Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 604(7905): 371-376, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35388216

RESUMEN

The outer membrane of Gram-negative bacteria has an external leaflet that is largely composed of lipopolysaccharide, which provides a selective permeation barrier, particularly against antimicrobials1. The final and crucial step in the biosynthesis of lipopolysaccharide is the addition of a species-dependent O-antigen to the lipid A core oligosaccharide, which is catalysed by the O-antigen ligase WaaL2. Here we present structures of WaaL from Cupriavidus metallidurans, both in the apo state and in complex with its lipid carrier undecaprenyl pyrophosphate, determined by single-particle cryo-electron microscopy. The structures reveal that WaaL comprises 12 transmembrane helices and a predominantly α-helical periplasmic region, which we show contains many of the conserved residues that are required for catalysis. We observe a conserved fold within the GT-C family of glycosyltransferases and hypothesize that they have a common mechanism for shuttling the undecaprenyl-based carrier to and from the active site. The structures, combined with genetic, biochemical, bioinformatics and molecular dynamics simulation experiments, offer molecular details on how the ligands come in apposition, and allows us to propose a mechanistic model for catalysis. Together, our work provides a structural basis for lipopolysaccharide maturation in a member of the GT-C superfamily of glycosyltransferases.


Asunto(s)
Ligasas , Lipopolisacáridos , Antígenos O , Proteínas Bacterianas/química , Ligasas de Carbono-Oxígeno/química , Ligasas de Carbono-Oxígeno/genética , Microscopía por Crioelectrón , Glicosiltransferasas , Bacterias Gramnegativas , Lipopolisacáridos/química , Lipopolisacáridos/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(24): e2302580120, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37276423

RESUMEN

AmiA and AmiB are peptidoglycan-hydrolyzing enzymes from Escherichia coli that are required to break the peptidoglycan layer during bacterial cell division and maintain integrity of the cell envelope. In vivo, the activity of AmiA and AmiB is tightly controlled through their interactions with the membrane-bound FtsEX-EnvC complex. Activation of AmiA and AmiB requires access to a groove in the amidase-activating LytM domain of EnvC which is gated by ATP-driven conformational changes in FtsEX-EnvC complex. Here, we present a high-resolution structure of the isolated AmiA protein, confirming that it is autoinhibited in the same manner as AmiB and AmiC, and a complex of the AmiB enzymatic domain bound to the activating EnvC LytM domain. In isolation, the active site of AmiA is blocked by an autoinhibitory helix that binds directly to the catalytic zinc and fills the volume expected to accommodate peptidoglycan binding. In the complex, binding of the EnvC LytM domain induces a conformational change that displaces the amidase autoinhibitory helix and reorganizes the active site for activity. Our structures, together with complementary mutagenesis work, defines the conformational changes required to activate AmiA and/or AmiB through their interaction with their cognate activator EnvC.


Asunto(s)
Proteínas de Escherichia coli , Proteínas de Escherichia coli/metabolismo , Peptidoglicano/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Escherichia coli/metabolismo , Amidohidrolasas/metabolismo , Proteínas Bacterianas/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(8): e2215237120, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36787358

RESUMEN

Acinetobacter baumannii is a gram-negative bacterial pathogen that causes challenging nosocomial infections. ß-lactam targeting of penicillin-binding protein (PBP)-mediated cell wall peptidoglycan (PG) formation is a well-established antimicrobial strategy. Exposure to carbapenems or zinc (Zn)-deprived growth conditions leads to a rod-to-sphere morphological transition in A. baumannii, an effect resembling that caused by deficiency in the RodA-PBP2 PG synthesis complex required for cell wall elongation. While it is recognized that carbapenems preferentially acylate PBP2 in A. baumannii and therefore block the transpeptidase function of the RodA-PBP2 system, the molecular details underpinning cell wall elongation inhibition upon Zn starvation remain undefined. Here, we report the X-ray crystal structure of A. baumannii PBP2, revealing an unexpected Zn coordination site in the transpeptidase domain required for protein stability. Mutations in the Zn-binding site of PBP2 cause a loss of bacterial rod shape and increase susceptibility to ß-lactams, therefore providing a direct rationale for cell wall shape maintenance and Zn homeostasis in A. baumannii. Furthermore, the Zn-coordinating residues are conserved in various ß- and γ-proteobacterial PBP2 orthologs, consistent with a widespread Zn-binding requirement for function that has been previously unknown. Due to the emergence of resistance to virtually all marketed antibiotic classes, alternative or complementary antimicrobial strategies need to be explored. These findings offer a perspective for dual inhibition of Zn-dependent PG synthases and metallo-ß-lactamases by metal chelating agents, considered the most sought-after adjuvants to restore ß-lactam potency against gram-negative bacteria.


Asunto(s)
Acinetobacter baumannii , Peptidil Transferasas , Acinetobacter baumannii/metabolismo , Peptidil Transferasas/metabolismo , Zinc/metabolismo , Forma de la Célula , Antibacterianos/farmacología , Antibacterianos/metabolismo , Proteínas de Unión a las Penicilinas/metabolismo , beta-Lactamas/farmacología , Carbapenémicos/farmacología , Quelantes/farmacología , Sitios de Unión , Proteínas Bacterianas/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33785594

RESUMEN

Survival in the human host requires bacteria to respond to unfavorable conditions. In the important Gram-positive pathogen Streptococcus pneumoniae, cell wall biosynthesis proteins MurM and MurN are tRNA-dependent amino acyl transferases which lead to the production of branched muropeptides. We demonstrate that wild-type cells experience optimal growth under mildly acidic stressed conditions, but ΔmurMN strain displays growth arrest and extensive lysis. Furthermore, these stress conditions compromise the efficiency with which alanyl-tRNAAla synthetase can avoid noncognate mischarging of tRNAAla with serine, which is toxic to cells. The observed growth defects are rescued by inhibition of the stringent response pathway or by overexpression of the editing domain of alanyl-tRNAAla synthetase that enables detoxification of tRNA misacylation. Furthermore, MurM can incorporate seryl groups from mischarged Seryl-tRNAAlaUGC into cell wall precursors with exquisite specificity. We conclude that MurM contributes to the fidelity of translation control and modulates the stress response by decreasing the pool of mischarged tRNAs. Finally, we show that enhanced lysis of ΔmurMN pneumococci is caused by LytA, and the murMN operon influences macrophage phagocytosis in a LytA-dependent manner. Thus, MurMN attenuates stress responses with consequences for host-pathogen interactions. Our data suggest a causal link between misaminoacylated tRNA accumulation and activation of the stringent response. In order to prevent potential corruption of translation, consumption of seryl-tRNAAla by MurM may represent a first line of defense. When this mechanism is overwhelmed or absent (ΔmurMN), the stringent response shuts down translation to avoid toxic generation of mistranslated/misfolded proteins.


Asunto(s)
Proteínas Bacterianas/metabolismo , División Celular , Pared Celular/metabolismo , Péptido Sintasas/metabolismo , ARN de Transferencia/metabolismo , Streptococcus pneumoniae/metabolismo , Animales , Proteínas Bacterianas/genética , Línea Celular , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Operón , Péptido Sintasas/genética , Fagocitosis , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad
5.
Plant Physiol ; 190(1): 165-179, 2022 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-35471580

RESUMEN

Accumulating evidence suggests that peptidoglycan, consistent with a bacterial cell wall, is synthesized around the chloroplasts of many photosynthetic eukaryotes, from glaucophyte algae to early-diverging land plants including pteridophyte ferns, but the biosynthetic pathway has not been demonstrated. Here, we employed mass spectrometry and enzymology in a two-fold approach to characterize the synthesis of peptidoglycan in chloroplasts of the moss Physcomitrium (Physcomitrella) patens. To drive the accumulation of peptidoglycan pathway intermediates, P. patens was cultured with the antibiotics fosfomycin, D-cycloserine, and carbenicillin, which inhibit key peptidoglycan pathway proteins in bacteria. Mass spectrometry of the trichloroacetic acid-extracted moss metabolome revealed elevated levels of five of the predicted intermediates from uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) through the uridine diphosphate N-acetylmuramic acid (UDP-MurNAc)-D,L-diaminopimelate (DAP)-pentapeptide. Most Gram-negative bacteria, including cyanobacteria, incorporate meso-diaminopimelic acid (D,L-DAP) into the third residue of the stem peptide of peptidoglycan, as opposed to L-lysine, typical of most Gram-positive bacteria. To establish the specificity of D,L-DAP incorporation into the P. patens precursors, we analyzed the recombinant protein UDP-N-acetylmuramoyl-L-alanyl-D-glutamate-2,6-diaminopimelate ligase (MurE) from both P. patens and the cyanobacterium Anabaena sp. (Nostoc sp. strain PCC 7120). Both ligases incorporated D,L-DAP in almost complete preference to L-Lys, consistent with the mass spectrophotometric data, with catalytic efficiencies similar to previously documented Gram-negative bacterial MurE ligases. We discuss how these data accord with the conservation of active site residues common to DL-DAP-incorporating bacterial MurE ligases and of the probability of a horizontal gene transfer event within the plant peptidoglycan pathway.


Asunto(s)
Pared Celular , Peptidoglicano , Bacterias/metabolismo , Pared Celular/metabolismo , Cloroplastos/metabolismo , Bacterias Gramnegativas/metabolismo , Ligasas/metabolismo , Lisina/metabolismo , Peptidoglicano/química , Peptidoglicano/genética , Peptidoglicano/metabolismo , Uridina Difosfato/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(45): 28355-28365, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33097670

RESUMEN

FtsEX is a bacterial ABC transporter that regulates the activity of periplasmic peptidoglycan amidases via its interaction with the murein hydrolase activator, EnvC. In Escherichia coli, FtsEX is required to separate daughter cells after cell division and for viability in low-osmolarity media. Both the ATPase activity of FtsEX and its periplasmic interaction with EnvC are required for amidase activation, but the process itself is poorly understood. Here we present the 2.1 Å structure of the FtsX periplasmic domain in complex with its periplasmic partner, EnvC. The EnvC-FtsX periplasmic domain complex has a 1-to-2 stoichiometry with two distinct FtsX-binding sites located within an antiparallel coiled coil domain of EnvC. Residues involved in amidase activation map to a previously identified groove in the EnvC LytM domain that is here found to be occluded by a "restraining arm" suggesting a self-inhibition mechanism. Mutational analysis, combined with bacterial two-hybrid screens and in vivo functional assays, verifies the FtsEX residues required for EnvC binding and experimentally test a proposed mechanism for amidase activation. We also define a predicted link between FtsEX and integrity of the outer membrane. Both the ATPase activity of FtsEX and its periplasmic interaction with EnvC are required for resistance to membrane-attacking antibiotics and detergents to which E. coli would usually be considered intrinsically resistant. These structural and functional data provide compelling mechanistic insight into FtsEX-mediated regulation of EnvC and its downstream control of periplasmic peptidoglycan amidases.


Asunto(s)
Bacterias/metabolismo , Proteínas Bacterianas/química , Proteínas de Ciclo Celular/química , División Celular/fisiología , Endopeptidasas/química , Periplasma/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Membrana Celular/metabolismo , Cristalografía por Rayos X , Análisis Mutacional de ADN , Endopeptidasas/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli , Modelos Moleculares , Mutación , N-Acetil Muramoil-L-Alanina Amidasa/genética , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Periplasma/química , Unión Proteica , Conformación Proteica , Dominios Proteicos , Dominios y Motivos de Interacción de Proteínas
7.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34884635

RESUMEN

Bacteria must maintain the ability to modify and repair the peptidoglycan layer without jeopardising its essential functions in cell shape, cellular integrity and intermolecular interactions. A range of new experimental techniques is bringing an advanced understanding of how bacteria regulate and achieve peptidoglycan synthesis, particularly in respect of the central role played by complexes of Sporulation, Elongation or Division (SEDs) and class B penicillin-binding proteins required for cell division, growth and shape. In this review we highlight relationships implicated by a bioinformatic approach between the outer membrane, cytoskeletal components, periplasmic control proteins, and cell elongation/division proteins to provide further perspective on the interactions of these cell division, growth and shape complexes. We detail the network of protein interactions that assist in the formation of peptidoglycan and highlight the increasingly dynamic and connected set of protein machinery and macrostructures that assist in creating the cell envelope layers in Gram-negative bacteria.


Asunto(s)
Membrana Celular/metabolismo , Bacterias Gramnegativas/metabolismo , Proteínas de Unión a las Penicilinas/metabolismo , Peptidoglicano/metabolismo , Proteínas Periplasmáticas/metabolismo , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo
8.
J Am Chem Soc ; 142(11): 5034-5048, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32048840

RESUMEN

Penicillin binding proteins (PBPs) catalyzing transpeptidation reactions that stabilize the peptidoglycan component of the bacterial cell wall are the targets of ß-lactams, the most clinically successful antibiotics to date. However, PBP-transpeptidation enzymology has evaded detailed analysis, because of the historical unavailability of kinetically competent assays with physiologically relevant substrates and the previously unappreciated contribution of protein cofactors to PBP activity. By re-engineering peptidoglycan synthesis, we have constructed a continuous spectrophotometric assay for transpeptidation of native or near native peptidoglycan precursors and fragments by Escherichia coli PBP1B, allowing us to (a) identify recognition elements of transpeptidase substrates, (b) reveal a novel mechanism of stereochemical editing within peptidoglycan transpeptidation, (c) assess the impact of peptidoglycan substrates on ß-lactam targeting of transpeptidation, and (d) demonstrate that both substrates have to be bound before transpeptidation occurs. The results allow characterization of high molecular weight PBPs as enzymes and not merely the targets of ß-lactam acylation.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Proteínas de Unión a las Penicilinas/química , Peptidoglicano Glicosiltransferasa/química , Peptidoglicano/química , Monosacáridos de Poliisoprenil Fosfato/química , Oligosacáridos de Poliisoprenil Fosfato/química , D-Ala-D-Ala Carboxipeptidasa de Tipo Serina/química , Proteínas de la Membrana Bacteriana Externa/química , Biocatálisis , Pruebas de Enzimas/métodos , Cinética , Estereoisomerismo , Especificidad por Sustrato
9.
PLoS Pathog ; 13(10): e1006667, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29077761

RESUMEN

The O-acetylation of the essential cell wall polymer peptidoglycan occurs in most Gram-positive bacterial pathogens, including species of Staphylococcus, Streptococcus and Enterococcus. This modification to peptidoglycan protects these pathogens from the lytic action of the lysozymes of innate immunity systems and, as such, is recognized as a virulence factor. The key enzyme involved, peptidoglycan O-acetyltransferase A (OatA) represents a particular challenge to biochemical study since it is a membrane associated protein whose substrate is the insoluble peptidoglycan cell wall polymer. OatA is predicted to be bimodular, being comprised of an N-terminal integral membrane domain linked to a C-terminal extracytoplasmic domain. We present herein the first biochemical and kinetic characterization of the C-terminal catalytic domain of OatA from two important human pathogens, Staphylococcus aureus and Streptococcus pneumoniae. Using both pseudosubstrates and novel biosynthetically-prepared peptidoglycan polymers, we characterized distinct substrate specificities for the two enzymes. In addition, the high resolution crystal structure of the C-terminal domain reveals an SGNH/GDSL-like hydrolase fold with a catalytic triad of amino acids but with a non-canonical oxyanion hole structure. Site-specific replacements confirmed the identity of the catalytic and oxyanion hole residues. A model is presented for the O-acetylation of peptidoglycan whereby the translocation of acetyl groups from a cytoplasmic source across the cytoplasmic membrane is catalyzed by the N-terminal domain of OatA for their transfer to peptidoglycan by its C-terminal domain. This study on the structure-function relationship of OatA provides a molecular and mechanistic understanding of this bacterial resistance mechanism opening the prospect for novel chemotherapeutic exploration to enhance innate immunity protection against Gram-positive pathogens.


Asunto(s)
Acetiltransferasas/metabolismo , Bacterias Grampositivas/metabolismo , Peptidoglicano/metabolismo , Staphylococcus aureus/efectos de los fármacos , Factores de Virulencia/metabolismo , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Resistencia a Medicamentos , Humanos , Peptidoglicano/farmacología , Staphylococcus aureus/patogenicidad , Especificidad por Sustrato/inmunología , Virulencia
10.
Molecules ; 24(11)2019 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-31174247

RESUMEN

Mammalian protein N-glycosylation requires the transfer of an oligosaccharide containing 2 residues of N-acetylglucosamine, 9 residues of mannose and 3 residues of glucose (Glc3Man9 GlcNAc2) from Glc3Man9GlcNAc2-diphospho (PP)-dolichol (DLO) onto proteins in the endoplasmic reticulum (ER). Under some pathophysiological conditions, DLO biosynthesis is perturbed, and truncated DLO is hydrolyzed to yield oligosaccharyl phosphates (OSP) via unidentified mechanisms. DLO diphosphatase activity (DLODP) was described in vitro, but its characterization is hampered by a lack of convenient non-radioactive substrates. Our objective was to develop a fluorescence-based assay for DLO hydrolysis. Using a vancomycin-based solid-phase extraction procedure coupled with thin layer chromatography (TLC) and mass spectrometry, we demonstrate that mouse liver membrane extracts hydrolyze fluorescent bacterial lipid II (LII: GlcNAc-MurNAc(dansyl-pentapeptide)-PP-undecaprenol) to yield GlcNAc-MurNAc(dansyl-pentapeptide)-P (GM5P). GM5P production by solubilized liver microsomal proteins shows similar biochemical characteristics to those reported for human hepatocellular carcinoma HepG2 cell DLODP activity. To conclude, we show, for the first time, hydrolysis of lipid II by a eukaryotic enzyme. As LII and DLO are hydrolyzed by the same, or closely related, enzymes, fluorescent lipid II analogs are convenient non-radioactive substrates for investigating DLODP and DLODP-like activities.


Asunto(s)
Acetilglucosamina/química , Retículo Endoplásmico/química , Hígado/química , Oligosacáridos/química , Animales , Bacterias/química , Retículo Endoplásmico/metabolismo , Células Eucariotas/química , Células Eucariotas/metabolismo , Glucosa/química , Glicosilación , Células Hep G2 , Humanos , Hidrólisis , Lípidos/química , Hígado/metabolismo , Manosa/química , Ratones , Oligosacáridos/metabolismo , Uridina Difosfato Ácido N-Acetilmurámico/análogos & derivados , Uridina Difosfato Ácido N-Acetilmurámico/química
11.
J Biol Chem ; 292(7): 2854-2865, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28062575

RESUMEN

Pneumococcus resists ß-lactams by expressing variants of its target enzymes, the penicillin-binding proteins (PBPs), with many amino acid substitutions. Up to 10% of the sequence can be modified. These altered PBPs have a much reduced reactivity with the drugs but retain their physiological activity of cross-linking the peptidoglycan, the major constituent of the bacterial cell wall. However, because ß-lactams are chemical and structural mimics of the natural substrate, resistance mediated by altered PBPs raises the following paradox: how PBPs that react poorly with the drugs maintain a sufficient level of activity with the physiological substrate. This question is addressed for the first time in this study, which compares the peptidoglycan cross-linking activity of PBP2b from susceptible and resistant strains with their inhibition by different ß-lactams. Unexpectedly, the enzymatic activity of the variants did not correlate with their antibiotic reactivity. This finding indicates that some of the numerous amino acid substitutions were selected to restore a viable level of enzymatic activity by a compensatory molecular mechanism.


Asunto(s)
Farmacorresistencia Bacteriana/efectos de los fármacos , Proteínas de Unión a las Penicilinas/metabolismo , Streptococcus pneumoniae/efectos de los fármacos , beta-Lactamas/farmacología , Secuencia de Aminoácidos , Cefalosporinas/metabolismo , Proteínas de Unión a las Penicilinas/química , Proteínas de Unión a las Penicilinas/genética , Desnaturalización Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Streptococcus pneumoniae/metabolismo
12.
Biochem Biophys Res Commun ; 499(3): 594-599, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29601819

RESUMEN

Bacterial peptidoglycan is constructed by cross-linking sugar chains carrying pentapeptide building blocks with two d-alanine residues at the C-terminus. Incorporation into the polymer and subsequent breakdown of peptidoglycan releases a tetrapeptide with a single d-alanine residue. Removal of this residue is necessary for the tripeptide to receive a new D-Ala-D-Ala dipeptide in the synthetic pathway, but proteases are generally unable to work with substrates having residues of unusual chirality close to the scissile bond. Processing of the tetrapeptide is carried out by a dedicated ld-carboxypeptidase, which is of interest as a novel drug target. We describe the high resolution crystal structure of the enzyme from E. coli, and demonstrate the dimeric structure is highly conserved.


Asunto(s)
Carboxipeptidasas/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Dominio Catalítico , Cristalografía por Rayos X , Peso Molecular , Multimerización de Proteína , Estructura Secundaria de Proteína
13.
Sci Prog ; 99(2): 153-182, 2016 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28742471

RESUMEN

Antibiotics save many lives, but their efficacy is under threat: overprescription, population growth, and global travel all contribute to the rapid origination and spread of resistant strains. Exacerbating this threat is the fact that no new major classes of antibiotics have been discovered in the last 30 years: this is the "discovery void." We discuss the traditional molecular targets of antibiotics as well as putative novel targets.

14.
Biochemistry ; 54(24): 3803-13, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-26031209

RESUMEN

A simulation model of prokaryotic Z-ring assembly, based on the observed behavior of FtsZ in vitro as well as on in vivo parameters, is used to integrate critical processes in cell division. According to the model, the cell's ability to divide depends on a "contraction parameter" (χ) that links the force of contraction to the dynamics of FtsZ. This parameter accurately predicts the outcome of division. Evaluating the GTP binding strength, the FtsZ polymerization rate, and the intrinsic GTP hydrolysis/dissociation activity, we find that inhibition of GTP-FtsZ binding is an inefficient antibacterial target. Furthermore, simulations indicate that the temperature sensitivity of the ftsZ84 mutation arises from the conversion of FtsZ to a dual-specificity NTPase. Finally, the sensitivity to temperature of the rate of ATP hydrolysis, over the critical temperature range, leads us to conclude that the ftsZ84 mutation affects the turnover rate of the Z-ring much less strongly than previously reported.


Asunto(s)
Proteínas Bacterianas/metabolismo , División Celular , Proteínas del Citoesqueleto/metabolismo , Bacterias Gramnegativas/citología , Bacterias Grampositivas/citología , Modelos Biológicos , Proteínas Bacterianas/genética , Simulación por Computador , Citocinesis , Proteínas del Citoesqueleto/genética , Citoesqueleto/metabolismo , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/metabolismo , Mutación
15.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 6): 1373-81, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26057677

RESUMEN

LytA is responsible for the autolysis of many Streptococcus species, including pathogens such as S. pneumoniae, S. pseudopneumoniae and S. mitis. However, how this major autolysin achieves full activity remains unknown. Here, the full-length structure of the S. pneumoniae LytA dimer is reported at 2.1 Å resolution. Each subunit has an N-terminal amidase domain and a C-terminal choline-binding domain consisting of six choline-binding repeats, which form five canonical and one single-layered choline-binding sites. Site-directed mutageneses combined with enzymatic activity assays indicate that dimerization and binding to choline are two independent requirements for the autolytic activity of LytA in vivo. Altogether, it is suggested that dimerization and full occupancy of all choline-binding sites through binding to choline-containing TA chains enable LytA to adopt a fully active conformation which allows the amidase domain to cleave two lactyl-amide bonds located about 103 Å apart on the peptidoglycan.


Asunto(s)
Proteínas Bacterianas/química , N-Acetil Muramoil-L-Alanina Amidasa/química , Streptococcus/química , Conformación Proteica
16.
Sci Prog ; 98(Pt 4): 313-45, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26790174

RESUMEN

Cell division is a key event in the bacterial life cycle. It involves constriction at the midcell, so that one cell can give rise to two daughter cells. This constriction is mediated by a ring composed offibrous multimers of the protein FtsZ. However a host of additional factors is involved in the formation and dynamics of this "Z-ring" and this complicated apparatus is collectively known as the "divisome". We review the literature, with an emphasis on mathematical modelling, and show how such theoretical efforts have helped experimentalists to make sense of the at times bewildering data, and plan further experiments.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , División Celular/fisiología , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Escherichia coli/citología , Escherichia coli/metabolismo , Modelos Biológicos , Proteínas Bacterianas/ultraestructura , Simulación por Computador , Proteínas del Citoesqueleto/ultraestructura , Citoesqueleto/fisiología , Modelos Químicos , Relación Estructura-Actividad
17.
J Biol Chem ; 288(46): 33439-48, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24064214

RESUMEN

Formation of the peptidoglycan stem pentapeptide requires the insertion of both L and D amino acids by the ATP-dependent ligase enzymes MurC, -D, -E, and -F. The stereochemical control of the third position amino acid in the pentapeptide is crucial to maintain the fidelity of later biosynthetic steps contributing to cell morphology, antibiotic resistance, and pathogenesis. Here we determined the x-ray crystal structure of Staphylococcus aureus MurE UDP-N-acetylmuramoyl-L-alanyl-D-glutamate:meso-2,6-diaminopimelate ligase (MurE) (E.C. 6.3.2.7) at 1.8 Šresolution in the presence of ADP and the reaction product, UDP-MurNAc-L-Ala-γ-D-Glu-L-Lys. This structure provides for the first time a molecular understanding of how this Gram-positive enzyme discriminates between L-lysine and D,L-diaminopimelic acid, the predominant amino acid that replaces L-lysine in Gram-negative peptidoglycan. Despite the presence of a consensus sequence previously implicated in the selection of the third position residue in the stem pentapeptide in S. aureus MurE, the structure shows that only part of this sequence is involved in the selection of L-lysine. Instead, other parts of the protein contribute substrate-selecting residues, resulting in a lysine-binding pocket based on charge characteristics. Despite the absolute specificity for L-lysine, S. aureus MurE binds this substrate relatively poorly. In vivo analysis and metabolomic data reveal that this is compensated for by high cytoplasmic L-lysine concentrations. Therefore, both metabolic and structural constraints maintain the structural integrity of the staphylococcal peptidoglycan. This study provides a novel focus for S. aureus-directed antimicrobials based on dual targeting of essential amino acid biogenesis and its linkage to cell wall assembly.


Asunto(s)
Proteínas Bacterianas/química , Pared Celular/enzimología , Lisina/química , Péptido Sintasas/química , Peptidoglicano/química , Staphylococcus aureus/enzimología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Pared Celular/genética , Cristalografía por Rayos X , Lisina/genética , Lisina/metabolismo , Metabolómica , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Peptidoglicano/biosíntesis , Peptidoglicano/genética , Estructura Terciaria de Proteína , Staphylococcus aureus/genética
18.
Microbiology (Reading) ; 160(Pt 8): 1628-1636, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24858082

RESUMEN

The polymerization of lipid intermediate II by the transglycosylase activity of penicillin-binding proteins (PBPs) represents an important target for antibacterial action, but limited methods are available for quantitative assay of this reaction, or screening potential inhibitors. A new labelling method for lipid II polymerization products using Sanger's reagent (fluoro-2,4-dinitrobenzene), followed by gel permeation HPLC analysis, has permitted the observation of intermediate polymerization products for Staphylococcus aureus monofunctional transglycosylase MGT. Peak formation is inhibited by 6 µM ramoplanin or enduracidin. Characterization by mass spectrometry indicates the formation of tetrasaccharide and octasaccharide intermediates, but not a hexasaccharide intermediate, suggesting a dimerization of a lipid-linked tetrasaccharide. Numerical modelling of the time-course data supports a kinetic model involving addition to lipid-linked tetrasaccharide of either lipid II or lipid-linked tetrasaccharide. Observation of free octasaccharide suggests that hydrolysis of the undecaprenyl diphosphate lipid carrier occurs at this stage in peptidoglycan transglycosylation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Glicosiltransferasas/metabolismo , Peptidoglicano/química , Staphylococcus aureus/enzimología , Uridina Difosfato Ácido N-Acetilmurámico/análogos & derivados , Proteínas Bacterianas/química , Biocatálisis , Pared Celular/química , Pared Celular/metabolismo , Glicosiltransferasas/química , Cinética , Peptidoglicano/metabolismo , Polimerizacion , Staphylococcus aureus/química , Factores de Tiempo , Uridina Difosfato Ácido N-Acetilmurámico/química , Uridina Difosfato Ácido N-Acetilmurámico/metabolismo
19.
Chembiochem ; 15(9): 1300-8, 2014 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-24895118

RESUMEN

Translocase MraY is the site of action of lysis protein E from bacteriophage ϕX174. Previous genetic studies have shown that mutation F288L in transmembrane helix 9 of E. coli MraY confers resistance to protein E. Construction of a helical wheel model for transmembrane helix 9 of MraY and the transmembrane domain of protein E enabled the identification of an Arg-Trp-x-x-Trp (RWxxW) motif in protein E that might interact with Phe288 of MraY and the neighbouring Glu287. This motif is also found in a number of cationic antimicrobial peptide sequences. Synthetic dipeptides and pentapeptides based on the RWxxW consensus sequence showed inhibition of particulate E. coli MraY activity (IC50 200-600 µM), and demonstrated antimicrobial activity against E. coli (MIC 31-125 µg mL(-1)). Cationic antimicrobial peptides at a concentration of 100 µg mL(-1) containing Arg-Trp sequences also showed 30-60 % inhibition of E. coli MraY activity. Assay of the synthetic peptide inhibitors against recombinant MraY enzymes from Bacillus subtilis, Pseudomonas aeruginosa, and Micrococcus flavus (all of which lack Phe288) showed reduced levels of enzyme inhibition, and assay against recombinant E. coli MraY F288L and an E287A mutant demonstrated either reduced or no detectable enzyme inhibition, thus indicating that these peptides interact at this site. The MIC of Arg-Trp-octyl ester against E. coli was increased eightfold by overexpression of mraY, and was further increased by overexpression of the mraY mutant F288L, also consistent with inhibition at the RWxxW site. As this site is on the exterior face of the cytoplasmic membrane, it constitutes a potential new site for antimicrobial action, and provides a new cellular target for cationic antimicrobial peptides.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Escherichia coli/enzimología , Péptidos/farmacología , Transferasas/antagonistas & inhibidores , Transferasas/química , Proteínas Virales/metabolismo , Proteínas Bacterianas/metabolismo , Sitios de Unión/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Péptidos/síntesis química , Péptidos/química , Conformación Proteica , Relación Estructura-Actividad , Transferasas/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)
20.
Nanotechnology ; 25(28): 285101, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24972373

RESUMEN

Surfactant-mediated removal of proteins from biomembranes invariably results in partial or complete loss of function and disassembly of multi-protein complexes. We determined the capacity of styrene-co-maleic acid (SMA) co-polymer to remove components of the cell division machinery from the membrane of drug-resistant staphylococcal cells. SMA-lipid nanoparticles solubilized FtsZ-PBP2-PBP2a complexes from intact cells, demonstrating the close physical proximity of these proteins within the lipid bilayer. Exposure of bacteria to (-)-epicatechin gallate, a polyphenolic agent that abolishes ß-lactam resistance in staphylococci, disrupted the association between PBP2 and PBP2a. Thus, SMA purification provides a means to remove native integral membrane protein assemblages with minimal physical disruption and shows promise as a tool for the interrogation of molecular aspects of bacterial membrane protein structure and function.


Asunto(s)
Proteínas Bacterianas/química , Proteínas de la Membrana/química , Proteínas de Unión a las Penicilinas/química , Péptido Sintasas/química , Staphylococcus aureus/química , Tensoactivos/química , Catequina/análogos & derivados , Catequina/química , División Celular/fisiología , Membrana Dobles de Lípidos/química , Maleatos/química , Poliestirenos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA