Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 138(6): 1164-73, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19766568

RESUMEN

Most bacterial exported proteins cross the cytoplasmic membrane as unfolded polypeptides. However, little is known about how they fold during or after this process due to the difficulty in detecting folding intermediates. Here we identify cotranslational and posttranslational folding intermediates of a periplasmic protein in which the protein and DsbA, a periplasmic disulfide bond-forming enzyme, are covalently linked by a disulfide bond. The cotranslational mixed-disulfide intermediate is, upon further chain elongation, resolved, releasing the oxidized polypeptide, thus allowing us to follow the folding process. This analysis reveals that two cysteines that are joined to form a structural disulfide can play different roles during the folding reaction and that the mode of translocation (cotranslational verse posttranslational) can affect the folding process of a protein in the periplasm. The latter finding leads us to propose that the activity of the ribosome (translation) can modulate protein folding even in an extracytosolic compartment.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Fosfatasa Alcalina/metabolismo , Cisteína/metabolismo , Periplasma/metabolismo , Biosíntesis de Proteínas , Pliegue de Proteína
2.
Mol Microbiol ; 111(4): 918-937, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30556355

RESUMEN

In bacteria, disulfide bonds confer stability on many proteins exported to the cell envelope or beyond, including bacterial virulence factors. Thus, proteins involved in disulfide bond formation represent good targets for the development of inhibitors that can act as antibiotics or anti-virulence agents, resulting in the simultaneous inactivation of several types of virulence factors. Here, we present evidence that the disulfide bond forming enzymes, DsbB and VKOR, are required for Pseudomonas aeruginosa pathogenicity and Mycobacterium tuberculosis survival respectively. We also report the results of a HTS of 216,767 compounds tested against P. aeruginosa DsbB1 and M. tuberculosis VKOR using Escherichia coli cells. Since both P. aeruginosa DsbB1 and M. tuberculosis VKOR complement an E. coli dsbB knockout, we screened simultaneously for inhibitors of each complemented E. coli strain expressing a disulfide-bond sensitive ß-galactosidase reported previously. The properties of several inhibitors obtained from these screens suggest they are a starting point for chemical modifications with potential for future antibacterial development.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Disulfuros/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Mycobacterium tuberculosis/enzimología , Pseudomonas aeruginosa/enzimología , Animales , Proteínas Bacterianas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Ensayos Analíticos de Alto Rendimiento , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/patogenicidad , Pseudomonas aeruginosa/patogenicidad , Bibliotecas de Moléculas Pequeñas/farmacología , Virulencia , Factores de Virulencia/metabolismo
3.
J Bacteriol ; 200(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29784887

RESUMEN

Disulfide bonds influence the stability and activity of many proteins. In Escherichia coli, the DsbA and DsbB enzymes promote disulfide bond formation. Other bacteria, including the Actinobacteria, use instead of DsbB the enzyme vitamin K epoxide reductase (VKOR), whose gene is found either fused to or in the same operon as a dsbA-like gene. Mycobacterium tuberculosis and other Gram-positive actinobacteria secrete many proteins with even numbers of cysteines to the cell envelope. These organisms have predicted oxidoreductases and VKOR orthologs. These findings indicate that such bacteria likely form disulfide bonds in the cell envelope. The M. tuberculosisvkor gene complements an E. colidsbB deletion strain, restoring the oxidation of E. coli DsbA. While we have suggested that the dsbA gene linked to the vkor gene may express VKOR's partner in mycobacteria, others have suggested that two other extracytoplasmic oxidoreductases (DsbE or DsbF) may be catalysts of protein disulfide bond formation. However, there is no direct evidence for interactions of VKOR with either DsbA, DsbE, or DsbF. To identify the actual substrate of VKOR, we identified two additional predicted extracytoplasmic DsbA-like proteins using bioinformatics analysis of the M. tuberculosis genome. Using the five potential DsbAs, we attempted to reconstitute disulfide bond pathways in E. coli and in Mycobacterium smegmatis, a close relative of M. tuberculosis Our results show that only M. tuberculosis DsbA is oxidized by VKOR. Comparison of the properties of dsbA- and vkor-null mutants in M. smegmatis shows parallels to the properties of dsb mutations in E. coliIMPORTANCE Disulfide bond formation has a great impact on bacterial pathogenicity. Thus, disulfide-bond-forming proteins represent new targets for the development of antibacterials, since the inhibition of disulfide bond formation would result in the simultaneous loss of the activity of several classes of virulence factors. Here, we identified five candidate proteins encoded by the M. tuberculosis genome as possible substrates of the M. tuberculosis VKOR protein involved in disulfide bond formation. We then reconstituted the mycobacterial disulfide bond formation pathway in E. coli and showed that of the five candidates, only M. tuberculosis DsbA is efficiently oxidized by VKOR in E. coli We also present evidence for the involvement of VKOR in DsbA oxidation in M. smegmatis.


Asunto(s)
Proteínas Bacterianas/genética , Disulfuros/metabolismo , Mycobacterium tuberculosis/genética , Tiorredoxinas/metabolismo , Vitamina K Epóxido Reductasas/genética , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/patogenicidad , Oxidación-Reducción , Oxidorreductasas , Proteína Disulfuro Isomerasas/metabolismo , Vitamina K Epóxido Reductasas/metabolismo
4.
J Biol Chem ; 292(16): 6529-6541, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28232484

RESUMEN

Disulfide bonds contribute to protein stability, activity, and folding in a variety of proteins, including many involved in bacterial virulence such as toxins, adhesins, flagella, and pili, among others. Therefore, inhibitors of disulfide bond formation enzymes could have profound effects on pathogen virulence. In the Escherichia coli disulfide bond formation pathway, the periplasmic protein DsbA introduces disulfide bonds into substrates, and then the cytoplasmic membrane protein DsbB reoxidizes DsbA's cysteines regenerating its activity. Thus, DsbB generates a protein disulfide bond de novo by transferring electrons to the quinone pool. We previously identified an effective pyridazinone-related inhibitor of DsbB enzymes from several Gram-negative bacteria. To map the protein residues that are important for the interaction with this inhibitor, we randomly mutagenized by error-prone PCR the E. coli dsbB gene and selected dsbB mutants that confer resistance to this drug using two approaches. We characterized in vivo and in vitro some of these mutants that map to two areas in the structure of DsbB, one located between the two first transmembrane segments where the quinone ring binds and the other located in the second periplasmic loop of DsbB, which interacts with DsbA. In addition, we show that a mutant version of a protein involved in lipopolysaccharide assembly, lptD4213, is synthetically lethal with the deletion of dsbB as well as with DsbB inhibitors. This finding suggests that drugs decreasing LptD assembly may be synthetically lethal with inhibitors of the Dsb pathway, potentiating the antibiotic effects.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Proteínas Bacterianas/química , Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Proteínas de la Membrana/química , Mutación , Antibacterianos/química , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/genética , Disulfuros/química , Escherichia coli/patogenicidad , Proteínas de Escherichia coli/genética , Biblioteca de Genes , Cinética , Lipopolisacáridos/química , Proteínas de la Membrana/genética , Mutagénesis , Reacción en Cadena de la Polimerasa , Proteína Disulfuro Isomerasas/química , Proteína Disulfuro Isomerasas/genética , Piridazinas/química , Quinonas/química , Espectrometría de Masa por Ionización de Electrospray , Relación Estructura-Actividad , Temperatura , Virulencia
5.
Mol Microbiol ; 103(3): 413-422, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27785850

RESUMEN

Disulfide bonds are found in many proteins associated with the cell wall of Escherichia coli, and for some of these proteins the disulfide bond is critical to their stability and function. One protein found to contain a disulfide bond is the essential cell division protein FtsN, but the importance of this bond to the protein's structural integrity is unclear. While it evidently plays a role in the proper folding of the SPOR domain of FtsN, this domain is non-essential, suggesting that the disulfide bond might also be dispensable. However, we find that FtsN mutants lacking cysteines give rise to filamentous growth. Furthermore, FtsN protein levels in strains expressing these mutants were significantly lower than in a strain expressing the wild-type allele, as were FtsN levels in strains incapable of making disulfide bonds (dsb- ) exposed to anaerobic conditions. These results strongly suggest that FtsN lacking a disulfide bond is unstable, thereby making this disulfide critical for function. We have previously found that dsb- strains fail to grow anaerobically, and the results presented here suggest that this growth defect may be due in part to misfolded FtsN. Thus, proper cell division in E. coli is dependent upon disulfide bond formation.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Proteínas de la Membrana/metabolismo , División Celular/fisiología , Pared Celular/metabolismo , Disulfuros , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Unión Proteica , Dominios Proteicos , Relación Estructura-Actividad
6.
Microbiology (Reading) ; 163(12): 1864-1879, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29139344

RESUMEN

Disulfide bonds confer stability and activity to proteins. Bioinformatic approaches allow predictions of which organisms make protein disulfide bonds and in which subcellular compartments disulfide bond formation takes place. Such an analysis, along with biochemical and protein structural data, suggests that many of the extremophile Crenarachaea make protein disulfide bonds in both the cytoplasm and the cell envelope. We have sought to determine the oxidative folding pathways in the sequenced genomes of the Crenarchaea, by seeking homologues of the enzymes known to be involved in disulfide bond formation in bacteria. Some Crenarchaea have two homologues of the cytoplasmic membrane protein VKOR, a protein required in many bacteria for the oxidation of bacterial DsbAs. We show that the two VKORs of Aeropyrum pernix assume opposite orientations in the cytoplasmic membrane, when expressed in E. coli. One has its active cysteines oriented toward the E. coli periplasm (ApVKORo) and the other toward the cytoplasm (ApVKORi). Furthermore, the ApVKORo promotes disulfide bond formation in the E. coli cell envelope, while the ApVKORi promotes disulfide bond formation in the E. coli cytoplasm via a co-expressed archaeal protein ApPDO. Amongst the VKORs from different archaeal species, the pairs of VKORs in each species are much more closely related to each other than to the VKORs of the other species. The results suggest two independent occurrences of the evolution of the two topologically inverted VKORs in archaea. Our results suggest a mechanistic basis for the formation of disulfide bonds in the cytoplasm of Crenarchaea.


Asunto(s)
Aeropyrum/metabolismo , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Disulfuros/química , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Aeropyrum/química , Aeropyrum/genética , Proteínas Arqueales/genética , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/metabolismo , Cisteína/química , Cisteína/metabolismo , Citoplasma/química , Citoplasma/genética , Citoplasma/metabolismo , Disulfuros/metabolismo , Proteínas de la Membrana/genética , Periplasma/genética , Periplasma/metabolismo , Pliegue de Proteína
7.
Nat Chem Biol ; 11(4): 292-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25686372

RESUMEN

In bacteria, disulfide bonds confer stability on many proteins exported to the cell envelope or beyond. These proteins include numerous bacterial virulence factors, and thus bacterial enzymes that promote disulfide bond formation represent targets for compounds inhibiting bacterial virulence. Here, we describe a new target- and cell-based screening methodology for identifying compounds that inhibit the disulfide bond-forming enzymes Escherichia coli DsbB (EcDsbB) or Mycobacterium tuberculosis VKOR (MtbVKOR), which can replace EcDsbB, although the two are not homologs. Initial screening of 51,487 compounds yielded six specifically inhibiting EcDsbB. These compounds share a structural motif and do not inhibit MtbVKOR. A medicinal chemistry approach led us to select related compounds, some of which are much more effective DsbB inhibitors than those found in the screen. These compounds inhibit purified DsbB and prevent anaerobic growth of E. coli. Furthermore, these compounds inhibit all but one of the DsbBs of nine other Gram-negative pathogenic bacteria tested.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Escherichia coli/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Mycobacterium tuberculosis/metabolismo , Agar/química , Antibacterianos/química , Dominio Catalítico , Química Farmacéutica/métodos , Técnicas Químicas Combinatorias , Disulfuros , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Transporte de Electrón , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/química , Espectrometría de Masas , Pruebas de Sensibilidad Microbiana , Mycobacterium smegmatis/metabolismo , Conformación Proteica , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/química , Pseudomonas aeruginosa/metabolismo
8.
Nature ; 463(7280): 507-12, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-20110994

RESUMEN

Vitamin K epoxide reductase (VKOR) generates vitamin K hydroquinone to sustain gamma-carboxylation of many blood coagulation factors. Here, we report the 3.6 A crystal structure of a bacterial homologue of VKOR from Synechococcus sp. The structure shows VKOR in complex with its naturally fused redox partner, a thioredoxin-like domain, and corresponds to an arrested state of electron transfer. The catalytic core of VKOR is a four transmembrane helix bundle that surrounds a quinone, connected through an additional transmembrane segment with the periplasmic thioredoxin-like domain. We propose a pathway for how VKOR uses electrons from cysteines of newly synthesized proteins to reduce a quinone, a mechanism confirmed by in vitro reconstitution of vitamin K-dependent disulphide bridge formation. Our results have implications for the mechanism of the mammalian VKOR and explain how mutations can cause resistance to the VKOR inhibitor warfarin, the most commonly used oral anticoagulant.


Asunto(s)
Oxigenasas de Función Mixta/química , Modelos Moleculares , Synechococcus/enzimología , Animales , Anticoagulantes , Proteínas Bacterianas/química , Dominio Catalítico , Disulfuros/química , Resistencia a Medicamentos/genética , Transporte de Electrón , Humanos , Proteínas de la Membrana/química , Oxigenasas de Función Mixta/genética , Estructura Terciaria de Proteína , Vitamina K Epóxido Reductasas , Warfarina
9.
Mol Cell ; 31(6): 896-908, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18922471

RESUMEN

We describe the NMR structure of DsbB, a polytopic helical membrane protein. DsbB, a bacterial cytoplasmic membrane protein, plays a key role in disulfide bond formation. It reoxidizes DsbA, the periplasmic protein disulfide oxidant, using the oxidizing power of membrane-embedded quinones. We determined the structure of an interloop disulfide bond form of DsbB, an intermediate in catalysis. Analysis of the structure and interactions with substrates DsbA and quinone reveals functionally relevant changes induced by these substrates. Analysis of the structure, dynamics measurements, and NMR chemical shifts around the interloop disulfide bond suggest how electron movement from DsbA to quinone through DsbB is regulated and facilitated. Our results demonstrate the extraordinary utility of NMR for functional characterization of polytopic integral membrane proteins and provide insights into the mechanism of DsbB catalysis.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Membrana Celular/enzimología , Disulfuros/metabolismo , Escherichia coli/citología , Escherichia coli/enzimología , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Sitios de Unión , Catálisis , Cisteína/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Proteínas de Escherichia coli/química , Membrana Dobles de Lípidos/metabolismo , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Oxidación-Reducción , Periplasma/enzimología , Proteína Disulfuro Isomerasas/química , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína , Soluciones , Ubiquinona
10.
Biochim Biophys Acta ; 1844(8): 1402-14, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24576574

RESUMEN

The formation of structural disulfide bonds is essential for the function and stability of a great number of proteins, particularly those that are secreted. There exists a variety of dedicated cellular catalysts and pathways from archaea to humans that ensure the formation of native disulfide bonds. In this review we describe the initial discoveries of these pathways and report progress in recent years in our understanding of the diversity of these pathways in prokaryotes, including those newly discovered in some archaea. We will also discuss the various successful efforts to achieve laboratory-based evolution and design of synthetic disulfide bond formation machineries in the bacterium Escherichia coli. These latter studies have also led to new more general insights into the redox environment of the cytoplasm and bacterial cell envelope. This article is part of a Special Issue entitled: Thiol-Based Redox Processes.


Asunto(s)
Disulfuros/química , Células Procariotas/metabolismo , Animales , Humanos , Oxidación-Reducción
11.
J Bacteriol ; 196(18): 3343-50, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25002543

RESUMEN

Targeted, translational LacZ fusions provided the initial support for the signal sequence hypothesis in prokaryotes and allowed for selection of the mutations that identified the Sec translocon. Many of these selections relied on the fact that expression of targeted, translational lacZ fusions like malE-lacZ and lamB-lacZ42-1 causes lethal toxicity as folded LacZ jams the translocation pore. However, there is another class of targeted LacZ fusions that do not jam the translocon. These targeted, nonjamming fusions also show toxic phenotypes that may be useful for selecting mutations in genes involved in posttranslocational protein folding and targeting; however, they have not been investigated to the same extent as their jamming counterparts. In fact, it is still unclear whether LacZ can be fully translocated in these fusions. It may be that they simply partition into the inner membrane where they can no longer participate in folding or assembly. In the present study, we systematically characterize the nonjamming fusions and determine their ultimate localization. We report that LacZ can be fully translocated into the periplasm, where it is toxic. We show that this toxicity is likely due to LacZ misfolding and that, in the absence of the periplasmic disulfide bond catalyst DsbA, LacZ folds in the periplasm. Using the novel phenotype of periplasmic ß-galactosidase activity, we show that the periplasmic chaperone FkpA contributes to LacZ folding in this nonnative compartment. We propose that targeted, nonjamming LacZ fusions may be used to further study folding and targeting in the periplasm of Escherichia coli.


Asunto(s)
Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Operón Lac/fisiología , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Genotipo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Isomerasa de Peptidilprolil/genética , Isomerasa de Peptidilprolil/metabolismo , Pliegue de Proteína , Proteínas Recombinantes , Transducción de Señal , Translocación Genética
12.
Trends Genet ; 26(11): 459-61, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20828856

RESUMEN

Marketers of genetic tests often openly or implicitly misrepresent the utility of genetic information. Scientists who are well aware of the current limitations to the utility of such tests are best placed to publicly counter misrepresentations of the science.


Asunto(s)
Información de Salud al Consumidor , Pruebas Genéticas , Genoma Humano , Investigación Biomédica , Información de Salud al Consumidor/tendencias , Humanos
13.
Nat Chem Biol ; 7(2): 101-5, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21186348

RESUMEN

The first and highly conserved step in glutathione (GSH) biosynthesis is formation of γ-glutamyl cysteine by the enzyme glutamate-cysteine ligase (GshA). However, bioinformatic analysis revealed that many prokaryotic species that encode GSH-dependent proteins lack the gene for this enzyme. To understand how bacteria cope without gshA, we isolated Escherichia coli ΔgshA multigenic suppressors that accumulated physiological levels of GSH. Mutations in both proB and proA, the first two genes in L-proline biosynthesis, provided a new pathway for γ-glutamyl cysteine formation via the selective interception of ProB-bound γ-glutamyl phosphate by amino acid thiols, likely through an S-to-N acyl shift mechanism. Bioinformatic analysis suggested that the L-proline biosynthetic pathway may have a second role in γ-glutamyl cysteine formation in prokaryotes. Also, we showed that this mechanism could be exploited to generate cytoplasmic redox buffers bioorthogonal to GSH.


Asunto(s)
Evolución Molecular , Glutatión/biosíntesis , Glutamato-Cisteína Ligasa/química , Glutamato-Cisteína Ligasa/genética , Mutación
14.
Proc Natl Acad Sci U S A ; 107(1): 297-301, 2010 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-20018758

RESUMEN

Blood coagulation in humans requires the activity of vitamin K epoxide reductase (VKOR), the target of the anticoagulant warfarin (Coumadin). Bacterial homologs of VKOR were recently found to participate in a pathway leading to disulfide bond formation in secreted proteins of many bacteria. Here we show that the VKOR homolog from the bacterium Mycobacterium tuberculosis, the causative agent of human tuberculosis, is inhibited by warfarin and that warfarin-resistant mutations of mycobacterial VKOR appear in similar locations to mutations found in human patients who require higher doses of warfarin. Deletion of VKOR results in a severe growth defect in mycobacteria, and the growth of M. tuberculosis is inhibited by warfarin. The bacterial VKOR homolog may represent a target for antibiotics and a model for genetic studies of human VKOR. We present a simple assay in Escherichia coli, based on a disulfide-sensitive beta-galactosidase, which can be used to screen for stronger inhibitors of the M. tuberculosis VKOR homolog.


Asunto(s)
Anticoagulantes/farmacología , Proteínas Bacterianas/metabolismo , Disulfuros/química , Oxigenasas de Función Mixta/antagonistas & inhibidores , Mycobacterium tuberculosis/efectos de los fármacos , Warfarina/farmacología , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Datos de Secuencia Molecular , Estructura Molecular , Mutación , Mycobacterium tuberculosis/enzimología , Oxidación-Reducción , Alineación de Secuencia , Vitamina K Epóxido Reductasas
15.
J Bacteriol ; 194(6): 1515-22, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22247510

RESUMEN

Production of deoxyribonucleotides for DNA synthesis is an essential and tightly regulated process. The class Ia ribonucleotide reductase (RNR), the product of the nrdAB genes, is required for aerobic growth of Escherichia coli. In catalyzing the reduction of ribonucleotides, two of the cysteines of RNR become oxidized, forming a disulfide bond. To regenerate active RNR, the cell uses thioredoxins and glutaredoxins to reduce the disulfide bond. Strains that lack thioredoxins 1 and 2 and glutaredoxin 1 do not grow because RNR remains in its oxidized, inactive form. However, suppressor mutations that lead to RNR overproduction allow glutaredoxin 3 to reduce sufficient RNR for growth of these mutant strains. We previously described suppressor mutations in the dnaA and dnaN genes that had such effects. Here we report the isolation of new mutations that lead to increased levels of RNR. These include mutations that were not known to influence production of RNR previously, such as a mutation in the hda gene and insertions in the nrdAB promoter region of insertion elements IS1 and IS5. Bioinformatic analysis raises the possibility that IS element insertion in this region represents an adaptive mechanism in nrdAB regulation in E. coli and closely related species. We also characterize mutations altering different amino acids in DnaA and DnaN from those isolated before.


Asunto(s)
Escherichia coli/enzimología , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Ribonucleótido Reductasas/biosíntesis , Ribonucleótido Reductasas/genética , Supresión Genética , Proteínas Bacterianas/genética , ADN Polimerasa III/genética , Proteínas de Unión al ADN/genética , Proteínas de Escherichia coli/genética , Genes Bacterianos , Mutagénesis Insercional , Regiones Promotoras Genéticas , Ribonucleósido Difosfato Reductasa/genética
16.
Proc Natl Acad Sci U S A ; 106(51): 21872-7, 2009 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-19995973

RESUMEN

Beta-lactam antibiotics inhibit penicillin binding proteins (PBPs) involved in peptidoglycan synthesis. Although inhibition of peptidoglycan biosynthesis is generally thought to induce cell lysis, the pattern and mechanism of cell lysis can vary substantially. Beta-lactams that inhibit FtsI, the only division specific PBP, block cell division and result in growth as filaments. These filaments ultimately lyse through a poorly understood mechanism. Here we find that one such beta-lactam, cephalexin, can, under certain conditions, lead instead to rapid lysis at nascent division sites through a process that requires the complete and ordered assembly of the divisome, the essential machinery involved in cell division. We propose that this assembly process (in which the localization of cell wall hydrolases depends on properly targeted FtsN, which in turn depends on the presence of FtsI) ensures that the biosynthetic machinery to form new septa is in place before the machinery to degrade septated daughter cells is enabled. Beta-lactams that target FtsI subvert this mechanism by inhibiting FtsI without perturbing the normal assembly of the cell division machinery and the consequent activation of cell wall hydrolases. One seemingly paradoxical implication of our results is that beta-lactam therapy may be improved by promoting active cell division.


Asunto(s)
División Celular/efectos de los fármacos , Cefalexina/farmacología , beta-Lactamas/farmacología
17.
J Bacteriol ; 193(18): 4988-92, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21784946

RESUMEN

FtsL and FtsB are two inner-membrane proteins that are essential constituents of the cell division apparatus of Escherichia coli. In this study, we demonstrate that the leucine zipper-like (LZ) motifs, located in the periplasmic domain of FtsL and FtsB, are required for an optimal interaction between these two essential proteins.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Leucina Zippers , Proteínas de la Membrana/metabolismo , Mapeo de Interacción de Proteínas , Proteínas de Ciclo Celular/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de la Membrana/genética , Unión Proteica
18.
Mol Microbiol ; 75(1): 13-28, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19968787

RESUMEN

Current dogma dictates that bacterial proteins with misoxidized disulfide bonds are shuffled into correctly oxidized states by DsbC. There are two proposed mechanisms for DsbC activity. The first involves a DsbC-only model of substrate disulfide rearrangement. The second invokes cycles of reduction and oxidation of substrate disulfide bonds by DsbC and DsbA respectively. Here, we addressed whether the second mechanism is important in vivo by identifying whether a periplasmic reductase could complement DsbC. We screened for naturally occurring periplasmic reductases in Bacteroides fragilis, a bacterium chosen because we predicted it encodes reductases and has a reducing periplasm. We found that the B. fragilis periplasmic protein TrxP has a thioredoxin fold with an extended N-terminal region; that it is a very active reductase but a poor isomerase; and that it fully complements dsbC. These results provide direct in vivo evidence that correctly folded protein is achievable via cycles of oxidation and reduction.


Asunto(s)
Bacteroides fragilis/enzimología , Oxidorreductasas/metabolismo , Proteínas Periplasmáticas/metabolismo , Proteína Disulfuro Isomerasas/química , Proteína Disulfuro Isomerasas/metabolismo , Pliegue de Proteína , Bacteroides fragilis/química , Bacteroides fragilis/genética , Cristalografía por Rayos X , Disulfuros/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Eliminación de Gen , Prueba de Complementación Genética , Modelos Biológicos , Modelos Moleculares , Oxidación-Reducción , Oxidorreductasas/química , Oxidorreductasas/genética , Proteínas Periplasmáticas/química , Proteínas Periplasmáticas/genética , Proteína Disulfuro Isomerasas/genética , Estructura Terciaria de Proteína
19.
Biochem J ; 430(3): 487-95, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20604742

RESUMEN

The Escherichia coli glutaredoxins 1 and 3 (Grx1 and Grx3) are structurally similar (37% sequence identity), yet have different activities in vivo. Unlike Grx3, Grx1 efficiently reduces protein disulfides in proteins such as RR (ribonucleotide reductase), whereas it is poor at reducing S-glutathionylated proteins. An E. coli strain lacking genes encoding thioredoxins 1 and 2 and Grx1 is not viable on either rich or minimal medium; however, a M43V mutation in Grx3 restores growth under these conditions and results in a Grx1-like protein [Ortenberg, Gon, Porat and Beckwith (2004) Proc. Natl. Acad. Sci. U.S.A. 101, 7439-7944]. To uncover the structural basis of this change in activity, we have compared wild-type and mutant Grx3 using CD and NMR spectroscopy. Ligand-induced stability measurements demonstrate that the Grx3(M43V/C65Y) mutant has acquired affinity for RR. Far-UV CD spectra reveal no significant differences, but differences are observed in the near-UV region indicative of tertiary structural changes. NMR (1)H-(15)N HSQC (heteronuclear single quantum coherence) spectra show that approximately half of the 82 residues experience significant (Deltadelta>0.03 p.p.m.) chemical shift deviations in the mutant, including nine residues experiencing extensive (Deltadelta > or =0.15 p.p.m.) deviations. To test whether the M43V mutation alters dynamic properties of Grx3, H/D (hydrogen/deuterium) exchange experiments were performed demonstrating that the rate at which backbone amides exchange protons with the solvent is dramatically enhanced in the mutant, particularly in the core of the protein. These data suggest that the Grx1-like activity of the Grx3(M43V/C65Y) mutant may be explained by enhanced intrinsic motion allowing for increased specificity towards larger substrates such as RR.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Glutarredoxinas/metabolismo , Proteínas Mutantes/metabolismo , Algoritmos , Sustitución de Aminoácidos , Dicroismo Circular , Estabilidad de Enzimas , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Glutarredoxinas/química , Glutarredoxinas/genética , Disulfuro de Glutatión/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutación , Oxidación-Reducción , Conformación Proteica , Estructura Terciaria de Proteína , Termodinámica
20.
Proc Natl Acad Sci U S A ; 105(18): 6735-40, 2008 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-18456836

RESUMEN

In Escherichia coli, the glutathione/glutaredoxin and thioredoxin pathways are essential for the reduction of cytoplasmic protein disulfide bonds, including those formed in the essential enzyme ribonucleotide reductase during its action on substrates. Double mutants lacking thioredoxin reductase (trxB) and glutathione reductase (gor) or glutathione biosynthesis (gshA) cannot grow. Growth of Deltagor DeltatrxB strains is restored by a mutant (ahpC*) of the peroxiredoxin AhpC, converting it to a disulfide reductase that generates reduced glutathione. Here, we show that ahpC* also restores growth to a DeltagshB DeltatrxB strain, which lacks glutathione and accumulates only its precursor gamma-glutamylcysteine (gamma-GC). It suppresses this strain by allowing accumulation of reduced gamma-GC, which can substitute for glutathione. Surprisingly, new ahpC suppressor mutations arose in a DeltagshA DeltatrxB strain lacking both glutathione and gamma-GC, a strain that ahpC* does not suppress. Some of these mutant AhpC proteins channel electrons into the disulfide-reducing pathways via either the thioredoxins or the glutaredoxins without, evidently, the intermediary of glutathione. Our results provide insights into the physiological functioning of the glutathione pathway and reveal surprising plasticity of a peroxidase because different mutant versions of AhpC can channel electrons into the disulfide-reducing pathways by at least four distinct routes. Despite the reductase activity of mutant AhpCs, these various suppressor strains exhibit an oxidizing cytoplasm and accumulate correctly folded disulfide-bonded proteins in their cytoplasm. Proteins most effectively oxidized vary between strains, potentially providing useful tools for expressing different disulfide-bonded proteins.


Asunto(s)
Evolución Biológica , Disulfuros/metabolismo , Escherichia coli/enzimología , Peroxidasa/metabolismo , Fosfatasa Alcalina/metabolismo , Cromatografía Líquida de Alta Presión , Cisteína/metabolismo , Citoplasma/enzimología , Dipéptidos/análisis , Proteínas de Escherichia coli/metabolismo , Glutarredoxinas/metabolismo , Cinética , Mutación/genética , NADH NADPH Oxidorreductasas/metabolismo , Oxidación-Reducción , Supresión Genética , Tiorredoxinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA