function of the protein has remained unclear. Here, the ability of peptide analogs of each CRR is determined to bind an iron-sulfur cluster in vitro. UV-vis and resonance Raman (RR) spectroscopies reveal that each CRR is capable of coordinating a [2Fe-2S] cluster with comparable affinities. The iron-sulfur cluster coordinated to the CRR-B domain of HapX displays particularly high stability. The data are consistent with HapX and SreA mediating responses to cellular iron levels through the direct coordination of [2Fe-2S] clusters. The high stability of the CRR-B peptide may also find use as a starting point for the development of new green catalysts.


Asunto(s)
Cisteína , Proteínas Fúngicas , Proteínas Hierro-Azufre , Péptidos , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Cisteína/metabolismo , Cisteína/química , Péptidos/metabolismo , Péptidos/química , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Aspergillus fumigatus/metabolismo , Aspergillus fumigatus/química , Aspergillus fumigatus/genética , Hierro/metabolismo , Unión Proteica , Espectrometría Raman , Factores de Transcripción/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética
8.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119723, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38599324

RESUMEN

Viruses have evolved complex mechanisms to exploit host factors for replication and assembly. In response, host cells have developed strategies to block viruses, engaging in a continuous co-evolutionary battle. This dynamic interaction often revolves around the competition for essential resources necessary for both host cell and virus replication. Notably, iron, required for the biosynthesis of several cofactors, including iron­sulfur (FeS) clusters, represents a critical element in the ongoing competition for resources between infectious agents and host. Although several recent studies have identified FeS cofactors at the core of virus replication machineries, our understanding of their specific roles and the cellular processes responsible for their incorporation into viral proteins remains limited. This review aims to consolidate our current knowledge of viral components that have been characterized as FeS proteins and elucidate how viruses harness these versatile cofactors to their benefit. Its objective is also to propose that viruses may depend on incorporation of FeS cofactors more extensively than is currently known. This has the potential to revolutionize our understanding of viral replication, thereby carrying significant implications for the development of strategies to target infections.


Asunto(s)
Proteínas Hierro-Azufre , Proteínas Virales , Replicación Viral , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/genética , Humanos , Proteínas Virales/metabolismo , Proteínas Virales/genética , Virus/metabolismo , Virus/genética , Virosis/metabolismo , Virosis/virología , Hierro/metabolismo , Animales , Interacciones Huésped-Patógeno
9.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119732, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38631440

RESUMEN

Iron-sulfur clusters serve as indispensable cofactors within proteins across all three domains of life. Fe/S clusters emerged early during the evolution of life on our planet and the biogeochemical cycle of sulfur is one of the most ancient and important element cycles. It is therefore no surprise that Fe/S proteins have crucial roles in the multiple steps of microbial sulfur metabolism. During dissimilatory sulfur oxidation in prokaryotes, Fe/S proteins not only serve as electron carriers in several steps, but also perform catalytic roles, including unprecedented reactions. Two cytoplasmic enzyme systems that oxidize sulfane sulfur to sulfite are of particular interest in this context: The rDsr pathway employs the reverse acting dissimilatory sulfite reductase rDsrAB as its key enzyme, while the sHdr pathway utilizes polypeptides resembling the HdrA, HdrB and HdrC subunits of heterodisulfide reductase from methanogenic archaea. Both pathways involve components predicted to bind unusual noncubane Fe/S clusters acting as catalysts for the formation of disulfide or sulfite. Mapping of Fe/S cluster machineries on the sulfur-oxidizing prokaryote tree reveals that ISC, SUF, MIS and SMS are all sufficient to meet the Fe/S cluster maturation requirements for operation of the sHdr or rDsr pathways. The sHdr pathway is dependent on lipoate-binding proteins that are assembled by a novel pathway, involving two Radical SAM proteins, namely LipS1 and LipS2. These proteins coordinate sulfur-donating auxiliary Fe/S clusters in atypical patterns by three cysteines and one histidine and act as lipoyl synthases by jointly inserting two sulfur atoms to an octanoyl residue. This article is part of a Special Issue entitled: Biogenesis and Function of Fe/S proteins.


Asunto(s)
Proteínas Hierro-Azufre , Oxidación-Reducción , Azufre , Azufre/metabolismo , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Archaea/metabolismo , Oxidorreductasas/metabolismo , Oxidorreductasas/genética
10.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119730, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38631441

RESUMEN

[FeS] clusters are co-factors that are essential for life and are synthesized by dedicated multiprotein cellular machineries. In this review, we present the current scenario for the emergence and the diversification of the [FeS] cluster biosynthesis machineries. In addition to well-known NIF, ISC and SUF machineries, two alternative minimal systems, SMS, and MIS, were recently identified. Taxonomic distribution and phylogeny analyses indicate that SMS and MIS were present in the Last Universal Common Ancestor (LUCA), well before the increase of oxygen on Earth. ISC, SUF and NIF systems emerged later in the history of life. The possible reasons for the emergence and diversification of these machineries are discussed.


Asunto(s)
Proteínas Hierro-Azufre , Hierro , Azufre , Azufre/metabolismo , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/genética , Hierro/metabolismo , Filogenia , Oxígeno/metabolismo , Evolución Molecular
11.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119731, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38631442

RESUMEN

Molybdenum cofactor (Moco) biosynthesis is a complex process that involves the coordinated function of several proteins. In the recent years it has become evident that the availability of Fe-S clusters play an important role for the biosynthesis of Moco. First, the MoaA protein binds two [4Fe-4S] clusters per monomer. Second, the expression of the moaABCDE and moeAB operons is regulated by FNR, which senses the availability of oxygen via a functional [4Fe-4S] cluster. Finally, the conversion of cyclic pyranopterin monophosphate to molybdopterin requires the availability of the L-cysteine desulfurase IscS, which is an enzyme involved in the transfer of sulfur to various acceptor proteins with a main role in the assembly of Fe-S clusters. In this review, we dissect the dependence of the production of active molybdoenzymes in detail, starting from the regulation of gene expression and further explaining sulfur delivery and Fe-S cluster insertion into target enzymes. Further, Fe-S cluster assembly is also linked to iron availability. While the abundance of selected molybdoenzymes is largely decreased under iron-limiting conditions, we explain that the expression of the genes is dependent on an active FNR protein. FNR is a very important transcription factor that represents the master-switch for the expression of target genes in response to anaerobiosis. Moco biosynthesis is further directly dependent on the presence of ArcA and also on an active Fur protein.


Asunto(s)
Coenzimas , Proteínas Hierro-Azufre , Metaloproteínas , Cofactores de Molibdeno , Pteridinas , Metaloproteínas/metabolismo , Metaloproteínas/genética , Metaloproteínas/biosíntesis , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/genética , Coenzimas/metabolismo , Coenzimas/biosíntesis , Coenzimas/genética , Pteridinas/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Hierro/metabolismo , Azufre/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Liasas de Carbono-Azufre/metabolismo , Liasas de Carbono-Azufre/genética , Regulación Bacteriana de la Expresión Génica , Operón , Isomerasas
12.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119733, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38641180

RESUMEN

Iron­sulfur (FeS) clusters are cofactors of numerous proteins involved in essential cellular functions including respiration, protein translation, DNA synthesis and repair, ribosome maturation, anti-viral responses, and isopropylmalate isomerase activity. Novel FeS proteins are still being discovered due to the widespread use of cryogenic electron microscopy (cryo-EM) and elegant genetic screens targeted at protein discovery. A complex sequence of biochemical reactions mediated by a conserved machinery controls biosynthesis of FeS clusters. In eukaryotes, a remarkable epistasis has been observed: the mitochondrial machinery, termed ISC (Iron-Sulfur Cluster), lies upstream of the cytoplasmic machinery, termed CIA (Cytoplasmic Iron­sulfur protein Assembly). The basis for this arrangement is the production of a hitherto uncharacterized intermediate, termed X-S or (Fe-S)int, produced in mitochondria by the ISC machinery, exported by the mitochondrial ABC transporter Atm1 (ABCB7 in humans), and then utilized by the CIA machinery for the cytoplasmic/nuclear FeS cluster assembly. Genetic and biochemical findings supporting this sequence of events are herein presented. New structural views of the Atm1 transport phases are reviewed. The key compartmental roles of glutathione in cellular FeS cluster biogenesis are highlighted. Finally, data are presented showing that every one of the ten core components of the mitochondrial ISC machinery and Atm1, when mutated or depleted, displays similar phenotypes: mitochondrial and cytoplasmic FeS clusters are both rendered deficient, consistent with the epistasis noted above.


Asunto(s)
Citoplasma , Proteínas Hierro-Azufre , Mitocondrias , Mitocondrias/metabolismo , Mitocondrias/genética , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/genética , Humanos , Citoplasma/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Glutatión/metabolismo
13.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119718, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38574823

RESUMEN

Reactions catalysed by iron-sulfur (Fe-S) enzymes appear in a variety of biosynthetic pathways that produce valuable natural products. Harnessing these biosynthetic pathways by expression in microbial cell factories grown on an industrial scale would yield enormous economic and environmental benefits. However, Fe-S enzymes often become bottlenecks that limits the productivity of engineered pathways. As a consequence, achieving the production metrics required for industrial application remains a distant goal for Fe-S enzyme-dependent pathways. Here, we identify and review three core challenges in harnessing Fe-S enzyme activity, which all stem from the properties of Fe-S clusters: 1) limited Fe-S cluster supply within the host cell, 2) Fe-S cluster instability, and 3) lack of specialized reducing cofactor proteins often required for Fe-S enzyme activity, such as enzyme-specific flavodoxins and ferredoxins. We highlight successful methods developed for a variety of Fe-S enzymes and electron carriers for overcoming these difficulties. We use heterologous nitrogenase expression as a grand case study demonstrating how each of these challenges can be addressed. We predict that recent breakthroughs in protein structure prediction and design will prove well-suited to addressing each of these challenges. A reliable toolkit for harnessing Fe-S enzymes in engineered metabolic pathways will accelerate the development of industry-ready Fe-S enzyme-dependent biosynthesis pathways.


Asunto(s)
Proteínas Hierro-Azufre , Biología Sintética , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/genética , Biología Sintética/métodos , Vías Biosintéticas , Nitrogenasa/metabolismo , Nitrogenasa/genética , Azufre/metabolismo , Ferredoxinas/metabolismo , Ferredoxinas/genética
14.
J Biol Chem ; 300(4): 107142, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38452854

RESUMEN

It was generally postulated that when intracellular free iron content is elevated in bacteria, the ferric uptake regulator (Fur) binds its corepressor a mononuclear ferrous iron to regulate intracellular iron homeostasis. However, the proposed iron-bound Fur had not been identified in any bacteria. In previous studies, we have demonstrated that Escherichia coli Fur binds a [2Fe-2S] cluster in response to elevation of intracellular free iron content and that binding of the [2Fe-2S] cluster turns on Fur as an active repressor to bind a specific DNA sequence known as the Fur-box. Here we find that the iron-sulfur cluster assembly scaffold protein IscU is required for the [2Fe-2S] cluster assembly in Fur, as deletion of IscU inhibits the [2Fe-2S] cluster assembly in Fur and prevents activation of Fur as a repressor in E. coli cells in response to elevation of intracellular free iron content. Additional studies reveal that IscU promotes the [2Fe-2S] cluster assembly in apo-form Fur and restores its Fur-box binding activity in vitro. While IscU is also required for the [2Fe-2S] cluster assembly in the Haemophilus influenzae Fur in E. coli cells, deletion of IscU does not significantly affect the [2Fe-2S] cluster assembly in the E. coli ferredoxin and siderophore-reductase FhuF. Our results suggest that IscU may have a unique role for the [2Fe-2S] cluster assembly in Fur and that regulation of intracellular iron homeostasis is closely coupled with iron-sulfur cluster biogenesis in E. coli.


Asunto(s)
Proteínas Bacterianas , Proteínas de Escherichia coli , Escherichia coli , Proteínas Hierro-Azufre , Hierro , Proteínas Represoras , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Hierro/metabolismo
15.
J Biol Chem ; 300(3): 105745, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38354784

RESUMEN

The NEET proteins, an important family of iron-sulfur (Fe-S) proteins, have generated a strong interest due to their involvement in diverse diseases such as cancer, diabetes, and neurodegenerative disorders. Among the human NEET proteins, CISD3 has been the least studied, and its functional role is still largely unknown. We have investigated the biochemical features of CISD3 at the atomic and in cellulo levels upon challenge with different stress conditions i.e., iron deficiency, exposure to hydrogen peroxide, and nitric oxide. The redox and cellular stability properties of the protein agree on a predominance of reduced form of CISD3 in the cells. Upon the addition of iron chelators, CISD3 loses its Fe-S clusters and becomes unstructured, and its cellular level drastically decreases. Chemical shift perturbation measurements suggest that, upon cluster oxidation, the protein undergoes a conformational change at the C-terminal CDGSH domain, which determines the instability of the oxidized state. This redox-associated conformational change may be the source of cooperative electron transfer via the two [Fe2S2] clusters in CISD3, which displays a single sharp voltammetric signal at -31 mV versus SHE. Oxidized CISD3 is particularly sensitive to the presence of hydrogen peroxide in vitro, whereas only the reduced form is able to bind nitric oxide. Paramagnetic NMR provides clear evidence that, upon NO binding, the cluster is disassembled but iron ions are still bound to the protein. Accordingly, in cellulo CISD3 is unaffected by oxidative stress induced by hydrogen peroxide but it becomes highly unstable in response to nitric oxide treatment.


Asunto(s)
Proteínas Hierro-Azufre , Proteínas Mitocondriales , Óxido Nítrico , Humanos , Peróxido de Hidrógeno/metabolismo , Hierro/metabolismo , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/metabolismo , Óxido Nítrico/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Células HEK293 , Estabilidad Proteica
16.
J Am Chem Soc ; 146(6): 3710-3720, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38308759

RESUMEN

1/2H and 13C hyperfine coupling constants to 5'-deoxyadenosyl (5'-dAdo•) radical trapped within the active site of the radical S-adenosyl-l-methionine (SAM) enzyme, pyruvate formate lyase-activating enzyme (PFL-AE), both in the absence of substrate and the presence of a reactive peptide-model of the PFL substrate, are completely characteristic of a classical organic free radical whose unpaired electron is localized in the 2pπ orbital of the sp2 C5'-carbon (J. Am. Chem. Soc. 2019, 141, 12139-12146). However, prior electron-nuclear double resonance (ENDOR) measurements had indicated that this 5'-dAdo• free radical is never truly "free": tight van der Waals contact with its target partners and active-site residues guide it in carrying out the exquisitely precise, regioselective reactions that are hallmarks of RS enzymes. Here, our understanding of how the active site chaperones 5'-dAdo• is extended through the finding that this apparently unexceptional organic free radical has an anomalous g-tensor and exhibits significant 57Fe, 13C, 15N, and 2H hyperfine couplings to the adjacent, isotopically labeled, methionine-bound [4Fe-4S]2+ cluster cogenerated with 5'-dAdo• during homolytic cleavage of cluster-bound SAM. The origin of the 57Fe couplings through nonbonded radical-cluster contact is illuminated by a formal exchange-coupling model and broken symmetry-density functional theory computations. Incorporation of ENDOR-derived distances from C5'(dAdo•) to labeled-methionine as structural constraints yields a model for active-site positioning of 5'-dAdo• with a short, nonbonded C5'-Fe distance (∼3 Å). This distance involves substantial motion of 5'-dAdo• toward the unique Fe of the [4Fe-4S]2+ cluster upon S-C(5') bond-cleavage, plausibly an initial step toward formation of the Fe-C5' bond of the organometallic complex, Ω, the central intermediate in catalysis by radical-SAM enzymes.


Asunto(s)
Proteínas Hierro-Azufre , S-Adenosilmetionina , S-Adenosilmetionina/metabolismo , Metionina , Espectroscopía de Resonancia por Spin del Electrón/métodos , Dominio Catalítico , Racemetionina , Radicales Libres/química , Proteínas Hierro-Azufre/química
17.
Sci Rep ; 14(1): 3026, 2024 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-38321125

RESUMEN

[NiFe]-hydrogenases have a bimetallic NiFe(CN)2CO cofactor in their large, catalytic subunit. The 136 Da Fe(CN)2CO group of this cofactor is preassembled on a distinct HypC-HypD scaffold complex, but the intracellular source of the iron ion is unresolved. Native mass spectrometric analysis of HypCD complexes defined the [4Fe-4S] cluster associated with HypD and identified + 26 to 28 Da and + 136 Da modifications specifically associated with HypC. A HypCC2A variant without the essential conserved N-terminal cysteine residue dissociated from its complex with native HypD lacked all modifications. Native HypC dissociated from HypCD complexes isolated from Escherichia coli strains deleted for the iscS or iscU genes, encoding core components of the Isc iron-sulfur cluster biogenesis machinery, specifically lacked the + 136 Da modification, but this was retained on HypC from suf mutants. The presence or absence of the + 136 Da modification on the HypCD complex correlated with the hydrogenase enzyme activity profiles of the respective mutant strains. Notably, the [4Fe-4S] cluster on HypD was identified in all HypCD complexes analyzed. These results suggest that the iron of the Fe(CN)2CO group on HypCD derives from the Isc machinery, while either the Isc or the Suf machinery can deliver the [4Fe-4S] cluster to HypD.


Asunto(s)
Proteínas de Escherichia coli , Hidrogenasas , Proteínas Hierro-Azufre , Escherichia coli/genética , Hierro/metabolismo , Proteínas de Escherichia coli/metabolismo , Hidrogenasas/metabolismo , Dominio Catalítico , Cisteína/química
18.
Elife ; 132024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38251655

RESUMEN

Mammalian ferredoxin 1 and 2 (FDX1/2) belong to an evolutionary conserved family of iron-sulfur cluster containing proteins and act as electron shutters between ferredoxin reductase (FDXR) and numerous proteins involved in critical biological pathways. FDX1 is involved in biogenesis of steroids and bile acids, Vitamin A/D metabolism, and lipoylation of tricarboxylic acid (TCA) cycle enzymes. FDX1 has been extensively characterized biochemically but its role in physiology and lipid metabolism has not been explored. In this study, we generated Fdx1-deficient mice and showed that knockout of both alleles of the Fdx1 gene led to embryonic lethality. We also showed that like Fdxr+/-+/-, Fdx1+/-+/- had a shorter life span and were prone to steatohepatitis. However, unlike Fdxr+/-+/-, Fdx1+/-+/- were not prone to spontaneous tumors. Additionally, we showed that FDX1 deficiency led to lipid droplet accumulation possibly via the ABCA1-SREBP1/2 pathway. Specifically, untargeted lipidomic analysis showed that FDX1 deficiency led to alterations in several classes of lipids, including cholesterol, triacylglycerides, acylcarnitines, ceramides, phospholipids and lysophospholipids. Taken together, our data indicate that FDX1 is essential for mammalian embryonic development and lipid homeostasis at both cellular and organismal levels.


Asunto(s)
Desarrollo Embrionario , Ferredoxinas , Animales , Ratones , Ferredoxinas/genética , Ferredoxinas/metabolismo , Homeostasis , Proteínas Hierro-Azufre/química , Lípidos , Mamíferos/metabolismo
19.
J Inorg Biochem ; 252: 112457, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38176366

RESUMEN

NsrR from Streptomyces coelicolor is a bacterial nitric oxide (NO) sensor/nitrosative stress regulator as its primary function, and has been shown to have differential response at low, mid, and high levels of NO. These must correspond to discrete structural changes at the protein-bound [4Fe-4S] cluster in response to stepwise nitrosylation of the cluster. We have investigated the effect of the monohapto carboxylate ligand in the site differentiated [4Fe-4S] cluster cofactor of the protein NsrR on modulating its reactivity to NO with a focus on indentifying mechanistic intermediates. We have prepared a synthetic model [4Fe-4S] cluster complex with tripodal ligand and one single site differentiated site occupied by either thiolate or carboxylate ligand. We report here the mechanistic details of sequential steps of nitrosylation as observed by ESI MS and IR spectroscopy. Parallel non-denaturing mass spectrometry analyses were performed using site-differentiated variants of NsrR with the native aspartic acid, cysteine, or alanine in the position of the forth ligand to the cluster. A mono-nitrosylated synthetic [4Fe-4S] cluster was observed for the first time in a biologically-relevant thiolate-based coordination environment. Combined synthetic and protein data give unprecedented clarity in the modulation of nitrosylation of a [4Fe-4S] cluster.


Asunto(s)
Proteínas Hierro-Azufre , Streptomyces coelicolor , Proteínas Hierro-Azufre/química , Óxido Nítrico/metabolismo , Proteínas Bacterianas/química , Ligandos , Espectroscopía de Resonancia por Spin del Electrón
20.
J Neurol Sci ; 456: 122808, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38043332

RESUMEN

There are scarce reports of riboflavin-responsive lipid storage myopathy in elderly patients with onset in their sixties. We describe three elderly patients with riboflavin-responsive lipid-storage myopathy. All three patients (aged 67-71 years on first examination) had subacute onset of neck extensors and proximal limb weakness progressing to inability to rise from a sitting position or to walk. Muscle biopsies showed vacuoles with lipid content, mainly in type 1 fibers. Genetic analysis failed to identify any pathogenic variant in one patient, identified a heterozygous variant of uncertain significance c.812 A > G; p.Tyr271Cys in the ETFDH gene in the second patient, and revealed a heterozygote likely pathogenic variant c.1286-2 A > C in the ETFDH gene predicted to cause abnormal splicing in the third patient. All patients responded to treatment with riboflavin and carnitine, and regained normal strength. This report emphasizes the importance of muscle biopsy in revealing treatable lipid storage myopathy in elderly patients with progressive myopathy of unidentifiable cause.


Asunto(s)
Proteínas Hierro-Azufre , Errores Innatos del Metabolismo Lipídico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa , Enfermedades Musculares , Distrofias Musculares , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH , Humanos , Anciano , Músculo Esquelético/metabolismo , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Flavoproteínas Transportadoras de Electrones/genética , Proteínas Hierro-Azufre/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Enfermedades Musculares/tratamiento farmacológico , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Riboflavina/uso terapéutico , Lípidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA