Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 6932, 2021 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-34836944

RESUMEN

Unsaturated fatty acids (UFAs) are essential for functional membrane phospholipids in most bacteria. The bifunctional dehydrogenase/isomerase FabX is an essential UFA biosynthesis enzyme in the widespread human pathogen Helicobacter pylori, a bacterium etiologically related to 95% of gastric cancers. Here, we present the crystal structures of FabX alone and in complexes with an octanoyl-acyl carrier protein (ACP) substrate or with holo-ACP. FabX belongs to the nitronate monooxygenase (NMO) flavoprotein family but contains an atypical [4Fe-4S] cluster absent in all other family members characterized to date. FabX binds ACP via its positively charged α7 helix that interacts with the negatively charged α2 and α3 helices of ACP. We demonstrate that the [4Fe-4S] cluster potentiates FMN oxidation during dehydrogenase catalysis, generating superoxide from an oxygen molecule that is locked in an oxyanion hole between the FMN and the active site residue His182. Both the [4Fe-4S] and FMN cofactors are essential for UFA synthesis, and the superoxide is subsequently excreted by H. pylori as a major resource of peroxide which may contribute to its pathogenic function in the corrosion of gastric mucosa.


Asunto(s)
Proteínas Bacterianas/ultraestructura , Ácidos Grasos Insaturados/biosíntesis , Helicobacter pylori/enzimología , Proteínas Hierro-Azufre/ultraestructura , Oxigenasas de Función Mixta/ultraestructura , Proteína Transportadora de Acilo/metabolismo , Proteína Transportadora de Acilo/ultraestructura , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico/genética , Cristalografía por Rayos X , Helicobacter pylori/genética , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Oxidación-Reducción
2.
Nat Commun ; 12(1): 5387, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34508071

RESUMEN

Photosynthesis and respiration rely upon a proton gradient to produce ATP. In photosynthesis, the Respiratory Complex I homologue, Photosynthetic Complex I (PS-CI) is proposed to couple ferredoxin oxidation and plastoquinone reduction to proton pumping across thylakoid membranes. However, little is known about the PS-CI molecular mechanism and attempts to understand its function have previously been frustrated by its large size and high lability. Here, we overcome these challenges by pushing the limits in sample size and spectroscopic sensitivity, to determine arguably the most important property of any electron transport enzyme - the reduction potentials of its cofactors, in this case the iron-sulphur clusters of PS-CI (N0, N1 and N2), and unambiguously assign them to the structure using double electron-electron resonance. We have thus determined the bioenergetics of the electron transfer relay and provide insight into the mechanism of PS-CI, laying the foundations for understanding of how this important bioenergetic complex functions.


Asunto(s)
Proteínas Bacterianas/metabolismo , Metabolismo Energético , Proteínas Hierro-Azufre/metabolismo , Complejo de Proteína del Fotosistema I/metabolismo , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/ultraestructura , Espectroscopía de Resonancia por Spin del Electrón , Transporte de Electrón , Proteínas Hierro-Azufre/ultraestructura , Complejo de Proteína del Fotosistema I/aislamiento & purificación , Complejo de Proteína del Fotosistema I/ultraestructura , Synechocystis/metabolismo
3.
J Biol Chem ; 296: 100474, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33640456

RESUMEN

Respiratory complex I (NADH:ubiquinone oxidoreductase), the first enzyme of the electron-transport chain, captures the free energy released by NADH oxidation and ubiquinone reduction to translocate protons across an energy-transducing membrane and drive ATP synthesis during oxidative phosphorylation. The cofactor that transfers the electrons directly to ubiquinone is an iron-sulfur cluster (N2) located in the NDUFS2/NUCM subunit. A nearby arginine residue (R121), which forms part of the second coordination sphere of the N2 cluster, is known to be posttranslationally dimethylated but its functional and structural significance are not known. Here, we show that mutations of this arginine residue (R121M/K) abolish the quinone-reductase activity, concomitant with disappearance of the N2 signature from the electron paramagnetic resonance (EPR) spectrum. Analysis of the cryo-EM structure of NDUFS2-R121M complex I at 3.7 Å resolution identified the absence of the cubane N2 cluster as the cause of the dysfunction, within an otherwise intact enzyme. The mutation further induced localized disorder in nearby elements of the quinone-binding site, consistent with the close connections between the cluster and substrate-binding regions. Our results demonstrate that R121 is required for the formation and/or stability of the N2 cluster and highlight the importance of structural analyses for mechanistic interpretation of biochemical and spectroscopic data on complex I variants.


Asunto(s)
Complejo I de Transporte de Electrón/química , Proteínas Fúngicas/química , Proteínas Hierro-Azufre/química , Proteínas Mitocondriales/química , Yarrowia/enzimología , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/ultraestructura , Proteínas Fúngicas/genética , Proteínas Fúngicas/ultraestructura , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/ultraestructura , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Estabilidad Proteica , Yarrowia/genética
4.
FEBS J ; 288(9): 2870-2883, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32979284

RESUMEN

Proteins destined to various intra- and extra-cellular locations must traverse membranes most frequently in an unfolded form. When the proteins being translocated need to remain in a folded state, specialized cellular transport machinery is used. One such machine is the membrane-bound AAA protein Bcs1 (Bcs1), which assists the iron-sulfur protein, an essential subunit of the respiratory Complex III, across the mitochondrial inner membrane. Recent structure determinations of mouse and yeast Bcs1 in three different nucleotide states reveal its homo-heptameric association and at least two dramatically different conformations. The apo and ADP-bound structures are similar, both containing a large substrate-binding cavity accessible to the mitochondrial matrix space, which contracts by concerted motion of the ATPase domains upon ATP binding, suggesting that bound substrate could then be pushed across the membrane. ATP hydrolysis drives substrate release and resets Bcs1 conformation back to the apo/ADP form. These structures shed new light on the mechanism of folded protein translocation across a membrane, provide better understanding on the assembly process of the respiratory Complex III, and correlate clinical presentations of disease-associated mutations with their locations in the 3D structure.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Chaperonas Moleculares/genética , Conformación Proteica , Proteínas de Saccharomyces cerevisiae/genética , ATPasas Asociadas con Actividades Celulares Diversas/ultraestructura , Adenosina Trifosfatasas/genética , Adenosina Trifosfato/genética , Animales , Complejo III de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/ultraestructura , Humanos , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/ultraestructura , Ratones , Mitocondrias/ultraestructura , Proteínas Mitocondriales/ultraestructura , Chaperonas Moleculares/ultraestructura , Dominios Proteicos/genética , Pliegue de Proteína , Transporte de Proteínas/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/ultraestructura
5.
FEBS J ; 288(9): 3010-3023, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33124176

RESUMEN

Metalloproteins play key roles across biology, and knowledge of their structure is essential to understand their physiological role. For those metalloproteins containing paramagnetic states, the enhanced relaxation caused by the unpaired electrons often makes signal detection unfeasible near the metal center, precluding adequate structural characterization right where it is more biochemically relevant. Here, we report a protein structure determination by NMR where two different sets of restraints, one containing Nuclear Overhauser Enhancements (NOEs) and another containing Paramagnetic Relaxation Enhancements (PREs), are used separately and eventually together. The protein PioC from Rhodopseudomonas palustris TIE-1 is a High Potential Iron-Sulfur Protein (HiPIP) where the [4Fe-4S] cluster is paramagnetic in both oxidation states at room temperature providing the source of PREs used as alternative distance restraints. Comparison of the family of structures obtained using NOEs only, PREs only, and the combination of both reveals that the pairwise root-mean-square deviation (RMSD) between them is similar and comparable with the precision within each family. This demonstrates that, under favorable conditions in terms of protein size and paramagnetic effects, PREs can efficiently complement and eventually replace NOEs for the structural characterization of small paramagnetic metalloproteins and de novo-designed metalloproteins by NMR. DATABASES: The 20 conformers with the lowest target function constituting the final family obtained using the full set of NMR restraints were deposited to the Protein Data Bank (PDB ID: 6XYV). The 20 conformers with the lowest target function obtained using NOEs only (PDB ID: 7A58) and PREs only (PDB ID: 7A4L) were also deposited to the Protein Data Bank. The chemical shift assignments were deposited to the BMRB (code 34487).


Asunto(s)
Proteínas Bacterianas/ultraestructura , Proteínas Hierro-Azufre/ultraestructura , Metaloproteínas/ultraestructura , Proteínas del Complejo del Centro de Reacción Fotosintética/ultraestructura , Conformación Proteica , Rhodopseudomonas/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Electrones , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Metaloproteínas/química , Metaloproteínas/genética , Modelos Moleculares , Proteínas del Complejo del Centro de Reacción Fotosintética/química , Proteínas del Complejo del Centro de Reacción Fotosintética/genética , Rhodopseudomonas/química
6.
FEBS J ; 287(11): 2312-2327, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31724821

RESUMEN

During its late steps, the mitochondrial iron-sulfur cluster (ISC) assembly machinery leads to the formation of [4Fe-4S] clusters. In vivo studies revealed that several proteins are implicated in the biosynthesis and trafficking of [4Fe-4S] clusters in mitochondria. However, they do not provide a clear picture into how these proteins cooperate. Here, we showed that three late-acting components of the mitochondrial ISC assembly machinery (GLRX5, BOLA3, and NFU1) are part of a ISC assembly pathway leading to the synthesis of a [4Fe-4S]2+ cluster on NFU1. We showed that the [2Fe-2S]2+ GLRX5-BOLA3 complex transfers its cluster to monomeric apo NFU1 to form, in the presence of a reductant, a [4Fe-4S]2+ cluster bound to dimeric NFU1. The cluster formation on NFU1 does not occur with [2Fe-2S]2+ GLRX5, and thus, the [4Fe-4S] cluster assembly pathway is activated only in the presence of BOLA3. These results define NFU1 as an 'assembler' of [4Fe-4S] clusters, that is, a protein able of converting two [2Fe-2S]2+ clusters into a [4Fe-4S]2+ cluster. Finally, we found that the [4Fe-4S]2+ cluster bound to NFU1 has a coordination site which is easily accessible to sulfur-containing ligands, as is typically observed in metallochaperones. This finding supports a role for NFU1 in promoting rapid and controlled cluster-exchange reaction.


Asunto(s)
Proteínas Portadoras/genética , Glutarredoxinas/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Proteínas Portadoras/ultraestructura , Glutarredoxinas/ultraestructura , Humanos , Hierro/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/ultraestructura , Mitocondrias/ultraestructura , Proteínas Mitocondriales/ultraestructura , Simulación del Acoplamiento Molecular , Biogénesis de Organelos , Transducción de Señal/genética , Azufre
7.
Mol Microbiol ; 112(6): 1769-1783, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31532036

RESUMEN

IscU is a central component of the ISC machinery and serves as a scaffold for the de novo assembly of iron-sulfur (Fe-S) clusters prior to their delivery to target apo-Fe-S proteins. However, the molecular mechanism is not yet fully understood. In this study, we have conducted mutational analysis of E. coli IscU using the recently developed genetic complementation system of a mutant that can survive without Fe-S clusters. The Fe-S cluster ligands (C37, C63, H105, C106) and the proximal D39 and K103 residues are essential for in vivo function of IscU and could not be substituted with any other amino acids. Furthermore, we found that substitution of Y3, a strictly conserved residue among IscU homologs, abolished in vivo functions. Surprisingly, a second-site suppressor mutation in IscS (A349V) reverted the defect caused by IscU Y3 substitutions. Biochemical analysis revealed that IscU Y3 was crucial for functional interaction with IscS and sulfur transfer between the two proteins. Our findings suggest that the critical role of IscU Y3 is linked to the conformational dynamics of the flexible loop of IscS, which is required for the ingenious sulfur transfer to IscU.


Asunto(s)
Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Aminoácidos/genética , Proteínas Bacterianas/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestructura , Hierro/metabolismo , Proteínas Hierro-Azufre/ultraestructura , Ligandos , Mutación/genética , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad , Azufre/metabolismo
8.
J Mol Biol ; 431(22): 4523-4526, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31473159

RESUMEN

Bacterial NADPH-dependent glutamate synthase (GltS) is a complex iron-sulfur flavoprotein that catalyzes the reductive synthesis of two L-Glu molecules from L-Gln and 2-oxo-glutarate. GltS functional unit hosts an α-subunit (αGltS) and a ß-subunit (ßGltS) that assemble in different αß oligomers in solution. Here, we present the cryo-electron microscopy structures of Azospirillum brasilense GltS in four different oligomeric states (α4ß3, α4ß4, α6ß4 and α6ß6, in the 3.5- to 4.1-Å resolution range). Our study provides a comprehensive GltS model that details the inter-protomeric assemblies and allows unequivocal location of the FAD cofactor and of two electron transfer [4Fe-4S]+1,+2 clusters within ßGltS.


Asunto(s)
Azospirillum brasilense/enzimología , Microscopía por Crioelectrón/métodos , Glutamato Sintasa/metabolismo , Glutamato Sintasa/ultraestructura , Catálisis , Transporte de Electrón , Mononucleótido de Flavina/metabolismo , Flavina-Adenina Dinucleótido/metabolismo , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/ultraestructura
9.
Annu Rev Biochem ; 88: 163-190, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31220976

RESUMEN

Many DNA-processing enzymes have been shown to contain a [4Fe4S] cluster, a common redox cofactor in biology. Using DNA electrochemistry, we find that binding of the DNA polyanion promotes a negative shift in [4Fe4S] cluster potential, which corresponds thermodynamically to a ∼500-fold increase in DNA-binding affinity for the oxidized [4Fe4S]3+ cluster versus the reduced [4Fe4S]2+ cluster. This redox switch can be activated from a distance using DNA charge transport (DNA CT) chemistry. DNA-processing proteins containing the [4Fe4S] cluster are enumerated, with possible roles for the redox switch highlighted. A model is described where repair proteins may signal one another using DNA-mediated charge transport as a first step in their search for lesions. The redox switch in eukaryotic DNA primases appears to regulate polymerase handoff, and in DNA polymerase δ, the redox switch provides a means to modulate replication in response to oxidative stress. We thus describe redox signaling interactions of DNA-processing [4Fe4S] enzymes, as well as the most interesting potential players to consider in delineating new DNA-mediated redox signaling networks.


Asunto(s)
ADN Glicosilasas/química , ADN Helicasas/química , ADN Polimerasa Dirigida por ADN/química , ADN/química , Endonucleasas/química , Genoma , Proteínas Hierro-Azufre/química , Animales , Bacterias/genética , Bacterias/metabolismo , ADN/metabolismo , ADN/ultraestructura , Daño del ADN , ADN Glicosilasas/metabolismo , ADN Glicosilasas/ultraestructura , ADN Helicasas/metabolismo , ADN Helicasas/ultraestructura , Reparación del ADN , Replicación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , ADN Polimerasa Dirigida por ADN/ultraestructura , Espectroscopía de Resonancia por Spin del Electrón , Endonucleasas/metabolismo , Endonucleasas/ultraestructura , Proteínas Hierro-Azufre/metabolismo , Proteínas Hierro-Azufre/ultraestructura , Oxidación-Reducción , Unión Proteica , Transducción de Señal , Termodinámica
10.
Nat Commun ; 10(1): 2210, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-31101807

RESUMEN

The core machinery for de novo biosynthesis of iron-sulfur clusters (ISC), located in the mitochondria matrix, is a five-protein complex containing the cysteine desulfurase NFS1 that is activated by frataxin (FXN), scaffold protein ISCU, accessory protein ISD11, and acyl-carrier protein ACP. Deficiency in FXN leads to the loss-of-function neurodegenerative disorder Friedreich's ataxia (FRDA). Here the 3.2 Å resolution cryo-electron microscopy structure of the FXN-bound active human complex, containing two copies of the NFS1-ISD11-ACP-ISCU-FXN hetero-pentamer, delineates the interactions of FXN with other component proteins of the complex. FXN binds at the interface of two NFS1 and one ISCU subunits, modifying the local environment of a bound zinc ion that would otherwise inhibit NFS1 activity in complexes without FXN. Our structure reveals how FXN facilitates ISC production through stabilizing key loop conformations of NFS1 and ISCU at the protein-protein interfaces, and suggests how FRDA clinical mutations affect complex formation and FXN activation.


Asunto(s)
Liasas de Carbono-Azufre/ultraestructura , Ataxia de Friedreich/patología , Proteínas de Unión a Hierro/ultraestructura , Proteínas Hierro-Azufre/ultraestructura , Mitocondrias/ultraestructura , Liasas de Carbono-Azufre/aislamiento & purificación , Liasas de Carbono-Azufre/metabolismo , Microscopía por Crioelectrón , Ataxia de Friedreich/genética , Hierro/metabolismo , Proteínas de Unión a Hierro/aislamiento & purificación , Proteínas de Unión a Hierro/metabolismo , Proteínas Hierro-Azufre/aislamiento & purificación , Proteínas Hierro-Azufre/metabolismo , Mitocondrias/metabolismo , Modelos Moleculares , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Azufre/metabolismo , Zinc/metabolismo , Frataxina
11.
J Struct Biol ; 202(3): 264-274, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29601957

RESUMEN

A systematic rationalization of the hundreds of proteins harboring iron-sulfur clusters and able to exhibit the most diverse biological functions is missing. In this picture we have already reviewed structure/electrochemistry of metalloproteins expressing single types of iron-sulfur centres [namely, {Fe(Cys)4}, {[Fe2S2](Cys)4}, {[Fe2S2](Cys)3(X)} (X = Asp, Arg, His), {[Fe2S2](Cys)2(His)2}, {[Fe3S4](Cys)3}, {[Fe4S4](Cys)4} and {[Fe4S4](SγCys)3(nonthiolate ligand)}] and their synthetic analogs. Recently we are focussing on structure/electrochemistry of metalloproteins containing iron-sulfur centres of different nuclearities. Having started such a subject with proteins harboring [4Fe-4S] and [2Fe-2S] (Zanello, 2017c) as well as [4Fe-4S] and [3Fe-4S] (Zanello, in press) clusters, we now provide the state of art of proteins harboring [4Fe-4S], [3Fe-4S] and [2Fe-2S] clusters, a subject that resulted strictly limited to enzymes active in the respiratory Complex II.


Asunto(s)
Proteínas Hierro-Azufre/ultraestructura , Metaloproteínas/ultraestructura , Conformación Proteica , Cisteína/química , Electroquímica , Espectroscopía de Resonancia por Spin del Electrón , Proteínas Hierro-Azufre/química , Metaloproteínas/química , Azufre/química
12.
J Struct Biol ; 202(3): 250-263, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29428558

RESUMEN

In the context of the plethora of proteins harboring iron-sulfur clusters we have already reviewed structure/electrochemistry of metalloproteins expressing single types of iron-sulfur clusters (namely: {Fe(Cys)4}, {[Fe2S2](Cys)4}, {[Fe2S2](Cys)3(X)} (X = Asp, Arg, His), {[Fe2S2](Cys)2(His)2}, {[Fe3S4](Cys)3}, {[Fe4S4](Cys)4} and {[Fe4S4](SγCys)3(nonthiolate ligand)} cores) and their synthetic analogs. More recently we are focussing on structure/electrochemistry of metalloproteins harboring iron-sulfur centres of different nuclearities. Having started such a subject with proteins harboring [4Fe-4S] and [2Fe-2S] clusters, we now depict the state of art of proteins containing [4Fe-4S] and [3Fe-4S] clusters.


Asunto(s)
Proteínas Hierro-Azufre/ultraestructura , Metaloproteínas/ultraestructura , Conformación Proteica , Cisteína/química , Electroquímica , Espectroscopía de Resonancia por Spin del Electrón , Proteínas Hierro-Azufre/química , Metaloproteínas/química , Azufre/química
13.
Science ; 357(6352): 699-703, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28818947

RESUMEN

In methanogenic archaea, the carbon dioxide (CO2) fixation and methane-forming steps are linked through the heterodisulfide reductase (HdrABC)-[NiFe]-hydrogenase (MvhAGD) complex that uses flavin-based electron bifurcation to reduce ferredoxin and the heterodisulfide of coenzymes M and B. Here, we present the structure of the native heterododecameric HdrABC-MvhAGD complex at 2.15-angstrom resolution. HdrB contains two noncubane [4Fe-4S] clusters composed of fused [3Fe-4S]-[2Fe-2S] units sharing 1 iron (Fe) and 1 sulfur (S), which were coordinated at the CCG motifs. Soaking experiments showed that the heterodisulfide is clamped between the two noncubane [4Fe-4S] clusters and homolytically cleaved, forming coenzyme M and B bound to each iron. Coenzymes are consecutively released upon one-by-one electron transfer. The HdrABC-MvhAGD atomic model serves as a structural template for numerous HdrABC homologs involved in diverse microbial metabolic pathways.


Asunto(s)
Proteínas Arqueales/química , Proteínas Hierro-Azufre/química , Methanococcaceae/enzimología , Oxidorreductasas/química , Secuencias de Aminoácidos , Proteínas Arqueales/ultraestructura , Coenzimas/química , Coenzimas/ultraestructura , Cristalografía por Rayos X , Transporte de Electrón , Ferredoxinas/química , Hierro/química , Proteínas Hierro-Azufre/ultraestructura , Redes y Vías Metabólicas , Oxidación-Reducción , Oxidorreductasas/ultraestructura , Dominios Proteicos , Azufre/química
14.
Nat Commun ; 8: 16110, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28726794

RESUMEN

Iron-sulfur clusters are ubiquitous in biology and function in electron transfer and catalysis. They are assembled from iron and cysteine sulfur on protein scaffolds. Iron is typically stored as iron oxyhydroxide, ferrihydrite, encapsulated in 12 nm shells of ferritin, which buffers cellular iron availability. Here we have characterized IssA, a protein that stores iron and sulfur as thioferrate, an inorganic anionic polymer previously unknown in biology. IssA forms nanoparticles reaching 300 nm in diameter and is the largest natural metalloprotein complex known. It is a member of a widely distributed protein family that includes nitrogenase maturation factors, NifB and NifX. IssA nanoparticles are visible by electron microscopy as electron-dense bodies in the cytoplasm. Purified nanoparticles appear to be generated from 20 nm units containing ∼6,400 Fe atoms and ∼170 IssA monomers. In support of roles in both iron-sulfur storage and cluster biosynthesis, IssA reconstitutes the [4Fe-4S] cluster in ferredoxin in vitro.


Asunto(s)
Proteínas Bacterianas/ultraestructura , Proteínas Hierro-Azufre/ultraestructura , Nanopartículas/ultraestructura , Pyrococcus furiosus/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Ferredoxinas/metabolismo , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/metabolismo , Microscopía Electrónica , Nanopartículas/química , Pyrococcus furiosus/química
15.
FEBS J ; 284(14): 2163-2166, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28165666

RESUMEN

The high-potential iron-sulfur protein (HiPIP) is a small (~ 80 residues) soluble metalloprotein functioning as an electron carrier in photosynthetic bacteria. HiPIP has one Fe4 S4 cluster at its molecular center. Its electronic structure is important for understanding electron transport. We recently succeeded in determining an ultra-high-resolution structure and analyzing the charge-density of HiPIP by using X-ray diffraction data at 0.48 Å resolution. The distribution of valence electrons in the iron-sulfur cluster and in the protein environment were clearly visualized, which is the first successful case for metalloproteins. In addition, a topological analysis of the charge density provided information about the electronic structure of the cluster.


Asunto(s)
Bacterias/química , Proteínas Bacterianas/química , Proteínas Bacterianas/ultraestructura , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/ultraestructura , Proteínas del Complejo del Centro de Reacción Fotosintética/química , Proteínas del Complejo del Centro de Reacción Fotosintética/ultraestructura , Bacterias/clasificación , Bacterias/ultraestructura , Cristalografía por Rayos X , Transporte de Electrón , Electrones , Fotosíntesis , Conformación Proteica
17.
J Mol Biol ; 426(19): 3287-3304, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25102080

RESUMEN

Recently, progress has been made toward understanding the functional diversity of bacterial microcompartment (MCP) systems, which serve as protein-based metabolic organelles in diverse microbes. New types of MCPs have been identified, including the glycyl-radical propanediol (Grp) MCP. Within these elaborate protein complexes, BMC-domain shell proteins [bacterial microcompartment (in reference to the shell protein domain)] assemble to form a polyhedral barrier that encapsulates the enzymatic contents of the MCP. Interestingly, the Grp MCP contains a number of shell proteins with unusual sequence features. GrpU is one such shell protein whose amino acid sequence is particularly divergent from other members of the BMC-domain superfamily of proteins that effectively defines all MCPs. Expression, purification, and subsequent characterization of the protein showed, unexpectedly, that it binds an iron-sulfur cluster. We determined X-ray crystal structures of two GrpU orthologs, providing the first structural insight into the homohexameric BMC-domain shell proteins of the Grp system. The X-ray structures of GrpU, both obtained in the apo form, combined with spectroscopic analyses and computational modeling, show that the metal cluster resides in the central pore of the BMC shell protein at a position of broken 6-fold symmetry. The result is a structurally polymorphic iron-sulfur cluster binding site that appears to be unique among metalloproteins studied to date.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/ultraestructura , Proteínas Hierro-Azufre/química , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Sitios de Unión , Clostridium/metabolismo , Cristalografía por Rayos X , Proteínas Hierro-Azufre/ultraestructura , Modelos Moleculares , Pectobacterium/metabolismo , Glicoles de Propileno/química , Glicoles de Propileno/metabolismo , Estructura Terciaria de Proteína
18.
PLoS Biol ; 11(9): e1001651, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24058293

RESUMEN

Allostery is a fundamental process by which ligand binding to a protein alters its activity at a distinct site. There is growing evidence that allosteric cooperativity can be communicated by modulation of protein dynamics without conformational change. The mechanisms, however, for communicating dynamic fluctuations between sites are debated. We provide a foundational theory for how allostery can occur as a function of low-frequency dynamics without a change in structure. We have generated coarse-grained models that describe the protein backbone motions of the CRP/FNR family transcription factors, CAP of Escherichia coli and GlxR of Corynebacterium glutamicum. The latter we demonstrate as a new exemplar for allostery without conformation change. We observe that binding the first molecule of cAMP ligand is correlated with modulation of the global normal modes and negative cooperativity for binding the second cAMP ligand without a change in mean structure. The theory makes key experimental predictions that are tested through an analysis of variant proteins by structural biology and isothermal calorimetry. Quantifying allostery as a free energy landscape revealed a protein "design space" that identified the inter- and intramolecular regulatory parameters that frame CRP/FNR family allostery. Furthermore, through analyzing CAP variants from diverse species, we demonstrate an evolutionary selection pressure to conserve residues crucial for allosteric control. This finding provides a link between the position of CRP/FNR transcription factors within the allosteric free energy landscapes and evolutionary selection pressures. Our study therefore reveals significant features of the mechanistic basis for allostery. Changes in low-frequency dynamics correlate with allosteric effects on ligand binding without the requirement for a defined spatial pathway. In addition to evolving suitable three-dimensional structures, CRP/FNR family transcription factors have been selected to occupy a dynamic space that fine-tunes biological activity and thus establishes the means to engineer allosteric mechanisms driven by low-frequency dynamics.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteína Receptora de AMP Cíclico/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Hierro-Azufre/metabolismo , Factores de Transcripción/metabolismo , Regulación Alostérica/fisiología , Proteínas Bacterianas/química , Proteínas Bacterianas/clasificación , Sitios de Unión , Corynebacterium glutamicum/metabolismo , Cristalografía por Rayos X , Proteína Receptora de AMP Cíclico/ultraestructura , Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestructura , Proteínas Hierro-Azufre/ultraestructura , Modelos Moleculares , Unión Proteica/fisiología , Conformación Proteica , Termodinámica , Factores de Transcripción/química , Factores de Transcripción/clasificación
19.
Biochemistry ; 50(27): 6133-45, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21651304

RESUMEN

Endonuclease III (EndoIII) is a base excision repair glycosylase that targets damaged pyrimidines and contains a [4Fe-4S] cluster. We have proposed a model where BER proteins that contain redox-active [4Fe-4S] clusters utilize DNA charge transport (CT) as a first step in the detection of DNA lesions. Here, several mutants of EndoIII were prepared to probe their efficiency of DNA/protein charge transport. Cyclic voltammetry experiments on DNA-modified electrodes show that aromatic residues F30, Y55, Y75, and Y82 help mediate charge transport between DNA and the [4Fe-4S] cluster. On the basis of circular dichroism studies to measure protein stability, mutations at residues W178 and Y185 are found to destabilize the protein; these residues may function to protect the [4Fe-4S] cluster. Atomic force microscopy studies furthermore reveal a correlation in the ability of mutants to carry out protein/DNA CT and their ability to relocalize onto DNA strands containing a single base mismatch; EndoIII mutants that are defective in carrying out DNA/protein CT do not redistribute onto mismatch-containing strands, consistent with our model. These results demonstrate a link between the ability of the repair protein to carry out DNA CT and its ability to relocalize near lesions, thus pointing to DNA CT as a key first step in the detection of base damage in the genome.


Asunto(s)
Daño del ADN/genética , Reparación del ADN/genética , ADN Bacteriano/genética , Desoxirribonucleasa (Dímero de Pirimidina)/genética , Proteínas de Escherichia coli/genética , Escherichia coli/enzimología , Escherichia coli/genética , Mutagénesis Sitio-Dirigida , Disparidad de Par Base/genética , ADN Glicosilasas/química , ADN Glicosilasas/genética , ADN Glicosilasas/ultraestructura , ADN Bacteriano/química , ADN Bacteriano/ultraestructura , Desoxirribonucleasa (Dímero de Pirimidina)/química , Desoxirribonucleasa (Dímero de Pirimidina)/ultraestructura , Electroquímica , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestructura , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/ultraestructura , Microscopía de Fuerza Atómica , Oxidación-Reducción , Transporte de Proteínas/genética , Transducción de Señal/genética
20.
Proc Natl Acad Sci U S A ; 102(39): 14058-62, 2005 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-16172380

RESUMEN

Sprouty proteins act as intracellular inhibitors of receptor tyrosine kinase signaling. Here we show that the mammalian Sprouty2 protein contains an iron-sulfur complex that can exist in an oxidized, reduced, or nitrosylated state. Purified Sprouty2 assembles into large monodisperse spheres containing approximately 24 polypeptides per particle. Biochemical experiments indicate that the charge state of the iron within Sprouty2 particles may be insulated from ambient intracellular redox. These features offer the possibility that Sprouty2 particles are capable of receiving, maintaining, and dissipating electrical charge in a manner formally equivalent to a battery.


Asunto(s)
Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/metabolismo , Proteínas/química , Proteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Factores de Crecimiento de Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas Hierro-Azufre/ultraestructura , Proteínas de la Membrana , Ratones , Microscopía Electrónica , Oxidación-Reducción , Conformación Proteica , Proteínas Serina-Treonina Quinasas , Proteínas/ultraestructura , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...