Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(2): e1011047, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36730465

RESUMEN

The obligate intracellular Chlamydiaceae do not need to resist osmotic challenges and thus lost their cell wall in the course of evolution. Nevertheless, these pathogens maintain a rudimentary peptidoglycan machinery for cell division. They build a transient peptidoglycan ring, which is remodeled during the process of cell division and degraded afterwards. Uncontrolled degradation of peptidoglycan poses risks to the chlamydial cell, as essential building blocks might get lost or trigger host immune response upon release into the host cell. Here, we provide evidence that a primordial enzyme class prevents energy intensive de novo synthesis and uncontrolled release of immunogenic peptidoglycan subunits in Chlamydia trachomatis. Our data indicate that the homolog of a Bacillus NlpC/P60 protein is widely conserved among Chlamydiales. We show that the enzyme is tailored to hydrolyze peptidoglycan-derived peptides, does not interfere with peptidoglycan precursor biosynthesis, and is targeted by cysteine protease inhibitors in vitro and in cell culture. The peptidase plays a key role in the underexplored process of chlamydial peptidoglycan recycling. Our study suggests that chlamydiae orchestrate a closed-loop system of peptidoglycan ring biosynthesis, remodeling, and recycling to support cell division and maintain long-term residence inside the host. Operating at the intersection of energy recovery, cell division and immune evasion, the peptidoglycan recycling NlpC/P60 peptidase could be a promising target for the development of drugs that combine features of classical antibiotics and anti-virulence drugs.


Asunto(s)
Chlamydia trachomatis , Peptidoglicano , Chlamydia trachomatis/metabolismo , Peptidoglicano/metabolismo , Evasión Inmune , Proteínas Bacterianas/metabolismo , División Celular , Pared Celular/metabolismo , Péptido Hidrolasas/metabolismo
2.
Immunity ; 45(5): 1013-1023, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27851910

RESUMEN

Activation of the innate immune response in Metazoans is initiated through the recognition of microbes by host pattern-recognition receptors. In Drosophila, diaminopimelic acid (DAP)-containing peptidoglycan from Gram-negative bacteria is detected by the transmembrane receptor PGRP-LC and by the intracellular receptor PGRP-LE. Here, we show that PGRP-SD acted upstream of PGRP-LC as an extracellular receptor to enhance peptidoglycan-mediated activation of Imd signaling. Consistent with this, PGRP-SD mutants exhibited impaired activation of the Imd pathway and increased susceptibility to DAP-type bacteria. PGRP-SD enhanced the localization of peptidoglycans to the cell surface and hence promoted signaling. Moreover, PGRP-SD antagonized the action of PGRP-LB, an extracellular negative regulator, to fine-tune the intensity of the immune response. These data reveal that Drosophila PGRP-SD functions as an extracellular receptor similar to mammalian CD14 and demonstrate that, comparable to lipopolysaccharide sensing in mammals, Drosophila relies on both intra- and extracellular receptors for the detection of bacteria.


Asunto(s)
Proteínas Portadoras/inmunología , Proteínas de Drosophila/inmunología , Drosophila melanogaster/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Factores Reguladores Miogénicos/inmunología , Transducción de Señal/inmunología , Animales , Modelos Animales de Enfermedad , Inmunidad Innata/inmunología , Peptidoglicano/inmunología , Reacción en Cadena de la Polimerasa , Receptores de Reconocimiento de Patrones/inmunología
3.
Nucleic Acids Res ; 49(2): 684-699, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33367813

RESUMEN

The sequence of tRNAs is submitted to evolutionary constraints imposed by their multiple interactions with aminoacyl-tRNA synthetases, translation elongation factor Tu in complex with GTP (EF-Tu•GTP), and the ribosome, each being essential for accurate and effective decoding of messenger RNAs. In Staphylococcus aureus, an additional constraint is imposed by the participation of tRNAGly isoacceptors in the addition of a pentaglycine side chain to cell-wall peptidoglycan precursors by transferases FmhB, FemA and FemB. Three tRNAGly isoacceptors poorly interacting with EF-Tu•GTP and the ribosome were previously identified. Here, we show that these 'non-proteogenic' tRNAs are preferentially recognized by FmhB based on kinetic analyses and on synthesis of stable aminoacyl-tRNA analogues acting as inhibitors. Synthesis of chimeric tRNAs and of helices mimicking the tRNA acceptor arms revealed that this discrimination involves identity determinants exclusively present in the D and T stems and loops of non-proteogenic tRNAs, which belong to an evolutionary lineage only present in the staphylococci. EF-Tu•GTP competitively inhibited FmhB by sequestration of 'proteogenic' aminoacyl-tRNAs in vitro. Together, these results indicate that competition for the Gly-tRNAGly pool is restricted by both limited recognition of non-proteogenic tRNAs by EF-Tu•GTP and limited recognition of proteogenic tRNAs by FmhB.


Asunto(s)
Peptidoglicano/biosíntesis , ARN Bacteriano/metabolismo , ARN de Transferencia de Glicerina/metabolismo , Staphylococcus aureus/metabolismo , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Unión Competitiva , Pared Celular/metabolismo , Guanosina Trifosfato/metabolismo , Modelos Moleculares , Conformación de Ácido Nucleico , Factor Tu de Elongación Peptídica/metabolismo , Unión Proteica
4.
Mol Microbiol ; 115(6): 1357-1378, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33469978

RESUMEN

Francisella tularensis is a Gram-negative, intracellular bacterium that causes the zoonotic disease tularemia. Intracellular pathogens, including F. tularensis, have evolved mechanisms to survive in the harsh environment of macrophages and neutrophils, where they are exposed to cell envelope-damaging molecules. The bacterial cell wall, primarily composed of peptidoglycan (PG), maintains cell morphology, structure, and membrane integrity. Intracellular Gram-negative bacteria protect themselves from macrophage and neutrophil killing by recycling and repairing damaged PG--a process that involves over 50 different PG synthesis and recycling enzymes. Here, we identified a PG recycling enzyme, L,D-carboxypeptidase A (LdcA), of F. tularensis that is responsible for converting PG tetrapeptide stems to tripeptide stems. Unlike E. coli LdcA and most other orthologs, F. tularensis LdcA does not localize to the cytoplasm and also exhibits L,D-endopeptidase activity, converting PG pentapeptide stems to tripeptide stems. Loss of F. tularensis LdcA led to altered cell morphology and membrane integrity, as well as attenuation in a mouse pulmonary infection model and in primary and immortalized macrophages. Finally, an F. tularensis ldcA mutant protected mice against virulent Type A F. tularensis SchuS4 pulmonary challenge.


Asunto(s)
Carboxipeptidasas A/metabolismo , Pared Celular/metabolismo , Francisella tularensis/patogenicidad , Peptidoglicano/metabolismo , Tularemia/patología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Francisella tularensis/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C3H , Neutrófilos/microbiología , Alineación de Secuencia , Virulencia
5.
Proc Natl Acad Sci U S A ; 116(12): 5623-5632, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30819893

RESUMEN

Long-term intracellular symbiosis (or endosymbiosis) is widely distributed across invertebrates and is recognized as a major driving force in evolution. However, the maintenance of immune homeostasis in organisms chronically infected with mutualistic bacteria is a challenging task, and little is known about the molecular processes that limit endosymbiont immunogenicity and host inflammation. Here, we investigated peptidoglycan recognition protein (PGRP)-encoding genes in the cereal weevil Sitophilus zeamais's association with Sodalis pierantonius endosymbiont. We discovered that weevil pgrp-lb generates three transcripts via alternative splicing and differential regulation. A secreted isoform is expressed in insect tissues under pathogenic conditions through activation of the PGRP-LC receptor of the immune deficiency pathway. In addition, cytosolic and transmembrane isoforms are permanently produced within endosymbiont-bearing organ, the bacteriome, in a PGRP-LC-independent manner. Bacteriome isoforms specifically cleave the tracheal cytotoxin (TCT), a peptidoglycan monomer released by endosymbionts. pgrp-lb silencing by RNAi results in TCT escape from the bacteriome to other insect tissues, where it chronically activates the host systemic immunity through PGRP-LC. While such immune deregulations did not impact endosymbiont load, they did negatively affect host physiology, as attested by a diminished sexual maturation of adult weevils. Whereas pgrp-lb was first described in pathogenic interactions, this work shows that, in an endosymbiosis context, specific bacteriome isoforms have evolved, allowing endosymbiont TCT scavenging and preventing chronic endosymbiont-induced immune responses, thus promoting host homeostasis.


Asunto(s)
Proteínas Portadoras/fisiología , Interacciones Microbiota-Huesped/inmunología , Simbiosis/inmunología , Animales , Bacterias/inmunología , Bacterias/metabolismo , Proteínas Portadoras/inmunología , Citotoxinas , Interacciones Microbiota-Huesped/fisiología , Proteínas de Insectos/genética , Larva/metabolismo , Peptidoglicano/inmunología , Peptidoglicano/metabolismo , Isoformas de Proteínas , Gorgojos/genética , Gorgojos/metabolismo
6.
Anal Chem ; 93(4): 2342-2350, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33470796

RESUMEN

Peptidoglycan (PGN) is an essential structure found in the bacterial cell wall. During the bacterial life cycle, PGN continuously undergoes biosynthesis and degradation to ensure bacterial growth and division. The resulting PGN fragments (muropeptides and peptides), which are generated by the bacterial autolytic system, are usually transported into the cytoplasm to be recycled. On the other hand, PGN fragments can act as messenger molecules involved in the bacterial cell wall stress response as in the case of ß-lactamase induction in the presence of ß-lactam antibiotic or in triggering mammalian innate immune response. During their cellular life, bacteria modulate their PGN degradation by their autolytic system or their recognition by the mammalian innate immune system by chemically modifying their PGN. Among these modifications, the amidation of the ε-carboxyl group of meso-diaminopimelic acid present in the PGN peptide chain is frequently observed. Currently, the detection and quantitation of PGN-derived peptides is still challenging because of the difficulty in separating these highly hydrophilic molecules by RP-HPLC as these compounds are eluted closely after the column void volume or coeluted in many cases. Here, we report the use of capillary zone electrophoresis coupled via an electrospray-based CE-MS interface to high-resolution mass spectrometry for the quantitation of three PGN peptides of interest and their amidated derivatives in bacterial cytoplasmic extracts. The absolute quantitation of the tripeptide based on the [13C,15N] isotopically labeled standard was also performed in crude cytoplasmic extracts of bacteria grown in the presence or absence of a ß-lactam antibiotic (cephalosporin C). Despite the high complexity of the samples, the repeatability of the CZE-MS quantitation results was excellent, with relative standard deviations close to 1%. The global reproducibility of the method including biological handling was better than 20%.


Asunto(s)
Bacillus subtilis/metabolismo , Citoplasma/química , Electroforesis Capilar/métodos , Péptidos/química , Peptidoglicano/química , Espectrometría de Masa por Ionización de Electrospray/métodos , Bacillus subtilis/efectos de los fármacos , Cefalosporinas/farmacología , Peptidoglicano/metabolismo
7.
PLoS Pathog ; 15(1): e1007537, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30668586

RESUMEN

c-di-AMP is an important second messenger molecule that plays a pivotal role in regulating fundamental cellular processes, including osmotic and cell wall homeostasis in many Gram-positive organisms. In the opportunistic human pathogen Staphylococcus aureus, c-di-AMP is produced by the membrane-anchored DacA enzyme. Inactivation of this enzyme leads to a growth arrest under standard laboratory growth conditions and a re-sensitization of methicillin-resistant S. aureus (MRSA) strains to ß-lactam antibiotics. The gene coding for DacA is part of the conserved three-gene dacA/ybbR/glmM operon that also encodes the proposed DacA regulator YbbR and the essential phosphoglucosamine mutase GlmM, which is required for the production of glucosamine-1-phosphate, an early intermediate of peptidoglycan synthesis. These three proteins are thought to form a complex in vivo and, in this manner, help to fine-tune the cellular c-di-AMP levels. To further characterize this important regulatory complex, we conducted a comprehensive structural and functional analysis of the S. aureus DacA and GlmM enzymes by determining the structures of the S. aureus GlmM enzyme and the catalytic domain of DacA. Both proteins were found to be dimers in solution as well as in the crystal structures. Further site-directed mutagenesis, structural and enzymatic studies showed that multiple DacA dimers need to interact for enzymatic activity. We also show that DacA and GlmM form a stable complex in vitro and that S. aureus GlmM, but not Escherichia coli or Pseudomonas aeruginosa GlmM, acts as a strong inhibitor of DacA function without the requirement of any additional cellular factor. Based on Small Angle X-ray Scattering (SAXS) data, a model of the complex revealed that GlmM likely inhibits DacA by masking the active site of the cyclase and preventing higher oligomer formation. Together these results provide an important mechanistic insight into how c-di-AMP production can be regulated in the cell.


Asunto(s)
Inhibidores de Adenilato Ciclasa/metabolismo , Adenilil Ciclasas/metabolismo , Adenilil Ciclasas/ultraestructura , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Fosfatos de Dinucleósidos/antagonistas & inhibidores , Fosfatos de Dinucleósidos/metabolismo , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/metabolismo , Operón/genética , Fosfoglucomutasa/metabolismo , Liasas de Fósforo-Oxígeno/metabolismo , Dominios Proteicos , Dispersión del Ángulo Pequeño , Sistemas de Mensajero Secundario/genética , Infecciones Estafilocócicas/genética , Staphylococcus aureus/metabolismo , Staphylococcus aureus/fisiología , Difracción de Rayos X/métodos
8.
PLoS Pathog ; 15(9): e1007972, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31487328

RESUMEN

The biogenesis of bacterial cell-envelope polysaccharides requires the translocation, across the plasma membrane, of sugar sub-units that are produced inside the cytoplasm. To this end, the hydrophilic sugars are anchored to a lipid phosphate carrier (undecaprenyl phosphate (C55-P)), yielding membrane intermediates which are translocated to the outer face of the membrane. Finally, the glycan moiety is transferred to a nascent acceptor polymer, releasing the carrier in the "inactive" undecaprenyl pyrophosphate (C55-PP) form. Thus, C55-P is generated through the dephosphorylation of C55-PP, itself arising from either de novo synthesis or recycling. Two types of integral membrane C55-PP phosphatases were described: BacA enzymes and a sub-group of PAP2 enzymes (type 2 phosphatidic acid phosphatases). The human pathogen Helicobacter pylori does not contain BacA homologue but has four membrane PAP2 proteins: LpxE, LpxF, HP0350 and HP0851. Here, we report the physiological role of HP0851, renamed HupA, via multiple and complementary approaches ranging from a detailed biochemical characterization to the assessment of its effect on cell envelope metabolism and microbe-host interactions. HupA displays a dual function as being the main C55-PP pyrophosphatase (UppP) and phosphatidylglycerol phosphate phosphatase (PGPase). Although not essential in vitro, HupA was essential in vivo for stomach colonization. In vitro, the remaining UppP activity was carried out by LpxE in addition to its lipid A 1-phosphate phosphatase activity. Both HupA and LpxE have crucial roles in the biosynthesis of several cell wall polysaccharides and thus constitute potential targets for new therapeutic strategies.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Helicobacter pylori/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de la Membrana Bacteriana Externa/fisiología , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Pared Celular/metabolismo , Proteínas de Unión al ADN , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Femenino , Helicobacter pylori/patogenicidad , Ratones , Ratones Endogámicos , Pruebas de Sensibilidad Microbiana , Fosfatidato Fosfatasa , Monoéster Fosfórico Hidrolasas/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Polimixina B/farmacología , Pirofosfatasas/metabolismo , Estómago
9.
Int J Mol Sci ; 22(10)2021 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-34070225

RESUMEN

One of the major families of membrane proteins found in prokaryote genome corresponds to the transporters. Among them, the resistance-nodulation-cell division (RND) transporters are highly studied, as being responsible for one of the most problematic mechanisms used by bacteria to resist to antibiotics, i.e., the active efflux of drugs. In Gram-negative bacteria, these proteins are inserted in the inner membrane and form a tripartite assembly with an outer membrane factor and a periplasmic linker in order to cross the two membranes to expulse molecules outside of the cell. A lot of information has been collected to understand the functional mechanism of these pumps, especially with AcrAB-TolC from Escherichia coli, but one missing piece from all the suggested models is the role of peptidoglycan in the assembly. Here, by pull-down experiments with purified peptidoglycans, we precise the MexAB-OprM interaction with the peptidoglycan from Escherichia coli and Pseudomonas aeruginosa, highlighting a role of the peptidoglycan in stabilizing the MexA-OprM complex and also differences between the two Gram-negative bacteria peptidoglycans.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Escherichia coli/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Peptidoglicano/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Pared Celular/metabolismo , Farmacorresistencia Bacteriana , Escherichia coli/efectos de los fármacos , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/genética , Modelos Moleculares , Peptidoglicano/química , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética
10.
J Bacteriol ; 202(23)2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-32958631

RESUMEN

Colicin M is an enzymatic bacteriocin produced by some Escherichia coli strains which provokes cell lysis of competitor strains by hydrolysis of the cell wall peptidoglycan undecaprenyl-PP-MurNAc(-pentapeptide)-GlcNAc (lipid II) precursor. The overexpression of a gene, cbrA (formerly yidS), was shown to protect E. coli cells from the deleterious effects of this colicin, but the underlying resistance mechanism was not established. We report here that a major structural modification of the undecaprenyl-phosphate carrier lipid and of its derivatives occurred in membranes of CbrA-overexpressing cells, which explains the acquisition of resistance toward this bacteriocin. Indeed, a main fraction of these lipids, including the lipid II peptidoglycan precursor, now displayed a saturated isoprene unit at the α-position, i.e., the unit closest to the colicin M cleavage site. Only unsaturated forms of these lipids were normally detectable in wild-type cells. In vitro and in vivo assays showed that colicin M did not hydrolyze α-saturated lipid II, clearly identifying this substrate modification as the resistance mechanism. These saturated forms of undecaprenyl-phosphate and lipid II remained substrates of the different enzymes participating in peptidoglycan biosynthesis and carrier lipid recycling, allowing this colicin M-resistance mechanism to occur without affecting this essential pathway.IMPORTANCE Overexpression of the chromosomal cbrA gene allows E. coli to resist colicin M (ColM), a bacteriocin specifically hydrolyzing the undecaprenyl-PP-MurNAc(-pentapeptide)-GlcNAc (lipid II) peptidoglycan precursor of targeted cells. This resistance results from a CbrA-dependent modification of the precursor structure, i.e., reduction of the α-isoprenyl bond of C55-carrier lipid moiety that is proximal to ColM cleavage site. This modification, observed here for the first time in eubacteria, annihilates the ColM activity without affecting peptidoglycan biogenesis. These data, which further increase our knowledge of the substrate specificity of this colicin, highlight the capability of E. coli to generate reduced forms of C55-carrier lipid and its derivatives. Whether the function of this modification is only relevant with respect to ColM resistance is now questioned.


Asunto(s)
Antibacterianos/farmacología , Colicinas/farmacología , Farmacorresistencia Bacteriana , Proteínas de Escherichia coli/metabolismo , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Flavoproteínas/metabolismo , Peptidoglicano/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Flavoproteínas/genética , Peptidoglicano/química , Uridina Difosfato Ácido N-Acetilmurámico/análogos & derivados , Uridina Difosfato Ácido N-Acetilmurámico/metabolismo
11.
Microbiology (Reading) ; 166(6): 567-578, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375990

RESUMEN

Clostridium difficile 630 possesses a cryptic but functional gene cluster vanGCd homologous to the vanG operon of Enterococcus faecalis. Expression of vanGCd in the presence of subinhibitory concentrations of vancomycin is accompanied by peptidoglycan amidation on the meso-DAP residue. In this paper, we report the presence of two potential asparagine synthetase genes named asnB and asnB2 in the C. difficile genome whose products were potentially involved in this peptidoglycan structure modification. We found that asnB expression was only induced when C. difficile was grown in the presence of vancomycin, yet independently from the vanGCd resistance and regulation operons. In addition, peptidoglycan precursors were not amidated when asnB was inactivated. No change in vancomycin MIC was observed in the asnB mutant strain. In contrast, overexpression of asnB resulted in the amidation of most of the C. difficile peptidoglycan precursors and in a weak increase of vancomycin susceptibility. AsnB activity was confirmed in E. coli. In contrast, the expression of the second asparagine synthetase, AsnB2, was not induced in the presence of vancomycin. In summary, our results demonstrate that AsnB is responsible for peptidoglycan amidation of C. difficile in the presence of vancomycin.


Asunto(s)
Antibacterianos/farmacología , Aspartatoamoníaco Ligasa/metabolismo , Proteínas Bacterianas/metabolismo , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/enzimología , Peptidoglicano/metabolismo , Vancomicina/farmacología , Aspartatoamoníaco Ligasa/genética , Proteínas Bacterianas/genética , Clostridioides difficile/genética , Farmacorresistencia Bacteriana , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Familia de Multigenes , Operón
12.
J Bacteriol ; 201(13)2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30988031

RESUMEN

Certain Pseudomonas aeruginosa strains produce a homolog of colicin M, namely, PaeM, that specifically inhibits peptidoglycan biosynthesis of susceptible P. aeruginosa strains by hydrolyzing the lipid II intermediate precursor. Two variants of this pyocin were identified whose sequences mainly differed in the N-terminal protein moiety, i.e., the region involved in the binding to the FiuA outer membrane receptor and translocation into the periplasm. The antibacterial activity of these two variants, PaeM1 and PaeM2, was tested against various P. aeruginosa strains comprising reference strains PAO1 and PA14, PaeM-producing strains, and 60 clinical isolates. Seven of these strains, including PAO1, were susceptible to only one variant (2 to PaeM1 and 5 to PaeM2), and 11 were affected by both. The remaining strains, including PA14 and four PaeM1 producers, were resistant to both variants. The differences in the antibacterial spectra of the two PaeM homologs prompted us to investigate the molecular determinants allowing their internalization into P. aeruginosa cells, taking the PAO1 strain that is susceptible to PaeM2 but resistant to PaeM1 as the indicator strain. Heterologous expression of fiuA gene orthologs from different strains into PAO1, site-directed mutagenesis experiments, and construction of PaeM chimeric proteins provided evidence that the cell susceptibility and discrimination differences between the PaeM variants resulted from a polymorphism of both the pyocin and the outer membrane receptor FiuA. Moreover, we found that a third component, TonB1, a protein involved in iron transport in P. aeruginosa, working together with FiuA and the ExbB/ExbD complex, was directly implicated in this discrimination.IMPORTANCE Bacterial antibiotic resistance constitutes a threat to human health, imposing the need for identification of new targets and development of new strategies to fight multiresistant pathogens. Bacteriocins and other weapons that bacteria have themselves developed to kill competitors are therefore of great interest and a valuable source of inspiration for us. Attention was paid here to two variants of a colicin M homolog (PaeM) produced by certain strains of P. aeruginosa that inhibit the growth of their congeners by blocking cell wall peptidoglycan synthesis. Molecular determinants allowing recognition of these pyocins by the outer membrane receptor FiuA were identified, and a receptor polymorphism affecting the susceptibility of P. aeruginosa clinical strains was highlighted, providing new insights into the potential use of these pyocins as an alternative to antibiotics.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , Farmacorresistencia Bacteriana , Polimorfismo Genético , Pseudomonas aeruginosa/genética , Piocinas/farmacología , Antibacterianos/farmacología , Pared Celular/química , Mutagénesis Sitio-Dirigida , Peptidoglicano/química , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Receptores de Superficie Celular
13.
Chemistry ; 24(22): 5743-5747, 2018 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-29389045

RESUMEN

The bacterial cell wall peptidoglycan contains unusual l- and d-amino acids assembled as branched peptides. Insight into the biosynthesis of the polymer has been hampered by limited access to substrates and to suitable polymerization assays. Here we report the full synthesis of the peptide stem of peptidoglycan precursors from two pathogenic bacteria, Enterococcus faecium and Mycobacterium tuberculosis, and the development of a sensitive post-derivatization assay for their cross-linking by l,d-transpeptidases. Access to series of stem peptides showed that amidation of free carboxyl groups is essential for optimal enzyme activity, in particular the amidation of diaminopimelate (DAP) residues for the cross-linking activity of the l,d-transpeptidase LdtMt2 from M. tuberculosis. Accordingly, construction of a conditional mutant established the essential role of AsnB indicating that this DAP amidotransferase is an attractive target for the development of anti-mycobacterial drugs.


Asunto(s)
Enterococcus faecium/enzimología , Mycobacterium tuberculosis/enzimología , Peptidoglicano/biosíntesis , Peptidil Transferasas/metabolismo , Transaminasas/metabolismo , Pared Celular/metabolismo , Enterococcus faecium/química , Enterococcus faecium/genética , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/genética , Peptidil Transferasas/efectos de los fármacos , beta-Lactamas/química
14.
Chemistry ; 24(56): 14911-14915, 2018 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-30020544

RESUMEN

Conjugation of RNA with multiple partners to obtain mimics of complex biomolecules is limited by the identification of orthogonal reactions. Here, lipid-carbohydrate-peptidyl-RNA conjugates were obtained by post-functionalization reactions, solid-phase synthesis, and enzymatic steps, to generate molecules mimicking the substrates of FmhB, an essential peptidoglycan synthesis enzyme of Staphylococcus aureus. Mimics of Gly-tRNAGly and lipid intermediate II (undecaprenyl-diphospho-disaccharide-pentapeptide) were combined in a single "bi-substrate" inhibitor (IC50 =56 nm). The synthetic route was exploited to generate substrates and inhibitors containing d-lactate residue (d-Lac) instead of d-Ala at the C-terminus of the pentapeptide stem, a modification responsible for vancomycin resistance in the enterococci. The substitution impaired recognition of peptidoglycan precursors by FmhB. The associated fitness cost may account for limited dissemination of vancomycin resistance genes in S. aureus.


Asunto(s)
Carbohidratos/química , Pared Celular/enzimología , Inhibidores Enzimáticos/química , Lípidos/química , ARN/química , Técnicas de Síntesis en Fase Sólida/métodos , Staphylococcus aureus/enzimología , Proteínas Bacterianas/antagonistas & inhibidores , Carbohidratos/síntesis química , Carbohidratos/farmacología , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Descubrimiento de Drogas , Farmacorresistencia Bacteriana , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Humanos , Lípidos/síntesis química , Lípidos/farmacología , Peptidoglicano/metabolismo , ARN/síntesis química , ARN/farmacología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/metabolismo , Especificidad por Sustrato
15.
Cell Mol Life Sci ; 74(12): 2319-2332, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28168443

RESUMEN

Type 2 phosphatidic acid phosphatases (PAP2s) can be either soluble or integral membrane enzymes. In bacteria, integral membrane PAP2s play major roles in the metabolisms of glycerophospholipids, undecaprenyl-phosphate (C55-P) lipid carrier and lipopolysaccharides. By in vivo functional experiments and biochemical characterization we show that the membrane PAP2 coded by the Bacillus subtilis yodM gene is the principal phosphatidylglycerol phosphate (PGP) phosphatase of B. subtilis. We also confirm that this enzyme, renamed bsPgpB, has a weaker activity on C55-PP. Moreover, we solved the crystal structure of bsPgpB at 2.25 Å resolution, with tungstate (a phosphate analog) in the active site. The structure reveals two lipid chains in the active site vicinity, allowing for PGP substrate modeling and molecular dynamic simulation. Site-directed mutagenesis confirmed the residues important for substrate specificity, providing a basis for predicting the lipids preferentially dephosphorylated by membrane PAP2s.


Asunto(s)
Bacillus subtilis/enzimología , Membrana Celular/enzimología , Fosfatidato Fosfatasa/química , Fosfatidato Fosfatasa/metabolismo , Bacillus subtilis/genética , Dominio Catalítico , Cristalografía por Rayos X , Escherichia coli/metabolismo , Genes Bacterianos , Prueba de Complementación Genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Fosfatidato Fosfatasa/genética , Fosfatidilgliceroles/metabolismo , Solubilidad , Especificidad por Sustrato
16.
J Immunol ; 194(10): 4641-9, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25870247

RESUMEN

Mucosal-associated invariant T (MAIT) cells recognize microbial compounds presented by the MHC-related 1 (MR1) protein. Although riboflavin precursor derivatives from Gram-positive bacteria have been characterized, some level of ligand heterogeneity has been suggested through the analysis of the MAIT cell TCR repertoire in humans and differential reactivity of human MAIT cell clones according to the bacteria. In this study, using Gram-negative bacteria mutated for the riboflavin biosynthetic pathway, we show a strict correlation between the ability to synthesize the 5-amino-ribityl-uracil riboflavin precursor and to activate polyclonal and quasi-monoclonal mouse MAIT cells. To our knowledge, we show for the first time that the semipurified bacterial fraction and the synthetic ligand activate murine MAIT cells in vitro and in vivo. We describe new MR1 ligands that do not activate MAIT cells but compete with bacterial and synthetic compounds activating MAIT cells, providing the capacity to modulate MAIT cell activation. Through competition experiments, we show that the most active synthetic MAIT cell ligand displays the same functional avidity for MR1 as does the microbial compound. Altogether, these results show that most, if not all, MAIT cell ligands found in Escherichia coli are related to the riboflavin biosynthetic pathway and display very limited heterogeneity.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Activación de Linfocitos/inmunología , Células T Asesinas Naturales/inmunología , Riboflavina/inmunología , Riboflavina/metabolismo , Animales , Modelos Animales de Enfermedad , Escherichia coli/inmunología , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/inmunología , Técnicas In Vitro , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Antígenos de Histocompatibilidad Menor , Membrana Mucosa/inmunología
17.
J Biol Chem ; 290(21): 13079-94, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25847251

RESUMEN

A gene named ltsA was earlier identified in Rhodococcus and Corynebacterium species while screening for mutations leading to increased cell susceptibility to lysozyme. The encoded protein belonged to a huge family of glutamine amidotransferases whose members catalyze amide nitrogen transfer from glutamine to various specific acceptor substrates. We here describe detailed physiological and biochemical investigations demonstrating the specific role of LtsA protein from Corynebacterium glutamicum (LtsACg) in the modification by amidation of cell wall peptidoglycan diaminopimelic acid (DAP) residues. A morphologically altered but viable ΔltsA mutant was generated, which displays a high susceptibility to lysozyme and ß-lactam antibiotics. Analysis of its peptidoglycan structure revealed a total loss of DAP amidation, a modification that was found in 80% of DAP residues in the wild-type polymer. The cell peptidoglycan content and cross-linking were otherwise not modified in the mutant. Heterologous expression of LtsACg in Escherichia coli yielded a massive and toxic incorporation of amidated DAP into the peptidoglycan that ultimately led to cell lysis. In vitro assays confirmed the amidotransferase activity of LtsACg and showed that this enzyme used the peptidoglycan lipid intermediates I and II but not, or only marginally, the UDP-MurNAc pentapeptide nucleotide precursor as acceptor substrates. As is generally the case for glutamine amidotransferases, either glutamine or NH4(+) could serve as the donor substrate for LtsACg. The enzyme did not amidate tripeptide- and tetrapeptide-truncated versions of lipid I, indicating a strict specificity for a pentapeptide chain length.


Asunto(s)
Amidas/química , Proteínas Bacterianas/metabolismo , Corynebacterium/metabolismo , Ácido Diaminopimélico/química , Muramidasa/metabolismo , Peptidoglicano/metabolismo , Transaminasas/metabolismo , Amidas/metabolismo , Secuencia de Aminoácidos , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Western Blotting , Pared Celular/metabolismo , Células Cultivadas , Corynebacterium/genética , Corynebacterium/crecimiento & desarrollo , Ácido Diaminopimélico/metabolismo , Técnicas para Inmunoenzimas , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Mutación/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transaminasas/genética
18.
Mol Microbiol ; 96(3): 437-47, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25532427

RESUMEN

Phages play critical roles in the spread of virulence factors and control of bacterial populations through their predation of bacteria. An essential step in the phage lifecycle is genome entry, where the infecting phage must productively interact with the components of the bacterial cell envelope in order to transmit its genome out of the viral particle and into the host cell cytoplasm. In this study, we characterize this process for the Escherichia coli phage HK97. We have discovered that HK97 genome injection requires the activities of the inner membrane glucose transporter protein, PtsG, and the periplasmic chaperone, FkpA. The requirements for PtsG and FkpA are determined by the sequence of the phage tape measure protein (TMP). We also identify a region of the TMP that mediates inhibition of phage genome injection by the HK97 superinfection exclusion protein, gp15. This region of the TMP also determines the PtsG requirement, and we show that gp15-mediated inhibition requires PtsG. Based on these data, we present a model for the in vivo genome injection process of phage HK97 and postulate a mechanism by which the inhibitory action of gp15 is reliant upon PtsG.


Asunto(s)
Colifagos/fisiología , ADN Viral/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/virología , Proteínas de la Membrana/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Proteínas de la Cola de los Virus/metabolismo , Internalización del Virus , Escherichia coli/metabolismo , Genoma Viral , Chaperonas Moleculares/metabolismo , Proteínas Periplasmáticas/metabolismo
19.
Angew Chem Int Ed Engl ; 55(43): 13553-13557, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27667506

RESUMEN

RNA functionalization is challenging due to the instability of RNA and the limited range of available enzymatic reactions. We developed a strategy based on solid phase synthesis and post-functionalization to introduce an electrophilic site at the 3' end of tRNA analogues. The squarate diester used as an electrophile enabled sequential amidation and provided asymmetric squaramides with high selectivity. The squaramate-RNAs specifically reacted with the lysine of UDP-MurNAc-pentapeptide, a peptidoglycan precursor used by the aminoacyl-transferase FemXWv for synthesis of the bacterial cell wall. The peptidyl-RNA obtained with squaramate-RNA and unprotected UDP-MurNAc-pentapeptide efficiently inhibited FemXWv . The squaramate unit also promoted specific cross-linking of RNA to the catalytic Lys of FemXWv but not to related transferases recognizing different aminoacyl-tRNAs. Thus, squaramate-RNAs provide specificity for cross-linking with defined groups in complex biomolecules due to its unique reactivity.


Asunto(s)
Aminoaciltransferasas/metabolismo , Reactivos de Enlaces Cruzados/metabolismo , Péptidos/metabolismo , ARN de Transferencia/metabolismo , ARN/biosíntesis , Uridina Difosfato Ácido N-Acetilmurámico/análogos & derivados , Aminoaciltransferasas/química , Reactivos de Enlaces Cruzados/química , Modelos Moleculares , Conformación Molecular , Péptidos/química , ARN/química , ARN de Transferencia/química , Uridina Difosfato Ácido N-Acetilmurámico/química , Uridina Difosfato Ácido N-Acetilmurámico/metabolismo
20.
Glycobiology ; 25(3): 319-30, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25344445

RESUMEN

Members of the GH73 glycosidase family cleave the ß-1,4-glycosidic bond between the N-acetylglucosaminyl (GlcNAc) and N-acetylmuramyl (MurNAc) moieties in bacterial peptidoglycan. A catalytic mechanism has been proposed for members FlgJ, Auto, AcmA and Atl(WM) and the structural analysis of FlgJ and Auto revealed a conserved α/ß fold reminiscent of the distantly related GH23 lysozyme. Comparison of the active site residues reveals variability in the nature of the catalytic general base suggesting two distinct catalytic mechanisms: an inverting mechanism involving two distant glutamate residues and a substrate-assisted mechanism involving anchimeric assistance by the C2-acetamido group of the GlcNAc moiety. Herein, we present the biochemical characterization and crystal structure of TM0633 from the hyperthermophilic bacterium Thermotoga maritima. TM0633 adopts the α/ß fold of the family and displays ß-N-acetylglucosaminidase activity on intact peptidoglycan sacculi. Site-directed mutagenesis identifies Glu34, Glu65 and Tyr118 as important residues for catalysis. A thorough bioinformatic analysis of the GH73 sequences identified five phylogenetic clusters. TM0633, FlgJ and Auto belong to a group of three clusters that conserve two carboxylate residues involved in a classical inverting acid-base mechanism. Members of the other two clusters lack a conserved catalytic general base supporting a substrate-assisted mechanism. Molecular modeling of representative members from each cluster suggests that variability in length of the ß-hairpin region above the active site confers ligand-binding specificity and modulates the catalytic mechanisms within the GH73 family.


Asunto(s)
Acetilglucosaminidasa/química , Proteínas Bacterianas/química , Thermotoga maritima/enzimología , Acetilglucosaminidasa/genética , Acetilglucosaminidasa/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Datos de Secuencia Molecular , Filogenia , Thermotoga maritima/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA