Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Phage (New Rochelle) ; 5(2): 53-62, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39119210

RESUMEN

Background: Endolysins are phage-encoded lytic enzymes that degrade bacterial peptidoglycan at the end of phage lytic cycles to release new phage particles. These enzymes are being explored as an alternative to small-molecule antibiotics. Methods: The crystal structure of KTN6 Gp46 was determined and compared with a ColabFold model. Cleavage specificity was examined using a peptidoglycan digest and reversed-phase high-performance liquid chromatography coupled to mass spectrometry (HPLC/MS). Results: The structure of KTN6 Gp46 could be determined at 1.4 Å resolution, and key differences in loops of the putative peptidoglycan binding domain were identified in comparison with its closest known homologue, the endolysin of phage SPN1S. Reversed-phase HPLC/MS analysis of the reaction products following peptidoglycan digestion confirmed the muramidase activity of Gp46, consistent with structural predictions. Conclusion: These insights into the structure and function of endolysins further expand the toolbox for endolysin engineering and explore their potential in enzyme-based antibacterial design strategies.

2.
Mol Microbiol ; 122(2): 230-242, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38994873

RESUMEN

Enterococcus faecalis is an opportunistic pathogen frequently causing nosocomial infections. The virulence of this organism is underpinned by its capacity to evade phagocytosis, allowing dissemination in the host. Immune evasion requires a surface polysaccharide produced by all enterococci, known as the enterococcal polysaccharide antigen (EPA). EPA consists of a cell wall-anchored rhamnose backbone substituted by strain-specific polysaccharides called 'decorations', essential for the biological activity of this polymer. However, the structural determinants required for innate immune evasion remain unknown, partly due to a lack of suitable validated assays. Here, we describe a quantitative, in vitro assay to investigate how EPA decorations alter phagocytosis. Using the E. faecalis model strain OG1RF, we demonstrate that a mutant with a deletion of the locus encoding EPA decorations can be used as a platform strain to express heterologous decorations, thereby providing an experimental system to investigate the inhibition of phagocytosis by strain-specific decorations. We show that the aggregation of cells lacking decorations is increasing phagocytosis and that this process does not involve the recognition of lipoproteins by macrophages. Collectively, our work provides novel insights into innate immune evasion by enterococci and paves the way for further studies to explore the structure/function relationship of EPA decorations.


Asunto(s)
Enterococcus faecalis , Evasión Inmune , Lipoproteínas , Macrófagos , Fagocitosis , Enterococcus faecalis/inmunología , Enterococcus faecalis/metabolismo , Enterococcus faecalis/genética , Lipoproteínas/metabolismo , Lipoproteínas/genética , Macrófagos/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Polisacáridos Bacterianos/metabolismo , Polisacáridos Bacterianos/inmunología , Humanos , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/genética , Inmunidad Innata , Virulencia , Animales , Ratones
3.
Virus Res ; 347: 199426, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38960003

RESUMEN

Enterococci are robust Gram-positive bacteria that pose a significant threat in healthcare settings due to antibiotic resistance, with vancomycin-resistant enterococci (VRE) most prominent. To tackle this issue, bacteriophages (bacterial viruses) can be exploited as they specifically and efficiently target bacteria. Here, we successfully isolated and characterised a set of novel phages: SHEF10, SHEF11, SHEF13, SHEF14, and SHEF16 which target E. faecalis (SHEF10,11,13), or E. faecium (SHEF13, SHEF14 & SHEF16) strains including a range of clinical and VRE isolates. Genomic analysis shows that all phages are strictly lytic and diverse in terms of genome size and content, quickly and effectively lysing strains at different multiplicity of infections. Detailed analysis of the broad host-range SHEF13 phage revealed the crucial role of the enterococcal polysaccharide antigen (EPA) variable region in its infection of E. faecalis V583. In parallel, the discovery of a carbohydrate-targeting domain (CBM22) found conserved within the three phage genomes indicates a role in cell surface interactions that may be important in phage-bacterial interactons. These findings advance our comprehension of phage-host interactions and pave the way for targeted therapeutic strategies against antibiotic-resistant enterococcal infections.


Asunto(s)
Bacteriófagos , Enterococcus faecalis , Genoma Viral , Especificidad del Huésped , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/clasificación , Bacteriófagos/aislamiento & purificación , Enterococcus faecalis/virología , Enterococcus faecalis/genética , Enterococcus faecium/virología , Enterococcus faecium/genética , Enterococcus/virología , Enterococcus/genética , Enterococos Resistentes a la Vancomicina/virología , Enterococos Resistentes a la Vancomicina/genética , Infecciones por Bacterias Grampositivas/microbiología , Humanos
4.
Methods Mol Biol ; 2836: 111-132, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38995539

RESUMEN

Peptidoglycan is a major and essential component of the bacterial cell envelope that confers cell shape and provides protection against internal osmotic pressure. This complex macromolecule is made of glycan strands cross-linked by short peptides, and its structure is continually modified throughout growth via a process referred to as "remodeling." Peptidoglycan remodeling allows cells to grow, adapt to their environment, and release fragments that can act as signaling molecules during host-pathogen interactions. Preparing peptidoglycan samples for structural analysis first requires purification of the peptidoglycan sacculus, followed by its enzymatic digestion into disaccharide peptides (muropeptides). These muropeptides can then be characterized by liquid chromatography coupled mass spectrometry (LC-MS) and used to infer the structure of intact peptidoglycan sacculi. Due to the presence of unusual crosslinks, noncanonical amino acids, and amino sugars, the analysis of peptidoglycan LC-MS datasets cannot be handled by traditional proteomics software. In this chapter, we describe a protocol to perform the analysis of peptidoglycan LC-MS datasets using the open-source software PGFinder. We provide a step-by-step strategy to deconvolute data from various mass spectrometry instruments, generate muropeptide databases, perform a PGFinder search, and process the data output.


Asunto(s)
Peptidoglicano , Programas Informáticos , Peptidoglicano/química , Peptidoglicano/metabolismo , Peptidoglicano/análisis , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Glicómica/métodos , Proteómica/métodos , Bacterias/metabolismo , Bacterias/química , Cromatografía Líquida con Espectrometría de Masas
5.
J Biol Chem ; 300(1): 105529, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38043796

RESUMEN

Clostridioides difficile is the leading cause of antibiotic-associated diarrhea worldwide with significant morbidity and mortality. This organism is naturally resistant to several beta-lactam antibiotics that inhibit the polymerization of peptidoglycan, an essential component of the bacteria cell envelope. Previous work has revealed that C. difficile peptidoglycan has an unusual composition. It mostly contains 3-3 cross-links, catalyzed by enzymes called L,D-transpeptidases (Ldts) that are poorly inhibited by beta-lactams. It was therefore hypothesized that peptidoglycan polymerization by these enzymes could underpin antibiotic resistance. Here, we investigated the catalytic activity of the three canonical Ldts encoded by C. difficile (LdtCd1, LdtCd2, and LdtCd3) in vitro and explored their contribution to growth and antibiotic resistance. We show that two of these enzymes catalyze the formation of novel types of peptidoglycan cross-links using meso-diaminopimelic acid both as a donor and an acceptor, also observed in peptidoglycan sacculi. We demonstrate that the simultaneous deletion of these three genes only has a minor impact on both peptidoglycan structure and resistance to beta-lactams. This unexpected result therefore implies that the formation of 3-3 peptidoglycan cross-links in C. difficile is catalyzed by as yet unidentified noncanonical Ldt enzymes.


Asunto(s)
Proteínas Bacterianas , Clostridioides difficile , Peptidoglicano , Peptidil Transferasas , Proteínas Bacterianas/química , Resistencia betalactámica , beta-Lactamas/farmacología , Catálisis , Clostridioides difficile/enzimología , Clostridioides difficile/genética , Peptidoglicano/química , Peptidil Transferasas/química , Peptidil Transferasas/genética
6.
J Biol Chem ; 300(1): 105494, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38006948

RESUMEN

Peptidoglycan is an essential component of the bacterial cell envelope that contains glycan chains substituted by short peptide stems. Peptide stems are polymerized by D,D-transpeptidases, which make bonds between the amino acid in position four of a donor stem and the third residue of an acceptor stem (4-3 cross-links). Some bacterial peptidoglycans also contain 3-3 cross-links that are formed by another class of enzymes called L,D-transpeptidases which contain a YkuD catalytic domain. In this work, we investigate the formation of unusual bacterial 1-3 peptidoglycan cross-links. We describe a version of the PGFinder software that can identify 1-3 cross-links and report the high-resolution peptidoglycan structure of Gluconobacter oxydans (a model organism within the Acetobacteraceae family). We reveal that G. oxydans peptidoglycan contains peptide stems made of a single alanine as well as several dipeptide stems with unusual amino acids at their C-terminus. Using a bioinformatics approach, we identified a G. oxydans mutant from a transposon library with a drastic reduction in 1-3 cross-links. Through complementation experiments in G. oxydans and recombinant protein production in a heterologous host, we identify an L,D-transpeptidase enzyme with a domain distantly related to the YkuD domain responsible for these non-canonical reactions. This work revisits the enzymatic capabilities of L,D-transpeptidases, a versatile family of enzymes that play a key role in bacterial peptidoglycan remodelling.


Asunto(s)
Proteínas Bacterianas , Gluconobacter oxydans , Modelos Moleculares , Peptidoglicano , Peptidil Transferasas , Aminoácidos/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico/genética , Peptidoglicano/química , Peptidoglicano/genética , Peptidoglicano/metabolismo , Peptidil Transferasas/química , Peptidil Transferasas/genética , Peptidil Transferasas/metabolismo , Programas Informáticos , Gluconobacter oxydans/enzimología , Gluconobacter oxydans/genética , Biología Computacional , Prueba de Complementación Genética , Estructura Terciaria de Proteína
7.
Commun Biol ; 6(1): 428, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37072531

RESUMEN

Control of cell size and morphology is of paramount importance for bacterial fitness. In the opportunistic pathogen Enterococcus faecalis, the formation of diplococci and short cell chains facilitates innate immune evasion and dissemination in the host. Minimisation of cell chain size relies on the activity of a peptidoglycan hydrolase called AtlA, dedicated to septum cleavage. To prevent autolysis, AtlA activity is tightly controlled, both temporally and spatially. Here, we show that the restricted localization of AtlA at the septum occurs via an unexpected mechanism. We demonstrate that the C-terminal LysM domain that allows the enzyme to bind peptidoglycan is essential to target this enzyme to the septum inside the cell before its translocation across the membrane. We identify a membrane-bound cytoplasmic protein partner (called AdmA) involved in the recruitment of AtlA via its LysM domains. This work reveals a moonlighting role for LysM domains, and a mechanism evolved to restrict the subcellular localization of a potentially lethal autolysin to its site of action.


Asunto(s)
Enterococcus faecalis , Peptidoglicano , Enterococcus faecalis/metabolismo , Peptidoglicano/metabolismo , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/genética , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Separación Celular
8.
Biomol NMR Assign ; 16(2): 247-251, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35665899

RESUMEN

Enterococcus faecalis is a major causative agent of hospital acquired infections. The ability of E. faecalis to evade the host immune system is essential during pathogenesis, which has been shown to be dependent on the complete separation of daughter cells by peptidoglycan hydrolases. AtlE is a peptidoglycan hydrolase which is predicted to bind to the cell wall of E. faecalis, via six C-terminal repeat sequences. Here, we report the near complete assignment of one of these six repeats, as well as the predicted backbone structure and dynamics. This data will provide a platform for future NMR studies to explore the ligand recognition motif of AtlE and help to uncover its potential role in E. faecalis virulence.


Asunto(s)
Enterococcus faecalis , N-Acetil Muramoil-L-Alanina Amidasa , Proteínas Bacterianas/metabolismo , Pared Celular/química , Pared Celular/metabolismo , Enterococcus faecalis/química , Enterococcus faecalis/metabolismo , Ligandos , N-Acetil Muramoil-L-Alanina Amidasa/análisis , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Resonancia Magnética Nuclear Biomolecular , Peptidoglicano/análisis , Peptidoglicano/química , Peptidoglicano/metabolismo
9.
Nat Commun ; 13(1): 2041, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440121

RESUMEN

Staphylococcus aureus frequently causes infections that are challenging to treat, leading to high rates of persistent and relapsing infection. Here, to understand how the host environment influences treatment outcomes, we study the impact of human serum on staphylococcal antibiotic susceptibility. We show that serum triggers a high degree of tolerance to the lipopeptide antibiotic daptomycin and several other classes of antibiotic. Serum-induced daptomycin tolerance is due to two independent mechanisms. Firstly, the host defence peptide LL-37 induces tolerance by triggering the staphylococcal GraRS two-component system, leading to increased peptidoglycan accumulation. Secondly, GraRS-independent increases in membrane cardiolipin abundance are required for full tolerance. When both mechanisms are blocked, S. aureus incubated in serum is as susceptible to daptomycin as when grown in laboratory media. Our work demonstrates that host factors can significantly modulate antibiotic susceptibility via diverse mechanisms, and combination therapy may provide a way to mitigate this.


Asunto(s)
Daptomicina , Infecciones Estafilocócicas , Antibacterianos/farmacología , Daptomicina/farmacología , Humanos , Pruebas de Sensibilidad Microbiana , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus
10.
J Biol Chem ; 298(5): 101915, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35398351

RESUMEN

The cleavage of septal peptidoglycan at the end of cell division facilitates the separation of the two daughter cells. The hydrolases involved in this process (called autolysins) are potentially lethal enzymes that can cause cell death; their activity, therefore, must be tightly controlled during cell growth. In Enterococcus faecalis, the N-acetylglucosaminidase AtlA plays a predominant role in cell separation. atlA mutants form long cell chains and are significantly less virulent in the zebrafish model of infection. The attenuated virulence of atlA mutants is underpinned by a limited dissemination of bacterial chains in the host organism and a more efficient uptake by phagocytes that clear the infection. AtlA has structural homologs in other important pathogens, such as Listeria monocytogenes and Salmonella typhimurium, and therefore represents an attractive model to design new inhibitors of bacterial pathogenesis. Here, we provide a 1.45 Å crystal structure of the E. faecalis AtlA catalytic domain that reveals a closed conformation of a conserved ß-hairpin and a complex network of hydrogen bonds that bring two catalytic residues to the ideal distance for an inverting mechanism. Based on the model of the AtlA-substrate complex, we identify key residues critical for substrate recognition and septum cleavage during bacterial growth. We propose that this work will provide useful information for the rational design of specific inhibitors targeting this enterococcal virulence factor and its orthologs in other pathogens.


Asunto(s)
Acetilglucosaminidasa , Enterococcus faecalis/enzimología , Acetilglucosaminidasa/química , Animales , Proteínas Bacterianas/metabolismo , Enterococcus faecalis/metabolismo , Peptidoglicano/metabolismo , Pez Cebra/metabolismo
11.
Elife ; 102021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34579805

RESUMEN

Many software solutions are available for proteomics and glycomics studies, but none are ideal for the structural analysis of peptidoglycan (PG), the essential and major component of bacterial cell envelopes. It icomprises glycan chains and peptide stems, both containing unusual amino acids and sugars. This has forced the field to rely on manual analysis approaches, which are time-consuming, labour-intensive, and prone to error. The lack of automated tools has hampered the ability to perform high-throughput analyses and prevented the adoption of a standard methodology. Here, we describe a novel tool called PGFinder for the analysis of PG structure and demonstrate that it represents a powerful tool to quantify PG fragments and discover novel structural features. Our analysis workflow, which relies on open-access tools, is a breakthrough towards a consistent and reproducible analysis of bacterial PGs. It represents a significant advance towards peptidoglycomics as a full-fledged discipline.


Asunto(s)
Bacterias/química , Peptidoglicano/química , Conformación de Carbohidratos , Conjuntos de Datos como Asunto , Glicómica , Espectrometría de Masas/métodos , Peptidoglicano/biosíntesis , Reproducibilidad de los Resultados , Programas Informáticos
12.
Int J Mol Sci ; 22(13)2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34281200

RESUMEN

The best-characterized members of the M23 family are glycyl-glycine hydrolases, such as lysostaphin (Lss) from Staphylococcus simulans or LytM from Staphylococcus aureus. Recently, enzymes with broad specificities were reported, such as EnpACD from Enterococcus faecalis, that cleaves D,L peptide bond between the stem peptide and a cross-bridge. Previously, the activity of EnpACD was demonstrated only on isolated peptidoglycan fragments. Herein we report conditions in which EnpACD lyses bacterial cells live with very high efficiency demonstrating great bacteriolytic potential, though limited to a low ionic strength environment. We have solved the structure of the EnpACD H109A inactive variant and analyzed it in the context of related peptidoglycan hydrolases structures to reveal the bases for the specificity determination. All M23 structures share a very conserved ß-sheet core which constitutes the rigid bottom of the substrate-binding groove and active site, while variable loops create the walls of the deep and narrow binding cleft. A detailed analysis of the binding groove architecture, specificity of M23 enzymes and D,L peptidases demonstrates that the substrate groove, which is particularly deep and narrow, is accessible preferably for peptides composed of amino acids with short side chains or subsequent L and D-isomers. As a result, the bottom of the groove is involved in interactions with the main chain of the substrate while the side chains are protruding in one plane towards the groove opening. We concluded that the selectivity of the substrates is based on their conformations allowed only for polyglycine chains and alternating chirality of the amino acids.


Asunto(s)
Endopeptidasas/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Péptido Hidrolasas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Enterococcus faecalis/genética , Enterococcus faecalis/metabolismo , Peptidoglicano/metabolismo , Profagos/genética , Profagos/metabolismo , Unión Proteica , Staphylococcus/metabolismo , Staphylococcus aureus/metabolismo , Especificidad por Sustrato
13.
Nat Microbiol ; 6(1): 19-26, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33139883

RESUMEN

Gram-negative bacteria have a cell envelope that comprises an outer membrane (OM), a peptidoglycan (PG) layer and an inner membrane (IM)1. The OM and PG are load-bearing, selectively permeable structures that are stabilized by cooperative interactions between IM and OM proteins2,3. In Escherichia coli, Braun's lipoprotein (Lpp) forms the only covalent tether between the OM and PG and is crucial for cell envelope stability4; however, most other Gram-negative bacteria lack Lpp so it has been assumed that alternative mechanisms of OM stabilization are present5. We used a glycoproteomic analysis of PG to show that ß-barrel OM proteins are covalently attached to PG in several Gram-negative species, including Coxiella burnetii, Agrobacterium tumefaciens and Legionella pneumophila. In C. burnetii, we found that four different types of covalent attachments occur between OM proteins and PG, with tethering of the ß-barrel OM protein BbpA becoming most abundant in the stationary phase and tethering of the lipoprotein LimB similar throughout the cell cycle. Using a genetic approach, we demonstrate that the cell cycle-dependent tethering of BbpA is partly dependent on a developmentally regulated L,D-transpeptidase (Ldt). We use our findings to propose a model of Gram-negative cell envelope stabilization that includes cell cycle control and an expanded role for Ldts in covalently attaching surface proteins to PG.


Asunto(s)
Agrobacterium tumefaciens/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Coxiella burnetii/metabolismo , Escherichia coli/metabolismo , Legionella pneumophila/metabolismo , Peptidoglicano/metabolismo , Ciclo Celular/fisiología , Membrana Celular/metabolismo , Pared Celular/metabolismo , Lipoproteínas/metabolismo , Simulación de Dinámica Molecular , Peptidil Transferasas/metabolismo , Unión Proteica/fisiología
14.
Cell Rep ; 31(12): 107813, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32579939

RESUMEN

Type VI secretion systems (T6SSs) are nanomachines used by bacteria to inject toxic effectors into competitors. The identity and mechanism of many effectors remain unknown. We characterized a Salmonella T6SS antibacterial effector called Tlde1 that is toxic in target-cell periplasm and is neutralized by its cognate immunity protein (Tldi1). Microscopy analysis reveals that cells expressing Tlde1 stop dividing and lose cell envelope integrity. Bioinformatic analysis uncovers similarities between Tlde1 and the catalytic domain of l,d-transpeptidases. Point mutations on conserved catalytic residues abrogate toxicity. Biochemical assays reveal that Tlde1 displays both l,d-carboxypeptidase activity by cleaving peptidoglycan tetrapeptides between meso-diaminopimelic acid3 and d-alanine4 and l,d-transpeptidase exchange activity by replacing d-alanine4 by a non-canonical d-amino acid. Phylogenetic analysis shows that Tlde1 homologs constitute a family of T6SS-associated effectors broadly distributed among Proteobacteria. This work expands our current knowledge about bacterial effectors used in interbacterial competition and reveals a different mechanism of bacterial antagonism.


Asunto(s)
Antibacterianos/farmacología , Peptidoglicano/metabolismo , Peptidil Transferasas/metabolismo , Sistemas de Secreción Tipo VI/metabolismo , Proteínas Bacterianas/metabolismo , División Celular/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Evolución Molecular , Periplasma/efectos de los fármacos , Periplasma/metabolismo , Proteobacteria/efectos de los fármacos , Proteobacteria/metabolismo , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/metabolismo
15.
mBio ; 11(2)2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32345640

RESUMEN

All enterococci produce a complex polysaccharide called the enterococcal polysaccharide antigen (EPA). This polymer is required for normal cell growth and division and for resistance to cephalosporins and plays a critical role in host-pathogen interaction. The EPA contributes to host colonization and is essential for virulence, conferring resistance to phagocytosis during the infection. Recent studies revealed that the "decorations" of the EPA polymer, encoded by genetic loci that are variable between isolates, underpin the biological activity of this surface polysaccharide. In this work, we investigated the structure of the EPA polymer produced by the high-risk enterococcal clonal complex Enterococcus faecalis V583. We analyzed purified EPA from the wild-type strain and a mutant lacking decorations and elucidated the structure of the EPA backbone and decorations. We showed that the rhamnan backbone of EPA is composed of a hexasaccharide repeat unit of C2- and C3-linked rhamnan chains, partially substituted in the C3 position by α-glucose (α-Glc) and in the C2 position by ß-N-acetylglucosamine (ß-GlcNAc). The so-called "EPA decorations" consist of phosphopolysaccharide chains corresponding to teichoic acids covalently bound to the rhamnan backbone. The elucidation of the complete EPA structure allowed us to propose a biosynthetic pathway, a first essential step toward the design of antimicrobials targeting the synthesis of this virulence factor.IMPORTANCE Enterococci are opportunistic pathogens responsible for hospital- and community-acquired infections. All enterococci produce a surface polysaccharide called EPA (enterococcal polysaccharide antigen) required for biofilm formation, antibiotic resistance, and pathogenesis. Despite the critical role of EPA in cell growth and division and as a major virulence factor, no information is available on its structure. Here, we report the complete structure of the EPA polymer produced by the model strain E. faecalis V583. We describe the structure of the EPA backbone, made of a rhamnan hexasaccharide substituted by Glc and GlcNAc residues, and show that teichoic acids are covalently bound to this rhamnan chain, forming the so-called "EPA decorations" essential for host colonization and pathogenesis. This report represents a key step in efforts to identify the structural properties of EPA that are essential for its biological activity and to identify novel targets to develop preventive and therapeutic approaches against enterococci.


Asunto(s)
Antígenos Bacterianos/química , Enterococcus faecalis/metabolismo , Polisacáridos/química , Antígenos Bacterianos/metabolismo , Desoxiazúcares/química , Desoxiazúcares/metabolismo , Humanos , Mananos/química , Mananos/metabolismo , Polisacáridos/metabolismo , Ácidos Teicoicos/química , Ácidos Teicoicos/metabolismo , Enterococos Resistentes a la Vancomicina/metabolismo
16.
Nat Chem Biol ; 16(1): 24-30, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31686030

RESUMEN

Lysostaphin is a bacteriolytic enzyme targeting peptidoglycan, the essential component of the bacterial cell envelope. It displays a very potent and specific activity toward staphylococci, including methicillin-resistant Staphylococcus aureus. Lysostaphin causes rapid cell lysis and disrupts biofilms, and is therefore a therapeutic agent of choice to eradicate staphylococcal infections. The C-terminal SH3b domain of lysostaphin recognizes peptidoglycans containing a pentaglycine crossbridge and has been proposed to drive the preferential digestion of staphylococcal cell walls. Here we elucidate the molecular mechanism underpinning recognition of staphylococcal peptidoglycan by the lysostaphin SH3b domain. We show that the pentaglycine crossbridge and the peptide stem are recognized by two independent binding sites located on opposite sides of the SH3b domain, thereby inducing a clustering of SH3b domains. We propose that this unusual binding mechanism allows synergistic and structurally dynamic recognition of S. aureus peptidoglycan and underpins the potent bacteriolytic activity of this enzyme.


Asunto(s)
Lisostafina/química , Peptidoglicano/química , Staphylococcus aureus/química , Bacteriólisis/efectos de los fármacos , Biopelículas , Pared Celular/química , Cromatografía Líquida de Alta Presión , Análisis Mutacional de ADN , Glicina/química , Ligandos , Espectroscopía de Resonancia Magnética , Mutagénesis Sitio-Dirigida , Péptidos/química , Unión Proteica , Dominios Proteicos , Proteínas Recombinantes/química , Dominios Homologos src
17.
Cell ; 179(3): 703-712.e7, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31587897

RESUMEN

Peptidoglycan (PG) is a defining feature of bacteria, involved in cell division, shape, and integrity. We previously reported that several genes related to PG biosynthesis were horizontally transferred from bacteria to the nuclear genome of mealybugs. Mealybugs are notable for containing a nested bacteria-within-bacterium endosymbiotic structure in specialized insect cells, where one bacterium, Moranella, lives in the cytoplasm of another bacterium, Tremblaya. Here we show that horizontally transferred genes on the mealybug genome work together with genes retained on the Moranella genome to produce a PG layer exclusively at the Moranella cell periphery. Furthermore, we show that an insect protein encoded by a horizontally transferred gene of bacterial origin is transported into the Moranella cytoplasm. These results provide a striking parallel to the genetic and biochemical mosaicism found in organelles, and prove that multiple horizontally transferred genes can become integrated into a functional pathway distributed between animal and bacterial endosymbiont genomes.


Asunto(s)
Bacterias/genética , Transferencia de Gen Horizontal , Hemípteros/genética , Peptidoglicano/biosíntesis , Simbiosis , Animales , Bacterias/patogenicidad , Genes Bacterianos , Hemípteros/microbiología , Interacciones Huésped-Patógeno , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Peptidoglicano/genética
18.
PLoS Pathog ; 15(5): e1007730, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31048927

RESUMEN

Enterococcus faecalis is an opportunistic pathogen with an intrinsically high resistance to lysozyme, a key effector of the innate immune system. This high level of resistance requires a complex network of transcriptional regulators and several genes (oatA, pgdA, dltA and sigV) acting synergistically to inhibit both the enzymatic and cationic antimicrobial peptide activities of lysozyme. We sought to identify novel genes modulating E. faecalis resistance to lysozyme. Random transposon mutagenesis carried out in the quadruple oatA/pgdA/dltA/sigV mutant led to the identification of several independent insertions clustered on the chromosome. These mutations were located in a locus referred to as the enterococcal polysaccharide antigen (EPA) variable region located downstream of the highly conserved epaA-epaR genes proposed to encode a core synthetic machinery. The epa variable region was previously proposed to be responsible for EPA decorations, but the role of this locus remains largely unknown. Here, we show that EPA decoration contributes to resistance towards charged antimicrobials and underpins virulence in the zebrafish model of infection by conferring resistance to phagocytosis. Collectively, our results indicate that the production of the EPA rhamnopolysaccharide backbone is not sufficient to promote E. faecalis infections and reveal an essential role of the modification of this surface polymer for enterococcal pathogenesis.


Asunto(s)
Antígenos de Superficie/inmunología , Enterococcus faecalis/patogenicidad , Infecciones por Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/microbiología , Muramidasa/inmunología , Polisacáridos/inmunología , Virulencia , Animales , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Enterococcus faecalis/genética , Enterococcus faecalis/inmunología , Infecciones por Bacterias Grampositivas/metabolismo , Muramidasa/metabolismo , Mutagénesis , Mutación , Polisacáridos/metabolismo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/inmunología , Pez Cebra/microbiología
19.
Nat Commun ; 9(1): 1263, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29593214

RESUMEN

Biopolymer composite cell walls maintain cell shape and resist forces in plants, fungi and bacteria. Peptidoglycan, a crucial antibiotic target and immunomodulator, performs this role in bacteria. The textbook structural model of peptidoglycan is a highly ordered, crystalline material. Here we use atomic force microscopy (AFM) to image individual glycan chains in peptidoglycan from Escherichia coli in unprecedented detail. We quantify and map the extent to which chains are oriented in a similar direction (orientational order), showing it is much less ordered than previously depicted. Combining AFM with size exclusion chromatography, we reveal glycan chains up to 200 nm long. We show that altered cell shape is associated with substantial changes in peptidoglycan biophysical properties. Glycans from E. coli in its normal rod shape are long and circumferentially oriented, but when a spheroid shape is induced (chemically or genetically) glycans become short and disordered.


Asunto(s)
Pared Celular/química , Imagen Molecular , Peptidoglicano/química , Polisacáridos/química , Antibacterianos/química , Bacillus subtilis/química , Escherichia coli/química , Microscopía de Fuerza Atómica , Microscopía de Contraste de Fase , Modelos Moleculares , Polímeros/química
20.
Elife ; 72018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29465397

RESUMEN

The bacterial cell wall is essential for viability, but despite its ability to withstand internal turgor must remain dynamic to permit growth and division. Peptidoglycan is the major cell wall structural polymer, whose synthesis requires multiple interacting components. The human pathogen Staphylococcus aureus is a prolate spheroid that divides in three orthogonal planes. Here, we have integrated cellular morphology during division with molecular level resolution imaging of peptidoglycan synthesis and the components responsible. Synthesis occurs across the developing septal surface in a diffuse pattern, a necessity of the observed septal geometry, that is matched by variegated division component distribution. Synthesis continues after septal annulus completion, where the core division component FtsZ remains. The novel molecular level information requires re-evaluation of the growth and division processes leading to a new conceptual model, whereby the cell cycle is expedited by a set of functionally connected but not regularly distributed components.


Asunto(s)
División Celular , Regulación Bacteriana de la Expresión Génica , Staphylococcus aureus/fisiología , Pared Celular/metabolismo , Redes Reguladoras de Genes , Modelos Biológicos , Peptidoglicano/metabolismo , Mapas de Interacción de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA