Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
J Am Chem Soc ; 144(13): 5713-5717, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35343688

RESUMEN

Human lipoyl synthase (LIAS) is an enzyme containing two [4Fe-4S] clusters (named FeSRS and FeSaux) involved in the biosynthesis of the lipoyl cofactor. The mechanism by which a [4Fe-4S] cluster is inserted into LIAS has thus far remained elusive. Here we show that NFU1 and ISCA1 of the mitochondrial iron-sulfur cluster assembly machinery, via forming a heterodimeric complex, are the key factors for the insertion of a [4Fe-4S] cluster into the FeSRS site of LIAS. In this process, the crucial actor is the C-domain of NFU1, which, by exploiting a protein-interaction affinity gradient increasing from ISCA1 to LIAS, drives the cluster to its final destination.


Asunto(s)
Proteínas Hierro-Azufre , Humanos , Hierro/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Azufre/metabolismo
2.
Nature ; 536(7615): 205-9, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27487209

RESUMEN

Genetic differences that specify unique aspects of human evolution have typically been identified by comparative analyses between the genomes of humans and closely related primates, including more recently the genomes of archaic hominins. Not all regions of the genome, however, are equally amenable to such study. Recurrent copy number variation (CNV) at chromosome 16p11.2 accounts for approximately 1% of cases of autism and is mediated by a complex set of segmental duplications, many of which arose recently during human evolution. Here we reconstruct the evolutionary history of the locus and identify bolA family member 2 (BOLA2) as a gene duplicated exclusively in Homo sapiens. We estimate that a 95-kilobase-pair segment containing BOLA2 duplicated across the critical region approximately 282 thousand years ago (ka), one of the latest among a series of genomic changes that dramatically restructured the locus during hominid evolution. All humans examined carried one or more copies of the duplication, which nearly fixed early in the human lineage--a pattern unlikely to have arisen so rapidly in the absence of selection (P < 0.0097). We show that the duplication of BOLA2 led to a novel, human-specific in-frame fusion transcript and that BOLA2 copy number correlates with both RNA expression (r = 0.36) and protein level (r = 0.65), with the greatest expression difference between human and chimpanzee in experimentally derived stem cells. Analyses of 152 patients carrying a chromosome 16p11. rearrangement show that more than 96% of breakpoints occur within the H. sapiens-specific duplication. In summary, the duplicative transposition of BOLA2 at the root of the H. sapiens lineage about 282 ka simultaneously increased copy number of a gene associated with iron homeostasis and predisposed our species to recurrent rearrangements associated with disease.


Asunto(s)
Cromosomas Humanos Par 16/genética , Variaciones en el Número de Copia de ADN/genética , Evolución Molecular , Predisposición Genética a la Enfermedad , Proteínas/genética , Animales , Trastorno Autístico/genética , Rotura Cromosómica , Duplicación de Gen , Homeostasis/genética , Humanos , Hierro/metabolismo , Pan troglodytes/genética , Pongo/genética , Proteínas/análisis , Recombinación Genética , Especificidad de la Especie , Factores de Tiempo
3.
Int J Mol Sci ; 22(9)2021 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-34063696

RESUMEN

Multiple mitochondrial dysfunctions syndrome (MMDS) is a rare neurodegenerative disorder associated with mutations in genes with a vital role in the biogenesis of mitochondrial [4Fe-4S] proteins. Mutations in one of these genes encoding for BOLA3 protein lead to MMDS type 2 (MMDS2). Recently, a novel phenotype for MMDS2 with complete clinical recovery was observed in a patient containing a novel variant (c.176G > A, p.Cys59Tyr) in compound heterozygosity. In this work, we aimed to rationalize this unique phenotype observed in MMDS2. To do so, we first investigated the structural impact of the Cys59Tyr mutation on BOLA3 by NMR, and then we analyzed how the mutation affects both the formation of a hetero-complex between BOLA3 and its protein partner GLRX5 and the iron-sulfur cluster-binding properties of the hetero-complex by various spectroscopic techniques and by experimentally driven molecular docking. We show that (1) the mutation structurally perturbed the iron-sulfur cluster-binding region of BOLA3, but without abolishing [2Fe-2S]2+ cluster-binding on the hetero-complex; (2) tyrosine 59 did not replace cysteine 59 as iron-sulfur cluster ligand; and (3) the mutation promoted the formation of an aberrant apo C59Y BOLA3-GLRX5 complex. All these aspects allowed us to rationalize the unique phenotype observed in MMDS2 caused by Cys59Tyr mutation.


Asunto(s)
Glutarredoxinas/genética , Mitocondrias/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Cisteína/genética , Glutarredoxinas/ultraestructura , Humanos , Proteínas Hierro-Azufre/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Simulación del Acoplamiento Molecular , Complejos Multiproteicos , Mutación , Resonancia Magnética Nuclear Biomolecular , Fenotipo
4.
Angew Chem Int Ed Engl ; 60(27): 14841-14845, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-33852169

RESUMEN

Human anamorsin is an iron-sulfur (Fe-S)-cluster-binding protein acting as an electron donor in the early steps of cytosolic iron-sulfur protein biogenesis. Human anamorsin belongs to the eukaryotic CIAPIN1 protein family and contains two highly conserved cysteine-rich motifs, each binding an Fe-S cluster. In vitro works by various groups have provided rather controversial results for the type of Fe-S clusters bound to the CIAPIN1 proteins. In order to unravel the knot on this topic, we used an in cellulo approach combining Mössbauer and EPR spectroscopies to characterize the iron-sulfur-cluster-bound form of human anamorsin. We found that the protein binds two [2Fe-2S] clusters at both its cysteine-rich motifs.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/química , Proteínas Hierro-Azufre/química , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Unión Proteica , Espectroscopía de Mossbauer
5.
J Am Chem Soc ; 142(24): 10794-10805, 2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32429669

RESUMEN

Human cytosolic monothiol glutaredoxin-3 (GLRX3) is a protein essential for the maturation of cytosolic [4Fe-4S] proteins. We show here that dimeric cluster-bridged GLRX3 transfers its [2Fe-2S]2+ clusters to the human P-loop NTPase NUBP1, an essential early component of the cytosolic iron-sulfur assembly (CIA) machinery. Specifically, we observed that [2Fe-2S]2+ clusters are transferred from GLRX3 to monomeric apo NUBP1 and reductively coupled to form [4Fe-4S]2+ clusters on both N-terminal CX13CX2CX5C and C-terminal CPXC motifs of NUBP1 in the presence of glutathione that acts as a reductant. In this process, cluster binding to the C-terminal motif of NUBP1 promotes protein dimerization, while cluster binding to the N-terminal motif does not affect the quaternary structure of NUBP1. The cluster transfer/assembly process is not complete on both N- and C-terminal motifs and indeed requires a reductant stronger than GSH to increase its efficiency. We also showed that the [4Fe-4S]2+ cluster formed at the N-terminal motif of NUBP1 is tightly bound, while the [4Fe-4S]2+ cluster bound at the C-terminal motif is labile. Our findings provide the first evidence for GLRX3 acting as a [2Fe-2S] cluster chaperone in the early stage of the CIA machinery.


Asunto(s)
Proteínas Portadoras/metabolismo , Citosol/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Hierro-Azufre/metabolismo , Hierro/metabolismo , Chaperonas Moleculares/metabolismo , Azufre/metabolismo , Proteínas Portadoras/química , Citosol/química , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Hierro/química , Proteínas Hierro-Azufre/química , Chaperonas Moleculares/química , Azufre/química
6.
J Am Chem Soc ; 140(43): 14401-14412, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30269484

RESUMEN

The maturation of mitochondrial iron-sulfur proteins requires a complex protein machinery. Human IBA57 protein was proposed to act in a late phase of this machinery, along with GLRX5, ISCA1, and ISCA2. However, a molecular picture on how these proteins cooperate is not defined yet. We show here that IBA57 forms a heterodimeric complex with ISCA2 by bridging a [2Fe-2S] cluster, that [2Fe-2S] cluster binding is absolutely required to promote the complex formation, and that the cysteine of the conserved motif characterizing IBA57 protein family and the three conserved cysteines of the ISCA protein family act as cluster ligands. The [2Fe-2S] heterodimeric complex is the final product when IBA57 is either exposed to [2Fe-2S] ISCA2 or in the presence of [2Fe-2S] GLRX5 and apo ISCA2. We also find that the [2Fe-2S] ISCA2-IBA57 complex is resistant to highly oxidative environments and is capable of reactivating apo aconitase in vitro. Collectively, our data delinate a [2Fe-2S] cluster transfer pathway involving three partner proteins of the mitochondrial ISC machinery, that is, GLRX5, ISCA2 and IBA57, which leads to the formation of a [2Fe-2S] ISCA2-IBA57 complex.

7.
J Biol Inorg Chem ; 23(4): 665-685, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29569085

RESUMEN

Iron-sulfur proteins were among the first class of metalloproteins that were actively studied using NMR spectroscopy tailored to paramagnetic systems. The hyperfine shifts, their temperature dependencies and the relaxation rates of nuclei of cluster-bound residues are an efficient fingerprint of the nature and the oxidation state of the Fe-S cluster. NMR significantly contributed to the analysis of the magnetic coupling patterns and to the understanding of the electronic structure occurring in [2Fe-2S], [3Fe-4S] and [4Fe-4S] clusters bound to proteins. After the first NMR structure of a paramagnetic protein was obtained for the reduced E. halophila HiPIP I, many NMR structures were determined for several Fe-S proteins in different oxidation states. It was found that differences in chemical shifts, in patterns of unobserved residues, in internal mobility and in thermodynamic stability are suitable data to map subtle changes between the two different oxidation states of the protein. Recently, the interaction networks responsible for maturing human mitochondrial and cytosolic Fe-S proteins have been largely characterized by combining solution NMR standard experiments with those tailored to paramagnetic systems. We show here the contribution of solution NMR in providing a detailed molecular view of "Fe-S interactomics". This contribution was particularly effective when protein-protein interactions are weak and transient, and thus difficult to be characterized at high resolution with other methodologies.


Asunto(s)
Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Mapas de Interacción de Proteínas , Electrones , Humanos
8.
J Biol Inorg Chem ; 23(4): 687, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29855713

RESUMEN

The article "The NMR contribution to protein-protein networking in Fe-S protein maturation", written by Lucia Banci, Francesca Camponeschi, Simone Ciofi­Baffoni, Mario Piccioli was originally published electronically on the publisher's internet portal (currently SpringerLink) on 22 March, 2018 without open access.

9.
J Am Chem Soc ; 139(28): 9479-9482, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28648056

RESUMEN

Human mitoNEET is a homodimeric protein anchored to the outer mitochondrial membrane and has a C-terminal [2Fe-2S] binding domain located in the cytosol. Recently, human mitoNEET has been shown to be implicated in Fe/S cluster repair of cytosolic iron regulatory protein 1 (IRP1), a key regulator of cellular iron homeostasis in mammalian cells. The Fe/S cluster repair function of mitoNEET is based on an Fe/S redox switch mechanism: under normal cellular conditions, reduced [2Fe-2S]+-mitoNEET is present and is inactive as an Fe/S cluster transfer protein; under conditions of oxidative cellular stress, the clusters of mitoNEET become oxidized, and the formed [2Fe-2S]2+-mitoNEET species reacts promptly to initiate Fe/S cluster transfer to IRP1, recycling the cytosolic apo-IRP1 into holo-aconitase. Until now, no clear data have been available on which is the system that reduces the mitoNEET clusters back once oxidative stress is not present anymore. In the present work, we used UV-vis and NMR spectroscopies to investigate the electron transfer process between mitoNEET and the cytosolic electron-donor Ndor1/anamorsin complex, a component of the cytosolic iron-sulfur protein assembly (CIA) machinery. The [2Fe-2S] clusters of mitoNEET are reduced via the formation of a transient complex that brings the [2Fe-2S] clusters of mitoNEET close to the redox-active [2Fe-2S] cluster of anamorsin. Our data provide in vitro evidence of a possible direct link between the CIA machinery and the mitoNEET cluster transfer repair pathway. This link might contribute to recovery of CIA machinery efficiency to mature cytosolic and nuclear Fe/S proteins.


Asunto(s)
Flavoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Hierro-Azufre/metabolismo , Proteínas Mitocondriales/metabolismo , Oxidorreductasas/metabolismo , Flavoproteínas/química , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas Hierro-Azufre/química , Proteínas Mitocondriales/química , Oxidorreductasas/química , Unión Proteica
10.
J Am Chem Soc ; 139(2): 719-730, 2017 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-27989128

RESUMEN

The cellular toxicity of copper is usually associated with its ability to generate reactive oxygen species. However, recent studies in bacterial organisms showed that copper toxicity is also strictly connected to iron-sulfur cluster proteins and to their assembly processes. Mitochondria of eukaryotic cells contain a labile copper(I) pool localized in the matrix where also the mitochondrial iron-sulfur (Fe/S) cluster assembly machinery resides to mature mitochondrial Fe/S cluster-containing proteins. Misregulation of copper homeostasis might therefore damage mitochondrial Fe/S protein maturation. To describe, from a molecular perspective, the effects of copper(I) toxicity on such a maturation process, we have here investigated the still unknown mechanism of [4Fe-4S] cluster formation conducted by the mitochondrial ISCA1/ISCA2 and GLRX5 proteins, and defined how copper(I) can impair this process. The molecular model here proposed indicates that the copper(I) and Fe/S protein maturation cellular pathways need to be strictly regulated to avoid copper(I) ion from blocking mitochondrial [4Fe-4S] protein maturation.


Asunto(s)
Cobre/toxicidad , Proteínas Hierro-Azufre , Hierro , Mitocondrias , Modelos Biológicos , Azufre , Cobre/química , Hierro/química , Proteínas Hierro-Azufre/química , Azufre/química
11.
Nat Chem Biol ; 11(10): 772-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26302480

RESUMEN

In eukaryotes, cytosolic monothiol glutaredoxins are proteins implicated in intracellular iron trafficking and sensing via their bound [2Fe-2S] clusters. We define a new role of human cytosolic monothiol glutaredoxin-3 (GRX3) in transferring its [2Fe-2S] clusters to human anamorsin, a physical and functional protein partner of GRX3 in the cytosol, whose [2Fe-2S] cluster-bound form is involved in the biogenesis of cytosolic and nuclear Fe-S proteins. Specific protein recognition between the N-terminal domains of the two proteins is the mandatory requisite to promote the [2Fe-2S] cluster transfer from GRX3 to anamorsin.


Asunto(s)
Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Hierro-Azufre/metabolismo , Hierro/metabolismo , Apoproteínas/química , Apoproteínas/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/genética , Citosol/metabolismo , Escherichia coli/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Modelos Biológicos , Simulación del Acoplamiento Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas
12.
Biochim Biophys Acta Gen Subj ; 1861(8): 2119-2131, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28483642

RESUMEN

Members of the monothiol glutaredoxin family and members of the BolA-like protein family have recently emerged as specific interacting partners involved in iron-sulfur protein maturation and redox regulation pathways. It is known that human mitochondrial BOLA1 and BOLA3 form [2Fe-2S] cluster-bridged dimeric heterocomplexes with the monothiol glutaredoxin GRX5. The structure and cluster coordination of the two [2Fe-2S] heterocomplexes as well as their molecular function are, however, not defined yet. Experimentally-driven structural models of the two [2Fe-2S] cluster-bridged dimeric heterocomplexes, the relative stability of the two complexes and the redox properties of the [2Fe-2S] cluster bound to these complexes are here presented on the basis of UV/vis, CD, EPR and NMR spectroscopies and computational protein-protein docking. While the BOLA1-GRX5 complex coordinates a reduced, Rieske-type [2Fe-2S]1+ cluster, an oxidized, ferredoxin-like [2Fe-2S]2+ cluster is present in the BOLA3-GRX5 complex. The [2Fe-2S] BOLA1-GRX5 complex is preferentially formed over the [2Fe-2S] BOLA3-GRX5 complex, as a result of a higher cluster binding affinity. All these observed differences provide the first indications discriminating the molecular function of the two [2Fe-2S] heterocomplexes.


Asunto(s)
Glutarredoxinas/química , Proteínas Hierro-Azufre/química , Proteínas Mitocondriales/química , Proteínas/química , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Espectroscopía de Resonancia Magnética
13.
Proc Natl Acad Sci U S A ; 111(17): 6203-8, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24733926

RESUMEN

Monothiol glutaredoxins play a crucial role in iron-sulfur (Fe/S) protein biogenesis. Essentially all of them can coordinate a [2Fe-2S] cluster and have been proposed to mediate the transfer of [2Fe-2S] clusters from scaffold proteins to target apo proteins, possibly by acting as cluster transfer proteins. The molecular basis of [2Fe-2S] cluster transfer from monothiol glutaredoxins to target proteins is a fundamental, but still unresolved, aspect to be defined in Fe/S protein biogenesis. In mitochondria monothiol glutaredoxin 5 (GRX5) is involved in the maturation of all cellular Fe/S proteins and participates in cellular iron regulation. Here we show that the structural plasticity of the dimeric state of the [2Fe-2S] bound form of human GRX5 (holo hGRX5) is the crucial factor that allows an efficient cluster transfer to the partner proteins human ISCA1 and ISCA2 by a specific protein-protein recognition mechanism. Holo hGRX5 works as a metallochaperone preventing the [2Fe-2S] cluster to be released in solution in the presence of physiological concentrations of glutathione and forming a transient, cluster-mediated protein-protein intermediate with two physiological protein partners receiving the [2Fe-2S] cluster. The cluster transfer mechanism defined here may extend to other mitochondrial [2Fe-2S] target proteins.


Asunto(s)
Proteínas Hierro-Azufre/metabolismo , Hierro/metabolismo , Proteínas Mitocondriales/metabolismo , Azufre/metabolismo , Apoproteínas/química , Apoproteínas/metabolismo , Glutarredoxinas/química , Glutarredoxinas/metabolismo , Glutatión/metabolismo , Humanos , Proteínas Hierro-Azufre/química , Espectroscopía de Resonancia Magnética , Proteínas Mitocondriales/química , Modelos Moleculares , Unión Proteica , Estructura Cuaternaria de Proteína , Soluciones , Espectrofotometría Ultravioleta
14.
Nature ; 465(7298): 645-8, 2010 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-20463663

RESUMEN

Copper is an essential trace element for eukaryotes and most prokaryotes. However, intracellular free copper must be strictly limited because of its toxic side effects. Complex systems for copper trafficking evolved to satisfy cellular requirements while minimizing toxicity. The factors driving the copper transfer between protein partners along cellular copper routes are, however, not fully rationalized. Until now, inconsistent, scattered and incomparable data on the copper-binding affinities of copper proteins have been reported. Here we determine, through a unified electrospray ionization mass spectrometry (ESI-MS)-based strategy, in an environment that mimics the cellular redox milieu, the apparent Cu(I)-binding affinities for a representative set of intracellular copper proteins involved in enzymatic redox catalysis, in copper trafficking to and within various cellular compartments, and in copper storage. The resulting thermodynamic data show that copper is drawn to the enzymes that require it by passing from one copper protein site to another, exploiting gradients of increasing copper-binding affinity. This result complements the finding that fast copper-transfer pathways require metal-mediated protein-protein interactions and therefore protein-protein specific recognition. Together with Cu,Zn-SOD1, metallothioneins have the highest affinity for copper(I), and may play special roles in the regulation of cellular copper distribution; however, for kinetic reasons they cannot demetallate copper enzymes. Our study provides the thermodynamic basis for the kinetic processes that lead to the distribution of cellular copper.


Asunto(s)
Proteínas Portadoras/metabolismo , Cobre/metabolismo , Espacio Intracelular/metabolismo , Animales , Biocatálisis , Cationes Monovalentes/metabolismo , Cobre/aislamiento & purificación , Ciclooxigenasa 2/química , Ciclooxigenasa 2/metabolismo , Ditiotreitol/metabolismo , Glutatión/metabolismo , Humanos , Transporte Iónico , Cinética , Ligandos , Metalotioneína/metabolismo , Mitocondrias Hepáticas , Oxidación-Reducción , Unión Proteica , Ratas , Espectrometría de Masa por Ionización de Electrospray , Termodinámica
15.
Proc Natl Acad Sci U S A ; 110(18): 7136-41, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23596212

RESUMEN

Biogenesis of iron-sulfur cluster proteins is a highly regulated process that requires complex protein machineries. In the cytosolic iron-sulfur protein assembly machinery, two human key proteins--NADPH-dependent diflavin oxidoreductase 1 (Ndor1) and anamorsin--form a stable complex in vivo that was proposed to provide electrons for assembling cytosolic iron-sulfur cluster proteins. The Ndor1-anamorsin interaction was also suggested to be implicated in the regulation of cell survival/death mechanisms. In the present work we unravel the molecular basis of recognition between Ndor1 and anamorsin and of the electron transfer process. This is based on the structural characterization of the two partner proteins, the investigation of the electron transfer process, and the identification of those protein regions involved in complex formation and those involved in electron transfer. We found that an unstructured region of anamorsin is essential for the formation of a specific and stable protein complex with Ndor1, whereas the C-terminal region of anamorsin, containing the [2Fe-2S] redox center, transiently interacts through complementary charged residues with the FMN-binding site region of Ndor1 to perform electron transfer. Our results propose a molecular model of the electron transfer process that is crucial for understanding the functional role of this interaction in human cells.


Asunto(s)
Flavoproteínas/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Proteínas Hierro-Azufre/biosíntesis , Oxidorreductasas/biosíntesis , Biosíntesis de Proteínas , Transporte de Electrón , Mononucleótido de Flavina/metabolismo , Flavoproteínas/química , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas Hierro-Azufre/química , Modelos Biológicos , Modelos Moleculares , Oxidorreductasas/química , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína
16.
J Am Chem Soc ; 137(51): 16133-43, 2015 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-26613676

RESUMEN

In eukaryotes, the interaction between members of the monothiol glutaredoxin family and members of the BolA-like protein family has been involved in iron metabolism. To investigate the still unknown functional role of the interaction between human glutaredoxin-3 (GRX3) and its protein partner BOLA2, we characterized at the atomic level the interaction of apo BOLA2 with the apo and holo states of GRX3 and studied the role of BOLA2 in the GRX3-dependent anamorsin maturation pathway. From these studies, it emerged that apo GRX3 and apo BOLA2 form a heterotrimeric complex, composed by two BOLA2 molecules and one GRX3 molecule. This complex is able to bind two [2Fe-2S](2+) clusters, each being bridged between a BOLA2 molecule and a monothiol glutaredoxin domain of GRX3, and to transfer both [2Fe-2S](2+) clusters to apo anamorsin producing its mature holo state. Collectively, the data suggest that the heterotrimeric complex can work as a [2Fe-2S](2+) cluster transfer component in cytosolic Fe/S protein maturation pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas/fisiología , Humanos
17.
J Am Chem Soc ; 136(46): 16240-50, 2014 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-25347204

RESUMEN

The generation of [4Fe-4S] clusters in mitochondria critically depends, in both yeast and human cells, on two A-type ISC proteins (in mammals named ISCA1 and ISCA2), which perform a nonredundant functional role forming in vivo a heterocomplex. The molecular function of ISCA1 and ISCA2 proteins, i.e., how these proteins help in generating [4Fe-4S] clusters, is still unknown. In this work we have structurally characterized the Fe/S cluster binding properties of human ISCA2 and investigated in vitro whether and how a [4Fe-4S] cluster is assembled when human ISCA1 and ISCA2 interact with the physiological [2Fe-2S](2+) cluster-donor human GRX5. We found that (i) ISCA2 binds either [2Fe-2S] or [4Fe-4S] cluster in a dimeric state, and (ii) two molecules of [2Fe-2S](2+) GRX5 donate their cluster to a heterodimeric ISCA1/ISCA2 complex. This complex acts as an "assembler" of [4Fe-4S] clusters; i.e., the two GRX5-donated [2Fe-2S](2+) clusters generate a [4Fe-4S](2+) cluster. The formation of the same [4Fe-4S](2+) cluster-bound heterodimeric species is also observed by having first one [2Fe-2S](2+) cluster transferred from GRX5 to each individual ISCA1 and ISCA2 proteins to form [2Fe-2S](2+) ISCA2 and [2Fe-2S](2+) ISCA1, and then mixing them together. These findings imply that such heterodimeric complex is the functional unit in mitochondria receiving [2Fe-2S] clusters from hGRX5 and assembling [4Fe-4S] clusters before their transfer to the final target apo proteins.


Asunto(s)
Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/metabolismo , Hierro/metabolismo , Mitocondrias/metabolismo , Azufre/metabolismo , Apoproteínas/química , Apoproteínas/metabolismo , Humanos , Modelos Moleculares , Multimerización de Proteína , Estructura Cuaternaria de Proteína
18.
J Biomol NMR ; 58(2): 123-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24414179

RESUMEN

A crucial factor for the understanding of structure-function relationships in metalloproteins is the identification of NMR signals from residues surrounding the metal cofactor. When the latter is paramagnetic, the NMR information in the proximity of the metal center may be scarce, because fast nuclear relaxation quenches signal intensity and coherence transfer efficiency. To identify residues at a short distance from a paramagnetic center, we developed a modified version of the ¹5N-HSQC experiment where (1) an inversion recovery filter is added prior to HSQC, (2) the INEPT period has been optimized according to fast relaxation of interested spins, (3) the inverse INEPT has been eliminated and signals acquired as antiphase doublets. The experiment has been successfully tested on a human [Fe2S2] protein which is involved in the biogenesis of iron-sulfur proteins. Thirteen HN resonances, unobserved with conventional HSQC experiments, could be identified. The structural arrangement of the protein scaffold in the proximity of the Fe/S cluster is fundamental to comprehend the molecular processes responsible for the transfer of Fe/S groups in the iron-sulfur protein assembly machineries.


Asunto(s)
Metaloproteínas/química , Resonancia Magnética Nuclear Biomolecular , Humanos , Isótopos de Nitrógeno/química , Resonancia Magnética Nuclear Biomolecular/métodos
19.
Proc Natl Acad Sci U S A ; 108(12): 4811-6, 2011 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-21383138

RESUMEN

Oxidative protein folding in the mitochondrial intermembrane space requires the transfer of a disulfide bond from MIA40 to the substrate. During this process MIA40 is reduced and regenerated to a functional state through the interaction with the flavin-dependent sulfhydryl oxidase ALR. Here we present the mechanistic basis of ALR-MIA40 interaction at atomic resolution by biochemical and structural analyses of the mitochondrial ALR isoform and its covalent mixed disulfide intermediate with MIA40. This ALR isoform contains a folded FAD-binding domain at the C-terminus and an unstructured, flexible N-terminal domain, weakly and transiently interacting one with the other. A specific region of the N-terminal domain guides the interaction with the MIA40 substrate binding cleft (mimicking the interaction of the substrate itself), without being involved in the import of ALR. The hydrophobicity-driven binding of this region ensures precise protein-protein recognition needed for an efficient electron transfer process.


Asunto(s)
Reductasas del Citocromo/química , Flavina-Adenina Dinucleótido/química , Proteínas de Transporte de Membrana Mitocondrial/química , Sitios de Unión , Reductasas del Citocromo/metabolismo , Transporte de Electrón/fisiología , Flavina-Adenina Dinucleótido/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Isoenzimas/química , Isoenzimas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Estructura Terciaria de Proteína , Especificidad por Sustrato/fisiología
20.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119786, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38901495

RESUMEN

Over the last decade, structural aspects involving iron­sulfur (Fe/S) protein biogenesis have played an increasingly important role in understanding the high mechanistic complexity of mitochondrial and cytosolic machineries maturing Fe/S proteins. In this respect, solution NMR has had a significant impact because of its ability to monitor transient protein-protein interactions, which are abundant in the networks of pathways leading to Fe/S cluster biosynthesis and transfer, as well as thanks to the developments of paramagnetic NMR in both terms of new methodologies and accurate data interpretation. Here, we review the use of solution NMR in characterizing the structural aspects of human Fe/S proteins and their interactions in the framework of Fe/S protein biogenesis. We will first present a summary of the recent advances that have been achieved by paramagnetic NMR and then we will focus our attention on the role of solution NMR in the field of human Fe/S protein biogenesis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA