Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Bacteriol ; 203(17): e0015321, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34152832

RESUMEN

Mechanisms of disulfide bond formation in the human pathogen Streptococcus pyogenes are currently unknown. To date, no disulfide bond-forming thiol-disulfide oxidoreductase (TDOR) has been described and at least one disulfide bonded protein is known in S. pyogenes. This protein is the superantigen SpeA, which contains 3 cysteine residues (Cys 87, Cys90, and Cys98) and has a disulfide bond formed between Cys87 and Cys98. In this study, candidate TDORs were identified from the genome sequence of S. pyogenes MGAS8232. Using mutational and biochemical approaches, one of the candidate proteins, SpyM18_2037 (named here SdbA), was shown to be the catalyst that introduces the disulfide bond in SpeA. SpeA in the culture supernatant remained reduced when sdbA was inactivated and restored to the oxidized state when a functional copy of sdbA was returned to the sdbA-knockout mutant. SdbA has a typical C46XXC49 active site motif commonly found in TDORs. Site-directed mutagenesis experiments showed that the cysteines in the CXXC motif were required for the disulfide bond in SpeA to form. Interactions between SdbA and SpeA were examined using cysteine variant proteins. The results showed that SdbAC49A formed a mixed disulfide with SpeAC87A, suggesting that the N-terminal Cys46 of SdbA and the C-terminal Cys98 of SpeA participated in the initial reaction. SpeA oxidized by SdbA displayed biological activities suggesting that SpeA was properly folded following oxidation by SdbA. In conclusion, formation of the disulfide bond in SpeA is catalyzed by SdbA and the findings represent the first report of disulfide bond formation in S. pyogenes. IMPORTANCE Here, we reported the first example of disulfide bond formation in Streptococcus pyogenes. The results showed that a thiol-disulfide oxidoreductase, named SdbA, is responsible for introducing the disulfide bond in the superantigen SpeA. The cysteine residues in the CXXC motif of SdbA are needed for catalyzing the disulfide bond in SpeA. The disulfide bond in SpeA and neighboring amino acids form a disulfide loop that is conserved among many superantigens, including those from Staphylococcus aureus. SpeA and staphylococcal enterotoxins lacking the disulfide bond are biologically inactive. Thus, the discovery of the enzyme that catalyzes the disulfide bond in SpeA is important for understanding the biochemistry of SpeA production and presents a target for mitigating the virulence of S. pyogenes.


Asunto(s)
Proteínas Bacterianas/metabolismo , Disulfuros/metabolismo , Exotoxinas/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Streptococcus pyogenes/enzimología , Secuencias de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Biocatálisis , Dominio Catalítico , Disulfuros/química , Exotoxinas/genética , Proteínas de la Membrana/genética , Mutagénesis Sitio-Dirigida , Proteína Disulfuro Reductasa (Glutatión)/química , Proteína Disulfuro Reductasa (Glutatión)/genética , Streptococcus pyogenes/química , Streptococcus pyogenes/genética
2.
Int J Mol Sci ; 22(2)2021 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-33466919

RESUMEN

Redox (reduction-oxidation) reactions control many important biological processes in all organisms, both prokaryotes and eukaryotes. This reaction is usually accomplished by canonical disulphide-based pathways involving a donor enzyme that reduces the oxidised cysteine residues of a target protein, resulting in the cleavage of its disulphide bonds. Focusing on human vitamin K epoxide reductase (hVKORC1) as a target and on four redoxins (protein disulphide isomerase (PDI), endoplasmic reticulum oxidoreductase (ERp18), thioredoxin-related transmembrane protein 1 (Tmx1) and thioredoxin-related transmembrane protein 4 (Tmx4)) as the most probable reducers of VKORC1, a comparative in-silico analysis that concentrates on the similarity and divergence of redoxins in their sequence, secondary and tertiary structure, dynamics, intraprotein interactions and composition of the surface exposed to the target is provided. Similarly, hVKORC1 is analysed in its native state, where two pairs of cysteine residues are covalently linked, forming two disulphide bridges, as a target for Trx-fold proteins. Such analysis is used to derive the putative recognition/binding sites on each isolated protein, and PDI is suggested as the most probable hVKORC1 partner. By probing the alternative orientation of PDI with respect to hVKORC1, the functionally related noncovalent complex formed by hVKORC1 and PDI was found, which is proposed to be a first precursor to probe thiol-disulphide exchange reactions between PDI and hVKORC1.


Asunto(s)
Dominios Proteicos , Pliegue de Proteína , Tiorredoxinas/química , Vitamina K Epóxido Reductasas/química , Algoritmos , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Simulación de Dinámica Molecular , Oxidación-Reducción , Proteína Disulfuro Reductasa (Glutatión)/química , Proteína Disulfuro Reductasa (Glutatión)/genética , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Proteína Disulfuro Isomerasas/química , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Homología de Secuencia de Aminoácido , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Vitamina K Epóxido Reductasas/genética , Vitamina K Epóxido Reductasas/metabolismo
3.
J Biomol NMR ; 74(10-11): 595-611, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32761504

RESUMEN

The presence of suitable cavities or pockets on protein structures is a general criterion for a therapeutic target protein to be classified as 'druggable'. Many disease-related proteins that function solely through protein-protein interactions lack such pockets, making development of inhibitors by traditional small-molecule structure-based design methods much more challenging. The 22 kDa bacterial thiol oxidoreductase enzyme, DsbA, from the gram-negative bacterium Burkholderia pseudomallei (BpsDsbA) is an example of one such target. The crystal structure of oxidized BpsDsbA lacks well-defined surface pockets. BpsDsbA is required for the correct folding of numerous virulence factors in B. pseudomallei, and genetic deletion of dsbA significantly attenuates B. pseudomallei virulence in murine infection models. Therefore, BpsDsbA is potentially an attractive drug target. Herein we report the identification of a small molecule binding site adjacent to the catalytic site of oxidized BpsDsbA. 1HN CPMG relaxation dispersion NMR measurements suggest that the binding site is formed transiently through protein dynamics. Using fragment-based screening, we identified a small molecule that binds at this site with an estimated affinity of KD ~ 500 µM. This fragment inhibits BpsDsbA enzymatic activity in vitro. The binding mode of this molecule has been characterized by NMR data-driven docking using HADDOCK. These data provide a starting point towards the design of more potent small molecule inhibitors of BpsDsbA.


Asunto(s)
Resonancia Magnética Nuclear Biomolecular/métodos , Proteína Disulfuro Reductasa (Glutatión)/química , Animales , Sitios de Unión , Burkholderia pseudomallei/enzimología , Burkholderia pseudomallei/patogenicidad , Dominio Catalítico , Ligandos , Ratones , Oxidación-Reducción , Unión Proteica , Conformación Proteica , Proteína Disulfuro Reductasa (Glutatión)/genética , Relación Estructura-Actividad Cuantitativa , Proteínas Recombinantes , Bibliotecas de Moléculas Pequeñas/química , Solubilidad , Tiazoles/química
4.
Protein Sci ; 28(1): 30-40, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30341785

RESUMEN

The formation of a disulfide bond is a critical step in the folding of numerous secretory and membrane proteins and catalyzed in vivo. A variety of mechanisms and protein structures have evolved to catalyze oxidative protein folding. Those enzymes that directly interact with a folding protein to accelerate its oxidative folding are mostly thiol-disulfide oxidoreductases that belong to the thioredoxin superfamily. The enzymes of this class often use a CXXC active-site motif embedded in their thioredoxin-like fold to promote formation, isomerization, and reduction of a disulfide bond in their target proteins. Over the past decade or so, an increasing number of substrates of the thiol-disulfide oxidoreductases that are present in the ER of mammalian cells have been discovered, revealing that the enzymes play unexpectedly diverse physiological functions. However, functions of some of these enzymes still remain unclear due to the lack of information on their substrates. Here, we review the methods used by researchers to identify the substrates of these enzymes and provide data that show the importance of using trichloroacetic acid in sample preparation for the substrate identification, hoping to aid future studies. We particularly focus on successful studies that have uncovered physiological substrates and functions of the enzymes in the periplasm of Gram-negative bacteria and the endoplasmic reticulum of mammalian cells. Similar approaches should be applicable to enzymes in other cellular compartments or in other organisms.


Asunto(s)
Retículo Endoplásmico/enzimología , Proteína Disulfuro Reductasa (Glutatión)/química , Pliegue de Proteína , Tiorredoxinas/química , Animales , Humanos , Oxidación-Reducción , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Especificidad por Sustrato , Tiorredoxinas/metabolismo
5.
Curr Opin Struct Biol ; 53: 140-150, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30223251

RESUMEN

Society has benefitted greatly from the use of antibiotics. Unfortunately, the misuse of these valuable molecules has resulted in increased levels of antibiotic resistance, a major global and public health issue. This resistance and the reliance on a small number of biological targets for the development of antibiotics emphasizes the need for new targets. A critical aspect guiding the development of new antimicrobials through a rational structure-guided approach is to understand the molecular structures of specific biological targets of interest. Here we give an overview of the structures of bacterial virulence enzyme targets involved in protein folding, peptidoglycan biosynthesis and cell wall modification. These include enzymes of the thiol-disulphide oxidoreductase pathway (DSB enzymes), peptidyl-proly cis/trans isomerases (Mips), enzymes from the Mur pathway and enzymes involved in lipopolysaccharide modification (EptA and ArnT). We also present progress towards inhibitor design of these targets for the development of novel anti-virulence therapeutic agents.


Asunto(s)
Antibacterianos/farmacología , Bacterias , Proteínas Bacterianas , Factores de Virulencia , Bacterias/enzimología , Bacterias/metabolismo , Infecciones Bacterianas/tratamiento farmacológico , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Diseño de Fármacos , Hexosiltransferasas/metabolismo , Humanos , Isomerasa de Peptidilprolil/química , Isomerasa de Peptidilprolil/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteína Disulfuro Reductasa (Glutatión)/química , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Dominios Proteicos , Estructura Cuaternaria de Proteína , Factores de Virulencia/química , Factores de Virulencia/metabolismo
6.
J Bacteriol ; 200(9)2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29440253

RESUMEN

The actinobacterium Corynebacterium matruchotii has been implicated in nucleation of oral microbial consortia leading to biofilm formation. Due to the lack of genetic tools, little is known about basic cellular processes, including protein secretion and folding, in this organism. We report here a survey of the C. matruchotii genome, which encodes a large number of exported proteins containing paired cysteine residues, and identified an oxidoreductase that is highly homologous to the Corynebacterium diphtheriae thiol-disulfide oxidoreductase MdbA (MdbACd). Crystallization studies uncovered that the 1.2-Å resolution structure of C. matruchotii MdbA (MdbACm) possesses two conserved features found in actinobacterial MdbA enzymes, a thioredoxin-like fold and an extended α-helical domain. By reconstituting the disulfide bond-forming machine in vitro, we demonstrated that MdbACm catalyzes disulfide bond formation within the actinobacterial pilin FimA. A new gene deletion method supported that mdbA is essential in C. matruchotii Remarkably, heterologous expression of MdbACm in the C. diphtheriae ΔmdbA mutant rescued its known defects in cell growth and morphology, toxin production, and pilus assembly, and this thiol-disulfide oxidoreductase activity required the catalytic motif CXXC. Altogether, the results suggest that MdbACm is a major thiol-disulfide oxidoreductase, which likely mediates posttranslocational protein folding in C. matruchotii by a mechanism that is conserved in ActinobacteriaIMPORTANCE The actinobacterium Corynebacterium matruchotii has been implicated in the development of oral biofilms or dental plaque; however, little is known about the basic cellular processes in this organism. We report here a high-resolution structure of a C. matruchotii oxidoreductase that is highly homologous to the Corynebacterium diphtheriae thiol-disulfide oxidoreductase MdbA. By biochemical analysis, we demonstrated that C. matruchotii MdbA catalyzes disulfide bond formation in vitro Furthermore, a new gene deletion method revealed that deletion of mdbA is lethal in C. matruchotii Remarkably, C. matruchotii MdbA can replace C. diphtheriae MdbA to maintain normal cell growth and morphology, toxin production, and pilus assembly. Overall, our studies support the hypothesis that C. matruchotii utilizes MdbA as a major oxidoreductase to catalyze oxidative protein folding.


Asunto(s)
Proteínas Bacterianas/química , Corynebacterium/enzimología , Corynebacterium/genética , Proteína Disulfuro Reductasa (Glutatión)/química , Proteínas Bacterianas/genética , Biopelículas , Catálisis , Corynebacterium diphtheriae/enzimología , Corynebacterium diphtheriae/genética , Disulfuros/química , Proteínas Fimbrias/química , Proteínas Fimbrias/genética , Eliminación de Gen , Genoma Bacteriano , Modelos Moleculares , Oxidación-Reducción , Oxidorreductasas/química , Oxidorreductasas/genética , Proteína Disulfuro Reductasa (Glutatión)/genética
7.
Antioxid Redox Signal ; 28(4): 296-310, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28899103

RESUMEN

AIMS: Living cells employ thioredoxin and glutaredoxin disulfide oxido-reductases to protect thiol groups in intracellular proteins. FrnE protein of Deinococcus radiodurans (drFrnE) is a disulfide oxido-reductase that is induced in response to Cd2+ exposure and is involved in cadmium and radiation tolerance. The aim of this study is to probe structure, function, and cellular localization of FrnE class of proteins. RESULTS: Here, we show drFrnE as a novel cytoplasmic oxido-reductase that could be functional in eubacteria under conditions where thioredoxin/glutaredoxin systems are inhibited or absent. Crystal structure analysis of drFrnE reveals thioredoxin fold with an alpha helical insertion domain and a unique, flexible, and functionally important C-terminal tail. The C-tail harbors a novel 239-CX4C-244 motif that interacts with the active site 22-CXXC-25 motif. Crystal structures with different active site redox states, including mixed disulfide (Cys22-Cys244), are reported here. The biochemical data show that 239-CX4C-244 motif channels electrons to the active site cysteines. drFrnE is more stable in the oxidized form, compared with the reduced form, supporting its role as a disulfide reductase. Using bioinformatics analysis and fluorescence microscopy, we show cytoplasmic localization of drFrnE. We have found "true" orthologs of drFrnE in several eubacterial phyla and, interestingly, all these groups apparently lack a functional glutaredoxin system. Innovation and Conclusion: We show that drFrnE represents a new class of hitherto unknown intracellular oxido-reductases that are abundantly present in eubacteria. Unlike other well-known oxido-reductases, FrnE harbors an additional dithiol motif that acts as a conduit to channel electrons to the active site during catalytic turnover. Antioxid. Redox Signal. 28, 296-310.


Asunto(s)
Citoplasma/enzimología , Deinococcus/química , Proteína Disulfuro Reductasa (Glutatión)/química , Secuencias de Aminoácidos/genética , Dominio Catalítico , Cristalografía por Rayos X , Citoplasma/química , Deinococcus/enzimología , Glutarredoxinas/química , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Proteína Disulfuro Reductasa (Glutatión)/genética , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Tiorredoxinas/química , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
8.
J Biol Chem ; 292(32): 13154-13167, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28634234

RESUMEN

In many Gram-negative bacteria, including Rhodobacter capsulatus, cytochrome c maturation (Ccm) is carried out by a membrane-integral machinery composed of nine proteins (CcmA to I). During this process, the periplasmic thiol-disulfide oxidoreductase DsbA is thought to catalyze the formation of a disulfide bond between the Cys residues at the apocytochrome c heme-binding site (CXXCH). Subsequently, a Ccm-specific thioreductive pathway involving CcmG and CcmH reduces this disulfide bond to allow covalent heme ligation. Currently, the sequence of thioredox reactions occurring between these components and apocytochrome c and the identity of their active Cys residues are unknown. In this work, we first investigated protein-protein interactions among the apocytochrome c, CcmG, and the heme-ligation components CcmF, CcmH, and CcmI. We found that they all interact with each other, forming a CcmFGHI-apocytochrome c complex. Using purified wild-type CcmG, CcmH, and apocytochrome c, as well as their respective Cys mutant variants, we determined the rates of thiol-disulfide exchange reactions between selected pairs of Cys residues from these proteins. We established that CcmG can efficiently reduce the disulfide bond of apocytochrome c and also resolve a mixed disulfide bond formed between apocytochrome c and CcmH. We further show that Cys-45 of CcmH and Cys-34 of apocytochrome c are most likely to form this mixed disulfide bond, which is consistent with the stereo-specificity of the heme-apocytochrome c ligation reaction. We conclude that CcmG confers efficiency, and CcmH ensures stereo-specificity during Ccm and present a comprehensive model for thioreduction reactions that lead to heme-apocytochrome c ligation.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Citocromos c/metabolismo , Modelos Biológicos , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Rhodobacter capsulatus/enzimología , Sustitución de Aminoácidos , Apoenzimas/química , Apoenzimas/metabolismo , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Cisteína/química , Cisteína/metabolismo , Cistina/química , Cistina/metabolismo , Citocromos c/química , Hemo/metabolismo , Mutación , Oxidación-Reducción , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteína Disulfuro Reductasa (Glutatión)/química , Proteína Disulfuro Reductasa (Glutatión)/genética , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Estereoisomerismo
9.
Acta Crystallogr D Struct Biol ; 72(Pt 10): 1100-1109, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27710931

RESUMEN

In bacteria, many Dsb-family proteins play diverse roles in the conversion between the oxidized and reduced states of cysteine residues of substrate proteins. Most Dsb enzymes catalyze disulfide formation in periplasmic or secreted substrate proteins. Recently, a DsbM protein has been found in a Gram-negative bacterium, and was characterized as a cytosolic Dsb member with the conserved CXXC motif on the basis of sequence homology to the Dsb-family proteins. The protein was implicated in the reduction of the cytoplasmic redox-sensor protein OxyR in Pseudomonas aeruginosa. Here, crystal structures of DsbM from P. aeruginosa are presented, revealing that it consists of a modified thioredoxin domain containing the CXXC motif and a lid domain surrounding the CXXC motif. In a glutathione-linked structure, a glutathione molecule is linked to the CXXC motif of DsbM and is bound in an elongated cavity region in the thioredoxin domain, which is also suited for substrate peptide binding. A striking structural similarity to a human glutathione S-transferase was found in the glutathione-binding pocket. Further, biochemical evidence is presented suggesting that DsbM is directly involved in the reduction of the disulfide of Cys199 and Cys208 in OxyR, resulting in the acceleration of OxyR reduction in the absence of reactive oxygen species stress. These findings may help to expand the understanding of the diverse roles of redox-related proteins that contain the CXXC motif.


Asunto(s)
Proteínas Bacterianas/química , Proteína Disulfuro Reductasa (Glutatión)/química , Pseudomonas aeruginosa/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Oxidación-Reducción , Conformación Proteica , Infecciones por Pseudomonas/microbiología , Alineación de Secuencia , Tiorredoxinas/química
10.
Biochemistry ; 55(2): 313-21, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26684934

RESUMEN

Glutaredoxins (GRXs) are thiol-disulfide oxidoreductases abundant in prokaryotes, although little is understood of these enzymes from the domain Archaea. The numerous characterized GRXs from the domain Bacteria utilize a diversity of low-molecular-weight thiols in addition to glutathione as reductants. We report here the biochemical and structural properties of a GRX-like protein named methanoredoxin (MRX) from Methanosarcina acetivorans of the domain Archaea. MRX utilizes coenzyme M (CoMSH) as reductant for insulin disulfide reductase activity, which adds to the diversity of thiol protectants in prokaryotes. Cell-free extracts of M. acetivorans displayed CoMS-SCoM reductase activity that complements the CoMSH-dependent activity of MRX. The crystal structure exhibits a classic thioredoxin-glutaredoxin fold comprising three α-helices surrounding four antiparallel ß-sheets. A pocket on the surface contains a CVWC motif, identifying the active site with architecture similar to GRXs. Although it is a monomer in solution, the crystal lattice has four monomers in a dimer of dimers arrangement. A cadmium ion is found within the active site of each monomer. Two such ions stabilize the N-terminal tails and dimer interfaces. Our modeling studies indicate that CoMSH and glutathione (GSH) bind to the active site of MRX similar to the binding of GSH in GRXs, although there are differences in the amino acid composition of the binding motifs. The results, combined with our bioinformatic analyses, show that MRX represents a class of GRX-like enzymes present in a diversity of methane-producing Archaea.


Asunto(s)
Proteínas Arqueales/metabolismo , Glutarredoxinas/metabolismo , Mesna/metabolismo , Methanosarcina/metabolismo , Proteínas Arqueales/química , Glutarredoxinas/química , Proteína Disulfuro Reductasa (Glutatión)/química , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Estructura Secundaria de Proteína
11.
Plant Physiol ; 170(2): 774-89, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26645455

RESUMEN

Most proteins produced in the endoplasmic reticulum (ER) of eukaryotic cells fold via disulfide formation (oxidative folding). Oxidative folding is catalyzed by protein disulfide isomerase (PDI) and PDI-related ER protein thiol disulfide oxidoreductases (ER oxidoreductases). In yeast and mammals, ER oxidoreductin-1s (Ero1s) supply oxidizing equivalent to the active centers of PDI. In this study, we expressed recombinant soybean Ero1 (GmERO1a) and found that GmERO1a oxidized multiple soybean ER oxidoreductases, in contrast to mammalian Ero1s having a high specificity for PDI. One of these ER oxidoreductases, GmPDIM, associated in vivo and in vitro with GmPDIL-2, was unable to be oxidized by GmERO1a. We therefore pursued the possible cooperative oxidative folding by GmPDIM, GmERO1a, and GmPDIL-2 in vitro and found that GmPDIL-2 synergistically accelerated oxidative refolding. In this process, GmERO1a preferentially oxidized the active center in the A': domain among the A: , A': , and B: domains of GmPDIM. A disulfide bond introduced into the active center of the A': domain of GmPDIM was shown to be transferred to the active center of the A: domain of GmPDIM and the A: domain of GmPDIM directly oxidized the active centers of both the A: or A': domain of GmPDIL-2. Therefore, we propose that the relay of an oxidizing equivalent from one ER oxidoreductase to another may play an essential role in cooperative oxidative folding by multiple ER oxidoreductases in plants.


Asunto(s)
Glycine max/enzimología , Oxidorreductasas/metabolismo , Proteína Disulfuro Reductasa (Glutatión)/química , Proteína Disulfuro Isomerasas/metabolismo , Catálisis , Disulfuros/metabolismo , Retículo Endoplásmico/enzimología , Oxidación-Reducción , Oxidorreductasas/genética , Proteína Disulfuro Reductasa (Glutatión)/genética , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Proteína Disulfuro Isomerasas/genética , Pliegue de Proteína , Proteínas Recombinantes , Glycine max/genética
12.
Mol Microbiol ; 98(6): 1037-50, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26294390

RESUMEN

The Gram-positive pathogen Corynebacterium diphtheriae exports through the Sec apparatus many extracellular proteins that include the key virulence factors diphtheria toxin and the adhesive pili. How these proteins attain their native conformations after translocation as unfolded precursors remains elusive. The fact that the majority of these exported proteins contain multiple cysteine residues and that several membrane-bound oxidoreductases are encoded in the corynebacterial genome suggests the existence of an oxidative protein-folding pathway in this organism. Here we show that the shaft pilin SpaA harbors a disulfide bond in vivo and alanine substitution of these cysteines abrogates SpaA polymerization and leads to the secretion of degraded SpaA peptides. We then identified a thiol-disulfide oxidoreductase (MdbA), whose structure exhibits a conserved thioredoxin-like domain with a CPHC active site. Remarkably, deletion of mdbA results in a severe temperature-sensitive cell division phenotype. This mutant also fails to assemble pilus structures and is greatly defective in toxin production. Consistent with these defects, the ΔmdbA mutant is attenuated in a guinea pig model of diphtheritic toxemia. Given its diverse cellular functions in cell division, pilus assembly and toxin production, we propose that MdbA is a component of the general oxidative folding machine in C. diphtheriae.


Asunto(s)
Corynebacterium diphtheriae/enzimología , Corynebacterium diphtheriae/patogenicidad , Proteínas Fimbrias/química , Proteínas Fimbrias/metabolismo , Proteína Disulfuro Reductasa (Glutatión)/aislamiento & purificación , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Corynebacterium diphtheriae/fisiología , Difteria/microbiología , Toxina Diftérica/biosíntesis , Toxina Diftérica/sangre , Fimbrias Bacterianas/química , Fimbrias Bacterianas/metabolismo , Cobayas , Viabilidad Microbiana , Mutación , Fenotipo , Proteína Disulfuro Reductasa (Glutatión)/química , Proteína Disulfuro Reductasa (Glutatión)/genética , Pliegue de Proteína , Toxemia/microbiología , Virulencia/genética
13.
J Biol Chem ; 290(35): 21393-405, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26170452

RESUMEN

Export of cell surface pilins in Gram-positive bacteria likely occurs by the translocation of unfolded precursor polypeptides; however, how the unfolded pilins gain their native conformation is presently unknown. Here, we present physiological studies to demonstrate that the FimA pilin of Actinomyces oris contains two disulfide bonds. Alanine substitution of cysteine residues forming the C-terminal disulfide bridge abrogates pilus assembly, in turn eliminating biofilm formation and polymicrobial interaction. Transposon mutagenesis of A. oris yielded a mutant defective in adherence to Streptococcus oralis, and revealed the essential role of a vitamin K epoxide reductase (VKOR) gene in pilus assembly. Targeted deletion of vkor results in the same defects, which are rescued by ectopic expression of VKOR, but not a mutant containing an alanine substitution in its conserved CXXC motif. Depletion of mdbA, which encodes a membrane-bound thiol-disulfide oxidoreductase, abrogates pilus assembly and alters cell morphology. Remarkably, overexpression of MdbA or a counterpart from Corynebacterium diphtheriae, rescues the Δvkor mutant. By alkylation assays, we demonstrate that VKOR is required for MdbA reoxidation. Furthermore, crystallographic studies reveal that A. oris MdbA harbors a thioredoxin-like fold with the conserved CXXC active site. Consistently, each MdbA enzyme catalyzes proper disulfide bond formation within FimA in vitro that requires the catalytic CXXC motif. Because the majority of signal peptide-containing proteins encoded by A. oris possess multiple Cys residues, we propose that MdbA and VKOR constitute a major folding machine for the secretome of this organism. This oxidative protein folding pathway may be a common feature in Actinobacteria.


Asunto(s)
Actinomyces/fisiología , Proteínas Bacterianas/metabolismo , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/metabolismo , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Vitamina K Epóxido Reductasas/metabolismo , Actinomyces/química , Actinomyces/citología , Actinomyces/genética , Actinomicosis/microbiología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Biopelículas/efectos de los fármacos , Cristalografía por Rayos X , Disulfuros/química , Disulfuros/metabolismo , Proteínas Fimbrias/química , Fimbrias Bacterianas/química , Eliminación de Gen , Humanos , Interacciones Microbianas , Modelos Moleculares , Conformación Proteica , Proteína Disulfuro Reductasa (Glutatión)/química , Proteína Disulfuro Reductasa (Glutatión)/genética , Pliegue de Proteína , Vitamina K Epóxido Reductasas/química , Vitamina K Epóxido Reductasas/genética
14.
BMC Microbiol ; 15: 135, 2015 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-26141380

RESUMEN

BACKGROUND: In the genome of H. pylori 26695, 149 proteins containing the CXXC motif characteristic of thioldisulfide oxidoreductases have been identified to date. However, only two of these proteins have a thioredoxin-like fold (i.e., HP0377 and HP0231) and are periplasm-located. We have previously shown that HP0231 is a dimeric oxidoreductase that catalyzes disulfide bond formation in the periplasm. Although HP0377 was originally described as DsbC homologue, its resolved structure and location of the hp0377 gene in the genome indicate that it is a counterpart of CcmG/DsbE. RESULTS: The present work shows that HP0377 is present in H. pylori cells only in a reduced form and that absence of the main periplasmic oxidase HP0231 influences its redox state. Our biochemical analysis indicates that HP0377 is a specific reductase, as it does not reduce insulin. However, it possesses disulfide isomerase activity, as it catalyzes the refolding of scrambled RNase. Additionally, although its standard redox potential is -176 mV, it is the first described CcmG protein having an acidic pKa of the N-terminal cysteine of the CXXC motif, similar to E. coli DsbA or E. coli DsbC. The CcmG proteins that play a role in a cytochrome c-maturation, both in system I and system II, are kept in the reduced form by an integral membrane protein DsbD or its analogue, CcdA. In H. pylori HP0377 is re-reduced by CcdA (HP0265); however in E. coli it remains in the oxidized state as it does not interact with E. coli DsbD. Our in vivo work also suggests that both HP0377, which plays a role in apocytochrome reduction, and HP0378, which is involved in heme transport and its ligation into apocytochrome, provide essential functions in H. pylori. CONCLUSIONS: The present data, in combination with the resolved three-dimensional structure of the HP0377, suggest that HP0377 is an unusual, multifunctional CcmG protein.


Asunto(s)
Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Secuencias de Aminoácidos , Proteínas Bacterianas/química , Genes Esenciales , Helicobacter pylori/química , Familia de Multigenes , Oxidación-Reducción , Proteína Disulfuro Reductasa (Glutatión)/química , Proteína Disulfuro Isomerasas/química , Replegamiento Proteico , Ribonucleasas/química
15.
PLoS One ; 9(9): e106247, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25181355

RESUMEN

BACKGROUND: Bacterial Dsb enzymes are involved in the oxidative folding of many proteins, through the formation of disulfide bonds between their cysteine residues. The Dsb protein network has been well characterized in cells of the model microorganism Escherichia coli. To gain insight into the functioning of the Dsb system in epsilon-Proteobacteria, where it plays an important role in the colonization process, we studied two homologs of the main Escherichia coli Dsb oxidase (EcDsbA) that are present in the cells of the enteric pathogen Campylobacter jejuni, the most frequently reported bacterial cause of human enteritis in the world. METHODS AND RESULTS: Phylogenetic analysis suggests the horizontal transfer of the epsilon-Proteobacterial DsbAs from a common ancestor to gamma-Proteobacteria, which then gave rise to the DsbL lineage. Phenotype and enzymatic assays suggest that the two C. jejuni DsbAs play different roles in bacterial cells and have divergent substrate spectra. CjDsbA1 is essential for the motility and autoagglutination phenotypes, while CjDsbA2 has no impact on those processes. CjDsbA1 plays a critical role in the oxidative folding that ensures the activity of alkaline phosphatase CjPhoX, whereas CjDsbA2 is crucial for the activity of arylsulfotransferase CjAstA, encoded within the dsbA2-dsbB-astA operon. CONCLUSIONS: Our results show that CjDsbA1 is the primary thiol-oxidoreductase affecting life processes associated with bacterial spread and host colonization, as well as ensuring the oxidative folding of particular protein substrates. In contrast, CjDsbA2 activity does not affect the same processes and so far its oxidative folding activity has been demonstrated for one substrate, arylsulfotransferase CjAstA. The results suggest the cooperation between CjDsbA2 and CjDsbB. In the case of the CjDsbA1, this cooperation is not exclusive and there is probably another protein to be identified in C. jejuni cells that acts to re-oxidize CjDsbA1. Altogether the data presented here constitute the considerable insight to the Epsilonproteobacterial Dsb systems, which have been poorly understood so far.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Biología Computacional/métodos , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Homología de Secuencia de Aminoácido , Aglutinación , Fosfatasa Alcalina/metabolismo , Arilsulfotransferasa/metabolismo , Campylobacter jejuni/enzimología , Escherichia coli/metabolismo , Prueba de Complementación Genética , Humanos , Insulina/metabolismo , Modelos Moleculares , Movimiento , Mutación/genética , Oxidación-Reducción , Filogenia , Agregado de Proteínas , Unión Proteica , Proteína Disulfuro Reductasa (Glutatión)/química , Pliegue de Proteína
17.
Antioxid Redox Signal ; 20(4): 606-17, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23901809

RESUMEN

AIMS: The intracellular pathogen Burkholderia pseudomallei causes the disease melioidosis, a major source of morbidity and mortality in southeast Asia and northern Australia. The need to develop novel antimicrobials is compounded by the absence of a licensed vaccine and the bacterium's resistance to multiple antibiotics. In a number of clinically relevant Gram-negative pathogens, DsbA is the primary disulfide oxidoreductase responsible for catalyzing the formation of disulfide bonds in secreted and membrane-associated proteins. In this study, a putative B. pseudomallei dsbA gene was evaluated functionally and structurally and its contribution to infection assessed. RESULTS: Biochemical studies confirmed the dsbA gene encodes a protein disulfide oxidoreductase. A dsbA deletion strain of B. pseudomallei was attenuated in both macrophages and a BALB/c mouse model of infection and displayed pleiotropic phenotypes that included defects in both secretion and motility. The 1.9 Å resolution crystal structure of BpsDsbA revealed differences from the classic member of this family Escherichia coli DsbA, in particular within the region surrounding the active site disulfide where EcDsbA engages with its partner protein E. coli DsbB, indicating that the interaction of BpsDsbA with its proposed partner BpsDsbB may be distinct from that of EcDsbA-EcDsbB. INNOVATION: This study has characterized BpsDsbA biochemically and structurally and determined that it is required for virulence of B. pseudomallei. CONCLUSION: These data establish a critical role for BpsDsbA in B. pseudomallei infection, which in combination with our structural characterization of BpsDsbA will facilitate the future development of rationally designed inhibitors against this drug-resistant organism.


Asunto(s)
Proteínas Bacterianas/química , Burkholderia pseudomallei/enzimología , Proteína Disulfuro Reductasa (Glutatión)/química , Animales , Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/patogenicidad , Dominio Catalítico , Línea Celular , Cristalografía por Rayos X , Femenino , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Oxidación-Reducción , Péptido Hidrolasas/metabolismo , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Estructura Secundaria de Proteína , Fosfolipasas de Tipo C/metabolismo , Virulencia
18.
Extremophiles ; 18(2): 219-28, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24306780

RESUMEN

Protein disulfide oxidoreductases (PDOs) are proteins involved in disulfide bond formation playing a crucial role in adaptation to extreme environment. This paper reports the functional and structural characterization of Sso1120, a PDO from the hyperthermophilic archaeon Sulfolobus solfataricus. The protein was expressed in Escherichia coli and purified to homogeneity. The functional characterization showed that the enzyme has reductase activity, as tested by insulin assay, but differently from the other PDOs, it does not present isomerase activity. In addition it is able to form a redox couple with the thioredoxin reductase that could be used in undiscovered pathways. The protein revealed a melting point of around 90 °C in CD spectroscopy-monitored thermal denaturation and high denaturant resistance. The X-ray crystallographic structure was solved at 1.80 Å resolution, showing differences with respect to other PDOs and an unexpected similarity with the N-terminal domain of the alkyl hydroperoxide reductase F component from Salmonella typhimurium. On the basis of the reported data and of bioinformatics and phylogenetic analyses, a possible involvement of this atypical PDO in a new antioxidant system of S. solfataricus has been proposed.


Asunto(s)
Proteínas Arqueales/química , Proteína Disulfuro Reductasa (Glutatión)/química , Sulfolobus solfataricus/enzimología , Secuencia de Aminoácidos , Proteínas Arqueales/genética , Proteínas Arqueales/metabolismo , Estabilidad de Enzimas , Datos de Secuencia Molecular , Oxidación-Reducción , Filogenia , Proteína Disulfuro Reductasa (Glutatión)/genética , Proteína Disulfuro Reductasa (Glutatión)/metabolismo , Estructura Terciaria de Proteína , Compuestos de Sulfhidrilo/metabolismo
19.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 5): 735-46, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23633582

RESUMEN

Maturation of cytochrome c is carried out in the bacterial periplasm, where specialized thiol-disulfide oxidoreductases provide the correct reduction of oxidized apocytochrome c before covalent haem attachment. HP0377 from Helicobacter pylori is a thioredoxin-fold protein that has been implicated as a component of system II for cytochrome c assembly and shows limited sequence similarity to Escherichia coli DsbC, a disulfide-bond isomerase. To better understand the role of HP0377, its crystal structures have been determined in both reduced and partially oxidized states, which are highly similar to each other. Sedimentation-equilibrium experiments indicate that HP0377 is monomeric in solution. HP0377 adopts a thioredoxin fold but shows distinctive variations as in other thioredoxin-like bacterial periplasmic proteins. The active site of HP0377 closely resembles that of E. coli DsbC. A reductase assay suggests that HP0377 may play a role as a reductase in the biogenesis of holocytochrome c553 (HP1227). Binding experiments indicate that it can form a covalent complex with HP0518, a putative L,D-transpeptidase with a catalytic cysteine residue, via a disulfide bond. Furthermore, physicochemical properties of HP0377 and its R86A variant have been determined. These results suggest that HP0377 may perform multiple functions as a reductase in H. pylori.


Asunto(s)
Proteínas Bacterianas/química , Helicobacter pylori/química , Proteína Disulfuro Reductasa (Glutatión)/química , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Cisteína/química , Citocromos c/metabolismo , Helicobacter pylori/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Periplasmáticas/química , Proteínas Periplasmáticas/metabolismo , Conformación Proteica , Proteína Disulfuro Reductasa (Glutatión)/metabolismo
20.
Biomol NMR Assign ; 7(1): 105-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22585086

RESUMEN

CcmG is a periplasmic, membrane-anchored protein widely distributed in a variety of species. In Escherichia coli, the CcmG protein always acts as a weak reductant in the electron transport chain during cytochrome c maturation (Ccm). Here we report (1)H, (15)N and (13)C backbone and side-chain resonance assignments of the reduced CcmG protein (residues 19-185, renumbered as 1-167) from E. coli. This work lays the essential basis for the further structural and functional analysis of reduced CcmG.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli , Resonancia Magnética Nuclear Biomolecular , Proteína Disulfuro Reductasa (Glutatión)/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...