Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 296
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 70(4): 614-627.e7, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29754824

RESUMEN

Bleach (HOCl) is a powerful oxidant that kills bacteria in part by causing protein aggregation. It inactivates ATP-dependent chaperones, rendering cellular proteins mostly dependent on holdases. Here we identified Escherichia coli CnoX (YbbN) as a folding factor that, when activated by bleach via chlorination, functions as an efficient holdase, protecting the substrates of the major folding systems GroEL/ES and DnaK/J/GrpE. Remarkably, CnoX uniquely combines this function with the ability to prevent the irreversible oxidation of its substrates. This dual activity makes CnoX the founding member of a family of proteins, the "chaperedoxins." Because CnoX displays a thioredoxin fold and a tetratricopeptide (TPR) domain, two structural motifs conserved in all organisms, this investigation sets the stage for the discovery of additional chaperedoxins in bacteria and eukaryotes that could cooperate with proteins from both the Hsp60 and Hsp70 families.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Glutatión/metabolismo , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Repeticiones de Tetratricopéptidos , Tiorredoxinas/metabolismo , Secuencia de Aminoácidos , Blanqueadores/farmacología , Chaperonina 10/metabolismo , Chaperonina 60/metabolismo , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/química , Glutatión/química , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Halogenación , Chaperonas Moleculares/química , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Conformación Proteica , Desnaturalización Proteica , Pliegue de Proteína , Homología de Secuencia , Tiorredoxinas/química
2.
Phys Chem Chem Phys ; 24(31): 18543-18551, 2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-35904932

RESUMEN

The dissimilatory sulfite reductase enzyme has very characteristic active site where the substrate binds to an iron site, ligated by a siroheme macrocycle and a thiol directly connected to a [Fe4S4] cluster. This arrangement gives the enzyme remarkable efficiency in reducing sulfite and nitrite all the way to hydrogen sulfide and ammonia. For the first time we present a theoretical study where substrate binding modalities and activation are elucidated using active site models containing proton supply side chains and the [Fe4S4] cluster. Density functional theory (DFT) was deployed in conjunction with the energy decomposition scheme (as implemented in AMS), the quantum theory of atoms in molecules (QTAIM), and conceptual DFT (cDFT) descriptors. We quantified the role of the electrostatic interactions inside the active site created by the side chains as well as the influence of the [Fe4S4] cluster on the substrate binding. Furthermore, using conceptual DFT results we shed light of the activation process, thus, laying foundation for further mechanistic studies. We found that the bonding of the ligands to the iron complex is dominated by electrostatic interactions, but the presence of the [Fe4S4] cubane leads to substantial changes in electronic interaction. The spin state of the cubane, however, affects the binding energy only marginally. The conceptual DFT results show that the presence of the [Fe4S4] cubane affects the reactivity of the active site as it is involved in electron transfer. This is corroborated by an increase in the electrophilicity index, thus making the active site more prone to react with the ligands. The interaction energies between the ligand and the siroheme group are also increased upon the presence of the cubane group, thus, suggesting that the siroheme group is not an innocent spectator but plays an active role in the reactivity of the dSIR active site.


Asunto(s)
Proteínas Hierro-Azufre , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Dominio Catalítico , Escherichia coli , Hierro/metabolismo , Proteínas Hierro-Azufre/química , Ligandos , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo
3.
Chem Soc Rev ; 50(4): 2486-2539, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33475096

RESUMEN

Heme-copper oxidases (HCO), nitric oxide reductases (NOR), and sulfite reductases (SiR) catalyze the multi-electron and multi-proton reductions of O2, NO, and SO32-, respectively. Each of these reactions is important to drive cellular energy production through respiratory metabolism and HCO, NOR, and SiR evolved to contain heteronuclear active sites containing heme/copper, heme/nonheme iron, and heme-[4Fe-4S] centers, respectively. The complexity of the structures and reactions of these native enzymes, along with their large sizes and/or membrane associations, make it challenging to fully understand the crucial structural features responsible for the catalytic properties of these active sites. In this review, we summarize progress that has been made to better understand these heteronuclear metalloenzymes at the molecular level though study of the native enzymes along with insights gained from biomimetic models comprising either small molecules or proteins. Further understanding the reaction selectivity of these enzymes is discussed through comparisons of their similar heteronuclear active sites, and we offer outlook for further investigations.


Asunto(s)
Materiales Biomiméticos/química , Metaloproteínas/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Oxidorreductasas/química , Catálisis , Dominio Catalítico , Hemo/metabolismo , Modelos Moleculares , Oxidación-Reducción , Conformación Proteica
4.
J Biol Chem ; 295(31): 10610-10623, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32434930

RESUMEN

Marine cyanobacteria are infected by phages whose genomes encode ferredoxin (Fd) electron carriers. These Fds are thought to redirect the energy harvested from light to phage-encoded oxidoreductases that enhance viral fitness, but it is unclear how the biophysical properties and partner specificities of phage Fds relate to those of photosynthetic organisms. Here, results of a bioinformatics analysis using a sequence similarity network revealed that phage Fds are most closely related to cyanobacterial Fds that transfer electrons from photosystems to oxidoreductases involved in nutrient assimilation. Structural analysis of myovirus P-SSM2 Fd (pssm2-Fd), which infects the cyanobacterium Prochlorococcus marinus, revealed high levels of similarity to cyanobacterial Fds (root mean square deviations of ≤0.5 Å). Additionally, pssm2-Fd exhibited a low midpoint reduction potential (-336 mV versus a standard hydrogen electrode), similar to other photosynthetic Fds, although it had lower thermostability (Tm = 28 °C) than did many other Fds. When expressed in an Escherichia coli strain deficient in sulfite assimilation, pssm2-Fd complemented bacterial growth when coexpressed with a P. marinus sulfite reductase, revealing that pssm2-Fd can transfer electrons to a host protein involved in nutrient assimilation. The high levels of structural similarity with cyanobacterial Fds and reactivity with a host sulfite reductase suggest that phage Fds evolved to transfer electrons to cyanobacterially encoded oxidoreductases.


Asunto(s)
Proteínas Bacterianas , Bacteriófagos/enzimología , Ferredoxinas , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Prochlorococcus , Proteínas Virales , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Ferredoxinas/química , Ferredoxinas/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Prochlorococcus/enzimología , Prochlorococcus/virología , Proteínas Virales/química , Proteínas Virales/metabolismo
5.
J Am Chem Soc ; 143(1): 335-348, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33372786

RESUMEN

Catalysis by canonical radical S-adenosyl-l-methionine (SAM) enzymes involves electron transfer (ET) from [4Fe-4S]+ to SAM, generating an R3S0 radical that undergoes regioselective homolytic reductive cleavage of the S-C5' bond to generate the 5'-dAdo· radical. However, cryogenic photoinduced S-C bond cleavage has regioselectively yielded either 5'-dAdo· or ·CH3, and indeed, each of the three SAM S-C bonds can be regioselectively cleaved in an RS enzyme. This diversity highlights a longstanding central question: what controls regioselective homolytic S-C bond cleavage upon SAM reduction? We here provide an unexpected answer, founded on our observation that photoinduced S-C bond cleavage in multiple canonical RS enzymes reveals two enzyme classes: in one, photolysis forms 5'-dAdo·, and in another it forms ·CH3. The identity of the cleaved S-C bond correlates with SAM ribose conformation but not with positioning and orientation of the sulfonium center relative to the [4Fe-4S] cluster. We have recognized the reduced-SAM R3S0 radical is a (2E) state with its antibonding unpaired electron in an orbital doublet, which renders R3S0 Jahn-Teller (JT)-active and therefore subject to vibronically induced distortion. Active-site forces induce a JT distortion that localizes the odd electron in a single priority S-C antibond, which undergoes regioselective cleavage. In photolytic cleavage those forces act through control of the ribose conformation and are transmitted to the sulfur via the S-C5' bond, but during catalysis thermally induced conformational changes that enable ET from a cluster iron generate dominant additional forces that specifically select S-C5' for cleavage. This motion also can explain how 5'-dAdo· subsequently forms the organometallic intermediate Ω.


Asunto(s)
Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , S-Adenosilmetionina/química , Proteínas Bacterianas/química , Proteínas Bacterianas/efectos de la radiación , Biocatálisis , Dominio Catalítico , Clostridium acetobutylicum/enzimología , Teoría Funcional de la Densidad , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/efectos de la radiación , Luz , Modelos Químicos , Estructura Molecular , Oxidación-Reducción/efectos de la radiación , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/efectos de la radiación , Fotólisis , S-Adenosilmetionina/efectos de la radiación , Thermotoga maritima/enzimología
6.
Nature ; 520(7549): 706-9, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25642962

RESUMEN

The six-electron reduction of sulfite to sulfide is the pivot point of the biogeochemical cycle of the element sulfur. The octahaem cytochrome c MccA (also known as SirA) catalyses this reaction for dissimilatory sulfite utilization by various bacteria. It is distinct from known sulfite reductases because it has a substantially higher catalytic activity and a relatively low reactivity towards nitrite. The mechanistic reasons for the increased efficiency of MccA remain to be elucidated. Here we show that anoxically purified MccA exhibited a 2- to 5.5-fold higher specific sulfite reductase activity than the enzyme isolated under oxic conditions. We determined the three-dimensional structure of MccA to 2.2 Å resolution by single-wavelength anomalous dispersion. We find a homotrimer with an unprecedented fold and haem arrangement, as well as a haem bound to a CX15CH motif. The heterobimetallic active-site haem 2 has a Cu(I) ion juxtaposed to a haem c at a Fe-Cu distance of 4.4 Å. While the combination of metals is reminiscent of respiratory haem-copper oxidases, the oxidation-labile Cu(I) centre of MccA did not seem to undergo a redox transition during catalysis. Intact MccA tightly bound SO2 at haem 2, a dehydration product of the substrate sulfite that was partially turned over due to photoreduction by X-ray irradiation, yielding the reaction intermediate SO. Our data show the biometal copper in a new context and function and provide a chemical rationale for the comparatively high catalytic activity of MccA.


Asunto(s)
Proteínas Bacterianas/química , Cobre/metabolismo , Hemo/análogos & derivados , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Wolinella/enzimología , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Cisteína/análogos & derivados , Cisteína/metabolismo , Hemo/metabolismo , Modelos Moleculares , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/aislamiento & purificación , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Sulfitos/metabolismo , Dióxido de Azufre/metabolismo
7.
Biochemistry ; 59(51): 4793-4798, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33326741

RESUMEN

Low G+C Gram-positive Firmicutes, such as the clinically important pathogens Staphylococcus aureus and Bacillus cereus, use the low-molecular weight thiol bacillithiol (BSH) as a defense mechanism to buffer the intracellular redox environment and counteract oxidative stress encountered by human neutrophils during infections. The protein YpdA has recently been shown to function as an essential NADPH-dependent reductase of oxidized bacillithiol disulfide (BSSB) resulting from stress responses and is crucial for maintaining the reduced pool of BSH and cellular redox balance. In this work, we present the first crystallographic structures of YpdAs, namely, those from S. aureus and B. cereus. Our analyses reveal a uniquely organized biological tetramer; however, the structure of the monomeric subunit is highly similar to those of other flavoprotein disulfide reductases. The absence of a redox active cysteine in the vicinity of the FAD isoalloxazine ring implies a new direct disulfide reduction mechanism, which is backed by the presence of a potentially gated channel, serving as a putative binding site for BSSB in the proximity of the FAD cofactor. We also report enzymatic activities for both YpdAs, which along with the structures presented in this work provide important structural and functional insight into a new class of FAD-containing NADPH-dependent oxidoreductases, related to the emerging fight against pathogenic bacteria.


Asunto(s)
Proteínas Bacterianas/química , Cisteína/análogos & derivados , Flavina-Adenina Dinucleótido/química , Glucosamina/análogos & derivados , NADP/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Bacillus cereus/enzimología , Cristalografía por Rayos X , Cisteína/química , Glucosamina/química , Oxidación-Reducción , Estructura Cuaternaria de Proteína , Staphylococcus aureus/enzimología
8.
J Biol Chem ; 294(49): 18820-18835, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31685660

RESUMEN

In the endoplasmic reticulum (ER), ER oxidoreductin 1 (ERO1) catalyzes intramolecular disulfide-bond formation within its substrates in coordination with protein-disulfide isomerase (PDI) and related enzymes. However, the molecular mechanisms that regulate the ERO1-PDI system in plants are unknown. Reduction of the regulatory disulfide bonds of the ERO1 from soybean, GmERO1a, is catalyzed by enzymes in five classes of PDI family proteins. Here, using recombinant proteins, vacuum-ultraviolet circular dichroism spectroscopy, biochemical and protein refolding assays, and quantitative immunoblotting, we found that GmERO1a activity is regulated by reduction of intramolecular disulfide bonds involving Cys-121 and Cys-146, which are located in a disordered region, similarly to their locations in human ERO1. Moreover, a GmERO1a variant in which Cys-121 and Cys-146 were replaced with Ala residues exhibited hyperactive oxidation. Soybean PDI family proteins differed in their ability to regulate GmERO1a. Unlike yeast and human ERO1s, for which PDI is the preferred substrate, GmERO1a directly transferred disulfide bonds to the specific active center of members of five classes of PDI family proteins. Of these proteins, GmPDIS-1, GmPDIS-2, GmPDIM, and GmPDIL7 (which are group II PDI family proteins) failed to catalyze effective oxidative folding of substrate RNase A when there was an unregulated supply of disulfide bonds from the C121A/C146A hyperactive mutant GmERO1a, because of its low disulfide-bond isomerization activity. We conclude that regulation of plant ERO1 activity is particularly important for effective oxidative protein folding by group II PDI family proteins.


Asunto(s)
Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Oxidación-Reducción , Proteína Disulfuro Isomerasas/química , Proteína Disulfuro Isomerasas/metabolismo , Pliegue de Proteína , Isoformas de Proteínas/metabolismo
9.
J Am Chem Soc ; 142(33): 14295-14306, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32787249

RESUMEN

Sulfide quinone oxidoreductase (SQOR) catalyzes the first step in sulfide clearance, coupling H2S oxidation to coenzyme Q reduction. Recent structures of human SQOR revealed a sulfur atom bridging the SQOR active site cysteines in a trisulfide configuration. Here, we assessed the importance of this cofactor using kinetic, crystallographic, and computational modeling approaches. Cyanolysis of SQOR proceeds via formation of an intense charge transfer complex that subsequently decays to eliminate thiocyanate. We captured a disulfanyl-methanimido thioate intermediate in the SQOR crystal structure, revealing how cyanolysis leads to reversible loss of SQOR activity that is restored in the presence of sulfide. Computational modeling and MD simulations revealed an ∼105-fold rate enhancement for nucleophilic addition of sulfide into the trisulfide versus a disulfide cofactor. The cysteine trisulfide in SQOR is thus critical for activity and provides a significant catalytic advantage over a cysteine disulfide.


Asunto(s)
Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Sulfuros/metabolismo , Cristalografía por Rayos X , Humanos , Cinética , Modelos Moleculares , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Sulfuros/química
10.
Biochem Biophys Res Commun ; 533(3): 481-485, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-32977945

RESUMEN

N-glycosylation is an important protein modification that generally occurs at the Asn residue in an Asn-X-Ser/Thr sequon. Ero1 and its homologs play key roles in catalyzing the oxidative folding in the endoplasmic reticulum (ER). Recently, we found that Arabidopsis (Arabidopsis thaliana) ERO1 and AtERO2 displayed different characteristics in catalyzing oxidative protein folding in the ER. All known Ero1s are glycosylated proteins, including AtERO1 and AtERO2 that were analyzed when they were transiently translated in mammalian cells. However, the exact N-glycosylation sites on AtERO1 and AtERO2 remains to be determined. In this work, using a plant transient expression system, we identified the N-glycosylation sites on both AtERO1 and AtERO2. We found that AtERO1 has one N-glycosylation site, while AtERO2 contains two, all in the N-X-S/T sequons. Interestingly, we found that Ero1 homologs from human, rice, soybean and Arabidopsis, all have a conserved N-glycosylation site near the inner active site that reduces molecular oxygen and provides the oxidizing equivalents. The identification of N-glycosylation sites on AtERO1/2 proteins will help understand the function of N-glycosylation not only in AtERO1/2, but also in other Ero1 homologs.


Asunto(s)
Proteínas de Arabidopsis/química , Glicoproteínas de Membrana/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Secuencia de Aminoácidos , Arabidopsis/metabolismo , Proteínas de Arabidopsis/metabolismo , Secuencia Conservada , Glicosilación , Glicoproteínas de Membrana/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Alineación de Secuencia , Análisis de Secuencia de Proteína
11.
Nat Chem Biol ; 14(11): 995-1004, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30177848

RESUMEN

Cysteine sulfinic acid or S-sulfinylation is an oxidative post-translational modification (OxiPTM) that is known to be involved in redox-dependent regulation of protein function but has been historically difficult to analyze biochemically. To facilitate the detection of S-sulfinylated proteins, we demonstrate that a clickable, electrophilic diazene probe (DiaAlk) enables capture and site-centric proteomic analysis of this OxiPTM. Using this workflow, we revealed a striking difference between sulfenic acid modification (S-sulfenylation) and the S-sulfinylation dynamic response to oxidative stress, which is indicative of different roles for these OxiPTMs in redox regulation. We also identified >55 heretofore-unknown protein substrates of the cysteine sulfinic acid reductase sulfiredoxin, extending its function well beyond those of 2-cysteine peroxiredoxins (2-Cys PRDX1-4) and offering new insights into the role of this unique oxidoreductase as a central mediator of reactive oxygen species-associated diseases, particularly cancer. DiaAlk therefore provides a novel tool to profile S-sulfinylated proteins and study their regulatory mechanisms in cells.


Asunto(s)
Cisteína/análogos & derivados , Nitrógeno/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Procesamiento Proteico-Postraduccional , Proteómica/métodos , Células A549 , Cisteína/química , Epítopos/química , Células HEK293 , Células HeLa , Humanos , Oxidación-Reducción , Estrés Oxidativo , Péptidos/química , Peroxirredoxinas , Especies Reactivas de Oxígeno/química , Proteínas Recombinantes/química , Ácidos Sulfénicos
12.
Chem Rev ; 118(3): 1253-1337, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29112440

RESUMEN

Signaling by H2S is proposed to occur via persulfidation, a posttranslational modification of cysteine residues (RSH) to persulfides (RSSH). Persulfidation provides a framework for understanding the physiological and pharmacological effects of H2S. Due to the inherent instability of persulfides, their chemistry is understudied. In this review, we discuss the biologically relevant chemistry of H2S and the enzymatic routes for its production and oxidation. We cover the chemical biology of persulfides and the chemical probes for detecting them. We conclude by discussing the roles ascribed to protein persulfidation in cell signaling pathways.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Sulfurtransferasas/metabolismo , Animales , Cistationina betasintasa/química , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/química , Cistationina gamma-Liasa/metabolismo , Humanos , Sulfuro de Hidrógeno/análisis , Sulfuro de Hidrógeno/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sulfito-Oxidasa/química , Sulfito-Oxidasa/metabolismo
13.
Biochem J ; 476(12): 1805-1815, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31167903

RESUMEN

In addition to nitric oxide (NO) synthases, molybdenum-dependent enzymes have been reported to reduce nitrite to produce NO. Here, we report the stoichiometric reduction in nitrite to NO by human sulfite oxidase (SO), a mitochondrial intermembrane space enzyme primarily involved in cysteine catabolism. Kinetic and spectroscopic studies provide evidence for direct nitrite coordination at the molybdenum center followed by an inner shell electron transfer mechanism. In the presence of the physiological electron acceptor cytochrome c, we were able to close the catalytic cycle of sulfite-dependent nitrite reduction thus leading to steady-state NO synthesis, a finding that strongly supports a physiological relevance of SO-dependent NO formation. By engineering SO variants with reduced intramolecular electron transfer rate, we were able to increase NO generation efficacy by one order of magnitude, providing a mechanistic tool to tune NO synthesis by SO.


Asunto(s)
Proteínas Mitocondriales/química , Óxido Nítrico/química , Nitritos/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Humanos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Óxido Nítrico/biosíntesis , Óxido Nítrico/genética , Nitritos/metabolismo , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo
14.
Int J Mol Sci ; 21(24)2020 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-33321986

RESUMEN

Erv1 (EC number 1.8.3.2) is an essential mitochondrial enzyme catalyzing protein import and oxidative folding in the mitochondrial intermembrane space. Erv1 has both oxidase and cytochrome c reductase activities. While both Erv1 and cytochrome c were reported to be membrane associated in mitochondria, it is unknown how the mitochondrial membrane environment may affect the function of Erv1. Here, in this study, we used liposomes to mimic the mitochondrial membrane and investigated the effect of liposomes and cardiolipin on the folding and function of yeast Erv1. Enzyme kinetics of both the oxidase and cytochrome c reductase activity of Erv1 were studied using oxygen consumption analysis and spectroscopic methods. Our results showed that the presence of liposomes has mild impacts on Erv1 oxidase activity, but significantly inhibited the catalytic efficiency of Erv1 cytochrome c reductase activity in a cardiolipin-dependent manner. Taken together, the results of this study provide important insights into the function of Erv1 in the mitochondria, suggesting that molecular oxygen is a better substrate than cytochrome c for Erv1 in the yeast mitochondria.


Asunto(s)
Cardiolipinas/química , Liposomas/química , Proteínas Mitocondriales/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Pliegue de Proteína , Proteínas de Saccharomyces cerevisiae/química , Cardiolipinas/farmacología , Cinética , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/metabolismo
15.
J Biol Chem ; 293(46): 17997-18009, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30242128

RESUMEN

Increased light scattering in the eye lens due to aggregation of the long-lived lens proteins, crystallins, is the cause of cataract disease. Several mutations in the gene encoding human γD-crystallin (HγD) cause misfolding and aggregation. Cataract-associated substitutions at Trp42 cause the protein to aggregate in vitro from a partially unfolded intermediate locked by an internal disulfide bridge, and proteomic evidence suggests a similar aggregation precursor is involved in age-onset cataract. Surprisingly, WT HγD can promote aggregation of the W42Q variant while itself remaining soluble. Here, a search for a biochemical mechanism for this interaction has revealed a previously unknown oxidoreductase activity in HγD. Using in vitro oxidation, mutational analysis, cysteine labeling, and MS, we have assigned this activity to a redox-active internal disulfide bond that is dynamically exchanged among HγD molecules. The W42Q variant acts as a disulfide sink, reducing oxidized WT and forming a distinct internal disulfide that kinetically traps the aggregation-prone intermediate. Our findings suggest a redox "hot potato" competition among WT and mutant or modified polypeptides wherein variants with the lowest kinetic stability are trapped in aggregation-prone intermediate states upon accepting disulfides from more stable variants. Such reactions may occur in other long-lived proteins that function in oxidizing environments. In these cases, aggregation may be forestalled by inhibiting disulfide flow toward mutant or damaged polypeptides.


Asunto(s)
Disulfuros/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , gamma-Cristalinas/metabolismo , Sustitución de Aminoácidos , Cisteína/química , Disulfuros/química , Escherichia coli , Humanos , Mutagénesis Sitio-Dirigida , Mutación , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Dominios Proteicos , Multimerización de Proteína , Desplegamiento Proteico , Proteómica , gamma-Cristalinas/química , gamma-Cristalinas/genética
16.
J Biol Chem ; 293(8): 2640-2649, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29305423

RESUMEN

Transglutaminase 2 (TG2) is a ubiquitously expressed, intracellular as well as extracellular protein with multiple modes of post-translational regulation, including an allosteric disulfide bond between Cys-370-Cys-371 that renders the enzyme inactive in the extracellular matrix. Although recent studies have established that extracellular TG2 is switched "on" by the redox cofactor protein thioredoxin-1 (TRX), it is unclear how TG2 is switched "off." Here, we demonstrate that TG2 oxidation by small-molecule biological oxidants, including glutathione, cystine, and hydrogen peroxide, is unlikely to be the inactivation mechanism. Instead, endoplasmic reticulum (ER)-resident protein 57 (ERp57), a protein in the ER that promotes folding of nascent proteins and is also present in the extracellular environment, has the cellular and biochemical characteristics for inactivating TG2. We found that ERp57 colocalizes with extracellular TG2 in cultured human umbilical vein endothelial cells (HUVECs). ERp57 oxidized TG2 with a rate constant that was 400-2000-fold higher than those of the aforementioned small molecule oxidants. Moreover, its specificity for TG2 was also markedly higher than those of other secreted redox proteins, including protein disulfide isomerase (PDI), ERp72, TRX, and quiescin sulfhydryl oxidase 1 (QSOX1). Lastly, siRNA-mediated ERp57 knockdown in HUVECs increased TG2-catalyzed transamidation in the extracellular environment. We conclude that, to the best of our knowledge, the disulfide bond switch in human TG2 represents the first example of a post-translational redox regulatory mechanism that is reversibly and allosterically modulated by two distinct proteins (ERp57 and TRX).


Asunto(s)
Matriz Extracelular/enzimología , Proteínas de Unión al GTP/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/metabolismo , Procesamiento Proteico-Postraduccional , Transglutaminasas/antagonistas & inhibidores , Regulación Alostérica/efectos de los fármacos , Biocatálisis/efectos de los fármacos , Células Cultivadas , Cistina/metabolismo , Enzimas Inmovilizadas/antagonistas & inhibidores , Enzimas Inmovilizadas/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Glutatión/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Oxidantes/metabolismo , Oxidantes/farmacología , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/genética , Proteína Glutamina Gamma Glutamiltransferasa 2 , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Transglutaminasas/química , Transglutaminasas/genética , Transglutaminasas/metabolismo
17.
Biochem Biophys Res Commun ; 520(2): 347-352, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31604522

RESUMEN

Typical 2-cysteine peroxiredoxins (2-Cys Prxs) are critical peroxidase sensors and could be deactivated by the hyperoxidation under oxidative stress. In plants, 2-Cys Prxs present at a high level in chloroplasts and are repaired by Sulfiredoxin. Whereas many studies have explored the mechanism of Sulfiredoxin from Homo sapiens (HsSrx), the molecular mechanism of Sulfiredoxin in plants with unique photosynthesis remains unclear. Here we report the crystal structure of Sulfiredoxin from Arabidopsis thaliana (AtSrx), which displayed a typical ParB/Srx fold with an ATP bound at a conservative nucleotide binding motif GCHR. Both the ADP binding pocket and the putative AtSrx-AtPrxA interaction surface of AtSrx are more positively charged comparing to HsSrx, suggesting a robust mechanism of AtSrx. These features illustrate the unique mechanisms of AtSrx, which are vital for figure out the strategies of plants to cope with oxidation stress.


Asunto(s)
Proteínas de Arabidopsis/química , Arabidopsis/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Antioxidantes/química , Antioxidantes/metabolismo , Arabidopsis/química , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Arginina/química , Arginina/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Modelos Moleculares , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Peroxirredoxinas/metabolismo , Conformación Proteica , Pliegue de Proteína
18.
Nitric Oxide ; 89: 22-31, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31002874

RESUMEN

The oxygen-independent nitrate-nitrite-nitric oxide (NO) pathway is considered as a substantial source of NO in mammals. Dietary nitrate/nitrite are distributed throughout the body and reduced to NO by the action of various enzymes. The intermembrane spaced (IMS), molybdenum cofactor-dependent sulfite oxidase (SO) was shown to catalyze such a nitrite reduction. In this study we asked whether the primary function of SO - sulfite oxidation - and its novel function - nitrite reduction - impact each other. First, we utilized benzyl viologen as artificial electron donor to investigate steady state NO synthesis by SO and found fast (kcat = 14 s-1) nitrite reduction of SO full-length and its isolated molybdenum domain at pH 6.5. Next, we determined the impact of nitrite on pre-steady state kinetics in SO catalysis and identified nitrite as a pH-dependent inhibitor of SO reductive and oxidative half reaction. Finally, we report on the time-dependent formation of the paramagnetic Mo(V) species following nitrite reduction and demonstrate that sulfite inhibits nitrite reduction. In conclusion, we propose a pH-dependent reciprocal regulation of sulfite oxidation and nitrite reduction by each substrate, thus facilitating quick responses to hypoxia induced changes in the IMS, which may function in protecting the cell from reactive oxygen species production.


Asunto(s)
Proteínas Mitocondriales/química , Nitritos/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Sulfitos/química , Bencil Viológeno/química , Catálisis , Escherichia coli/genética , Hemo/química , Humanos , Concentración de Iones de Hidrógeno , Cinética , Oxidación-Reducción
19.
Proc Natl Acad Sci U S A ; 113(47): E7564-E7571, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27821734

RESUMEN

Recent studies have pointed to protein S-nitrosylation as a critical regulator of cellular redox homeostasis. For example, S-nitrosylation of peroxiredoxin-2 (Prx2), a peroxidase widely expressed in mammalian neurons, inhibits both enzymatic activity and protective function against oxidative stress. Here, using in vitro and in vivo approaches, we identify a role and reaction mechanism of the reductase sulfiredoxin (Srxn1) as an enzyme that denitrosylates (thus removing -SNO) from Prx2 in an ATP-dependent manner. Accordingly, by decreasing S-nitrosylated Prx2 (SNO-Prx2), overexpression of Srxn1 protects dopaminergic neural cells and human-induced pluripotent stem cell (hiPSC)-derived neurons from NO-induced hypersensitivity to oxidative stress. The pathophysiological relevance of this observation is suggested by our finding that SNO-Prx2 is dramatically increased in murine and human Parkinson's disease (PD) brains. Our findings therefore suggest that Srxn1 may represent a therapeutic target for neurodegenerative disorders such as PD that involve nitrosative/oxidative stress.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Enfermedad de Parkinson/metabolismo , Peroxirredoxinas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/citología , Humanos , Hidrólisis , Células Madre Pluripotentes Inducidas/citología , Ratones , Óxido Nítrico/química , Estrés Oxidativo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Peroxirredoxinas/química , Fosforilación
20.
Biochemistry ; 57(3): 258-266, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29064673

RESUMEN

During oxidative protein folding, disulfide bond formation is catalyzed by thiol oxidoreductases. Through dedicated relay pathways, the disulfide is generated in donor enzymes, passed to carrier enzymes, and subsequently delivered to target proteins. The eukaryotic disulfide donors are flavoenzymes, Ero1 in the endoplasmic reticulum and Erv1 in mitochondria. In prokaryotes, disulfide generation is coupled to quinone reduction, catalyzed by intramembrane donor enzymes, DsbB and VKOR. To catalyze de novo disulfide formation, these different disulfide donors show striking structural convergence at several levels. They share a four-helix bundle core structure at their active site, which contains a CXXC motif at a helical end. They have also evolved a flexible loop with shuttle cysteines to transfer electrons to the active site and relay the disulfide bond to the carrier enzymes. Studies of the prokaryotic VKOR, however, have stirred debate about whether the human homologue adopts the same topology with four transmembrane helices and uses the same electron-transfer mechanism. The controversies have recently been resolved by investigating the human VKOR structure and catalytic process in living cells with a mass spectrometry-based approach. Structural convergence between human VKOR and the disulfide donors is found to underlie cofactor reduction, disulfide generation, and electron transfer.


Asunto(s)
Evolución Molecular , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Compuestos de Sulfhidrilo/química , Vitamina K Epóxido Reductasas/química , Catálisis , Secuencia Conservada , Disulfuros/química , Humanos , Isomerismo , Pliegue de Proteína , Estructura Secundaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA