Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Infect Dis ; 215(12): 1836-1845, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28863473

RESUMO

For many gram-positive pathogens, conjugative plasmid transfer is an important means of spreading antibiotic resistance . Therefore, the search for alternative treatments to fight and prevent infections caused by these bacteria has become of major interest. In the present study, we evaluated the protein TraM, from the conjugative plasmid pIP501, as a potential vaccine candidate. Anti-TraM antiserum mediated in vitro opsonophagocytic killing of the strain harboring the pIP501 plasmid and also proved to be cross-reactive against other clinically relevant enterococcal and staphylococcal strains. Specificity of antibodies toward TraM was confirmed by results of an opsonophagocytic inhibition assay and Western blot. In addition, conjugative transfer experiments proved that TraM is essential for the transfer of pIP501. Finally, immunization with either TraM or anti-TraM antiserum reduced significantly the colony counts in mice livers, demonstrating that TraM is a promising vaccine candidate against enterococci and other gram-positive pathogens.


Assuntos
Proteínas de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Farmacorresistência Bacteriana Múltipla/imunologia , Enterococcus faecalis/imunologia , Escherichia coli/imunologia , Sistemas de Secreção Tipo IV/imunologia , Animais , Proteínas de Bactérias/genética , Western Blotting , Enterococcus faecalis/genética , Escherichia coli/genética , Feminino , Fígado/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos , Transporte Proteico , Coelhos , Staphylococcus aureus/imunologia
2.
J Infect Dis ; 214(2): 189-95, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-26984142

RESUMO

Enterococcus faecium is a common cause of nosocomial infections, of which infective endocarditis is associated with substantial mortality. In this study, we used a microarray-based transposon mapping (M-TraM) approach to evaluate a rat endocarditis model and identified a gene, originally annotated as "fruA" and renamed "bepA," putatively encoding a carbohydrate phosphotransferase system (PTS) permease (biofilm and endocarditis-associated permease A [BepA]), as important in infective endocarditis. This gene is highly enriched in E. faecium clinical isolates and absent in commensal isolates that are not associated with infection. Confirmation of the phenotype was established in a competition experiment of wild-type and a markerless bepA mutant in a rat endocarditis model. In addition, deletion of bepA impaired biofilm formation in vitro in the presence of 100% human serum and metabolism of ß-methyl-D-glucoside. ß-glucoside metabolism has been linked to the metabolism of glycosaminoglycans that are exposed on injured heart valves, where bacteria attach and form vegetations. Therefore, we propose that the PTS permease BepA is directly implicated in E. faecium pathogenesis.


Assuntos
Biofilmes/crescimento & desenvolvimento , Endocardite Bacteriana/microbiologia , Endocardite Bacteriana/fisiopatologia , Enterococcus faecium/enzimologia , Enterococcus faecium/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Fatores de Virulência/metabolismo , Animais , Elementos de DNA Transponíveis , Modelos Animais de Doenças , Enterococcus faecium/patogenicidade , Feminino , Técnicas de Inativação de Genes , Testes Genéticos , Proteínas de Membrana Transportadoras/genética , Mutagênese Insercional , Fosfotransferases/genética , Fosfotransferases/metabolismo , Ratos Wistar , Fatores de Virulência/genética
3.
Infect Genet Evol ; 37: 215-24, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26611826

RESUMO

Most bacterial species produce capsular polysaccharides that contribute to disease pathogenesis through evasion of the host innate immune system and are also involved in inhibiting leukocyte killing. In the present study, we identified a gene in Enterococcus faecium U0317 with homologies to the polysaccharide biosynthesis protein CapD that is made up of 336 amino acids and putatively catalyzes N-linked glycosylation. A capD deletion mutant was constructed and complemented by homologous recombination that was confirmed by PCR and sequencing. The mutant revealed different growth behavior and morphological changes compared to wild-type by scanning electron microscopy, also the capD mutant showed a strong hydrophobicity and that was reversed in the reconstituted mutant. For further characterization and functional analyses, in-vitro cell culture and in-vivo a mouse infection models were used. Antibodies directed against alpha lipotechoic acid (αLTA) and the peptidyl-prolyl cis-trans isomerase (αPpiC), effectively mediated the opsonophagocytic killing in the capD knock-out mutant, while this activity was not observed in the wild-type and reconstituted mutant. By comparison more than 2-fold decrease was seen in mutant colonization and adherence to both T24 and Caco2 cells. However, a significant higher bacterial colonization was observed in capD mutant during bacteremia in the animal model, while virulence in a mouse UTI (urinary tract infection) model, there were no obvious differences. Further studies are needed to elucidate the function of capsular polysaccharide synthesis gene clusters and its involvement in the disease pathogenesis with the aim to develop targeted therapies to treat multidrug-resistant E. faecium infections.


Assuntos
Cápsulas Bacterianas/genética , Enterococcus faecium/crescimento & desenvolvimento , Infecções por Bactérias Gram-Positivas/microbiologia , Polissacarídeos/biossíntese , Animais , Aderência Bacteriana , Cápsulas Bacterianas/química , Cápsulas Bacterianas/metabolismo , Células CACO-2 , Linhagem Celular , Modelos Animais de Doenças , Enterococcus faecium/isolamento & purificação , Enterococcus faecium/patogenicidade , Humanos , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Mutação
4.
Sci Rep ; 5: 18255, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26675410

RESUMO

Enterococcus faecium is a commensal of the mammalian gastrointestinal tract, but is also found in non-enteric environments where it can grow between 10 °C and 45 °C. E. faecium has recently emerged as a multi-drug resistant nosocomial pathogen. We hypothesized that genes involved in the colonization and infection of mammals exhibit temperature-regulated expression control and we therefore performed a transcriptome analysis of the clinical isolate E. faecium E1162, during mid-exponential growth at 25 °C and 37 °C. One of the genes that exhibited differential expression between 25 °C and 37 °C, was predicted to encode a peptidoglycan-anchored surface protein. The N-terminal domain of this protein is unique to E. faecium and closely related enterococci, while the C-terminal domain is homologous to the Streptococcus agalactiae surface protein BibA. This region of the protein contains proline-rich repeats, leading us to name the protein PrpA for proline-rich protein A. We found that PrpA is a surface-exposed protein which is most abundant during exponential growth at 37 °C in E. faecium E1162. The heterologously expressed and purified N-terminal domain of PrpA was able to bind to the extracellular matrix proteins fibrinogen and fibronectin. In addition, the N-terminal domain of PrpA interacted with both non-activated and activated platelets.


Assuntos
Proteínas de Bactérias/metabolismo , Plaquetas/metabolismo , Enterococcus faecium/metabolismo , Fibrinogênio/metabolismo , Fibronectinas/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sequência de Bases , Sítios de Ligação/genética , Infecção Hospitalar/microbiologia , Enterococcus faecium/genética , Enterococcus faecium/ultraestrutura , Regulação Bacteriana da Expressão Gênica , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Peptidoglicano/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Temperatura
6.
PLoS One ; 10(7): e0132949, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26172831

RESUMO

In this study, we investigated the impact of the cell membrane composition of E. faecalis on its recognition by the host immune system. To this end, we employed an E. faecalis deletion mutant (ΔbgsA) that does not synthesize the major cell membrane glycolipid diglycosyl-diacylglycerol (DGlcDAG). Proteomic analysis revealed that 13 of a total of 21 upregulated surface-associated proteins of E. faecalis ΔbgsA were lipoproteins. This led to a total lipoprotein content in the cell membrane of 35.8% in ΔbgsA compared to only 9.4% in wild-type bacteria. Increased lipoprotein content strongly affected the recognition of ΔbgsA by mouse macrophages in vitro with an increased stimulation of TNF-α production by heat-fixed bacteria and secreted antigens. Inactivation of the prolipoprotein diacylglycerol transferase (lgt) in ΔbgsA abrogated TNF-α induction by a ΔbgsA_lgt double mutant indicating that lipoproteins mediate increased activation of mouse macrophages by ΔbgsA. Heat-fixed ΔbgsA bacteria, culture supernatant, or cell membrane lipid extract activated transfected HEK cells in a TLR2-dependent fashion; the same was not true of wild-type bacteria. In mice infected intraperitoneally with a sublethal dose of E. faecalis we observed a 70% greater mortality in mice infected with ΔbgsA compared with wild-type-infected mice. Increased mortality due to ΔbgsA infection was associated with elevated plasma levels of the inflammatory cytokines TNF-α, IL-6 and MIP-2. In summary, our results provide evidence that an E. faecalis mutant lacking its major bilayer forming glycolipid DGlcDAG upregulates lipoprotein expression leading to increased activation of the host innate immune system and virulence in vivo.


Assuntos
Membrana Celular/imunologia , Enterococcus faecalis/imunologia , Glicolipídeos/imunologia , Interações Hospedeiro-Patógeno/imunologia , Lipoproteínas/imunologia , Animais , Proteínas de Bactérias/imunologia , Linhagem Celular , Quimiocina CXCL2/sangue , Feminino , Células HEK293 , Humanos , Imunidade Inata/imunologia , Interleucina-6/sangue , Macrófagos , Lipídeos de Membrana/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Receptor 2 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/imunologia , Virulência/imunologia
7.
BMC Genomics ; 16: 175, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25887115

RESUMO

BACKGROUND: Enterococcus faecalis is a multifaceted microorganism known to act as a beneficial intestinal commensal bacterium. It is also a dreaded nosocomial pathogen causing life-threatening infections in hospitalised patients. Isolates of a distinct MLST type ST40 represent the most frequent strain type of this species, distributed worldwide and originating from various sources (animal, human, environmental) and different conditions (colonisation/infection). Since enterococci are known to be highly recombinogenic we determined to analyse the microevolution and niche adaptation of this highly distributed clonal type. RESULTS: We compared a set of 42 ST40 isolates by assessing key molecular determinants, performing whole genome sequencing (WGS) and a number of phenotypic assays including resistance profiling, formation of biofilm and utilisation of carbon sources. We generated the first circular closed reference genome of an E. faecalis isolate D32 of animal origin and compared it with the genomes of other reference strains. D32 was used as a template for detailed WGS comparisons of high-quality draft genomes of 14 ST40 isolates. Genomic and phylogenetic analyses suggest a high level of similarity regarding the core genome, also demonstrated by similar carbon utilisation patterns. Distribution of known and putative virulence-associated genes did not differentiate between ST40 strains from a commensal and clinical background or an animal or human source. Further analyses of mobile genetic elements (MGE) revealed genomic diversity owed to: (1) a modularly structured pathogenicity island; (2) a site-specifically integrated and previously unknown genomic island of 138 kb in two strains putatively involved in exopolysaccharide synthesis; and (3) isolate-specific plasmid and phage patterns. Moreover, we used different cell-biological and animal experiments to compare the isolate D32 with a closely related ST40 endocarditis isolate whose draft genome sequence was also generated. D32 generally showed a greater capacity of adherence to human cell lines and an increased pathogenic potential in various animal models in combination with an even faster growth in vivo (not in vitro). CONCLUSION: Molecular, genomic and phenotypic analysis of representative isolates of a major clone of E. faecalis MLST ST40 revealed new insights into the microbiology of a commensal bacterium which can turn into a conditional pathogen.


Assuntos
Enterococcus faecalis/genética , Genoma Bacteriano , Animais , Bacteriemia/microbiologia , Aderência Bacteriana , Biofilmes/crescimento & desenvolvimento , Sistemas CRISPR-Cas , Células CACO-2 , Carbono/metabolismo , Enterococcus faecalis/classificação , Enterococcus faecalis/metabolismo , Enterococcus faecalis/patogenicidade , Feminino , Genômica , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Sequências Repetitivas Dispersas , Lepidópteros/microbiologia , Camundongos Endogâmicos BALB C , Fenótipo , Plasmídeos/genética , Análise de Sequência de DNA
8.
PLoS Pathog ; 11(2): e1004653, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25706310

RESUMO

The microbiome and the phage meta-genome within the human gut are influenced by antibiotic treatments. Identifying a novel mechanism, here we demonstrate that bacteria use the universal communication molecule AI-2 to induce virulence genes and transfer them via phage release. High concentrations (i.e. 100 µM) of AI-2 promote dispersal of bacteria from already established biofilms, and is associated with release of phages capable of infecting other bacteria. Enterococcus faecalis V583ΔABC harbours 7 prophages in its genome, and a mutant deficient in one of these prophages (i.e. prophage 5) showed a greatly reduced dispersal of biofilm. Infection of a probiotic E. faecalis strain without lytic prophages with prophage 5 resulted in increased biofilm formation and also in biofilm dispersal upon induction with AI-2. Infection of the probiotic E. faecalis strain with phage-containing supernatants released through AI-2 from E. faecalis V583ΔABC resulted in a strong increase in pathogenicity of this strain. The polylysogenic probiotic strain was also more virulent in a mouse sepsis model and a rat endocarditis model. Both AI-2 and ciprofloxacin lead to phage release, indicating that conditions in the gastrointestinal tract of hospitalized patients treated with antibiotics might lead to distribution of virulence genes to apathogenic enterococci and possibly also to other commensals or even to beneficial probiotic strains.


Assuntos
Biofilmes/crescimento & desenvolvimento , Endocardite Bacteriana/microbiologia , Enterococcus faecalis , Prófagos/fisiologia , Percepção de Quorum , Sepse/microbiologia , Fatores de Virulência/metabolismo , Liberação de Vírus/fisiologia , Animais , Biofilmes/efeitos dos fármacos , Células CACO-2 , Ciprofloxacina/farmacologia , Endocardite Bacteriana/patologia , Enterococcus faecalis/patogenicidade , Enterococcus faecalis/fisiologia , Enterococcus faecalis/virologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Wistar , Sepse/patologia , Liberação de Vírus/efeitos dos fármacos
9.
Microbiol Res ; 172: 88-94, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25497915

RESUMO

Staphylococcus saprophyticus is an important cause of urinary tract infection, and its cell surface hydrophobicity may contribute to virulence by facilitating adherence of the organism to uroepithelia. S. saprophyticus expresses the surface protein SdrI, a member of the serine-aspartate repeat (SD) protein family, which has multifunctional properties. The SdrI knock out mutant has a reduced hydrophobicity index (HPI) of 25%, and expressed in the non-hydrophobic Staphylococcus carnosus strain TM300 causes hydrophobicity. Using hydrophobic interaction chromatography (HIC), we confined the hydrophobic site of SdrI to the N-terminal repeat region. S. saprophyticus strains carrying different plasmid constructs lacking either the N-terminal repeats, both B or SD-repeats were less hydrophobic than wild type and fully complemented SdrI mutant (HPI: 51%). The surface hydrophobicity and HPI of both wild type and the complemented strain were also influenced by calcium (Ca(2+)) and were reduced from 81.3% and 82.4% to 10.9% and 12.3%, respectively. This study confirms that the SdrI protein of S. saprophyticus is a crucial factor for surface hydrophobicity and also gives a first significant functional description of the N-terminal repeats, which in conjunction with the B-repeats form an optimal hydrophobic conformation.


Assuntos
Proteínas de Bactérias/química , Interações Hidrofóbicas e Hidrofílicas , Proteínas de Membrana/química , Estrutura Terciária de Proteína , Staphylococcus saprophyticus/química , Proteínas de Bactérias/genética , Expressão Gênica , Técnicas de Inativação de Genes , Teste de Complementação Genética , Proteínas de Membrana/genética , Staphylococcus saprophyticus/genética
10.
PLoS One ; 9(11): e111880, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25369230

RESUMO

Infections by opportunistic bacteria have significant contributions to morbidity and mortality of hospitalized patients and also lead to high expenses in healthcare. In this setting, one of the major clinical problems is caused by Gram-positive bacteria such as enterococci and staphylococci. In this study we extract, purify, identify and characterize immunogenic surface-exposed proteins present in the vancomycin resistant enterococci (VRE) strain Enterococcus faecium E155 using three different extraction methods: trypsin shaving, biotinylation and elution at high pH. Proteomic profiling was carried out by gel-free and gel-nanoLC-MS/MS analyses. The total proteins found with each method were 390 by the trypsin shaving, 329 by the elution at high pH, and 45 using biotinylation. An exclusively extracytoplasmic localization was predicted in 39 (10%) by trypsin shaving, in 47 (15%) by elution at high pH, and 27 (63%) by biotinylation. Comparison between the three extraction methods by Venn diagram and subcellular localization predictors (CELLO v.2.5 and Gpos-mPLoc) allowed us to identify six proteins that are most likely surface-exposed: the SCP-like extracellular protein, a low affinity penicillin-binding protein 5 (PBP5), a basic membrane lipoprotein, a peptidoglycan-binding protein LysM (LysM), a D-alanyl-D-alanine carboxypeptidase (DdcP) and the peptidyl-prolyl cis-trans isomerase (PpiC). Due to their close relationship with the peptidoglycan, we chose PBP5, LysM, DdcP and PpiC to test their potential as vaccine candidates. These putative surface-exposed proteins were overexpressed in Escherichia coli and purified. Rabbit polyclonal antibodies raised against the purified proteins were able to induce specific opsonic antibodies that mediated killing of the homologous strain E. faecium E155 as well as clinical strains E. faecium E1162, Enterococcus faecalis 12030, type 2 and type 5. Passive immunization with rabbit antibodies raised against these proteins reduced significantly the colony counts of E. faecium E155 in mice, indicating the effectiveness of these surface-related proteins as promising vaccine candidates to target different enterococcal pathogens.


Assuntos
Anticorpos Antibacterianos/sangue , Bacteriemia/prevenção & controle , Enterococcus faecalis/imunologia , Enterococcus faecium/imunologia , Infecções por Bactérias Gram-Positivas/prevenção & controle , Peptidoglicano/imunologia , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/imunologia , Reações Cruzadas , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Vacinação
11.
PLoS One ; 9(10): e110953, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25333799

RESUMO

Lipoteichoic acids (LTA) are amphiphilic polymers that are important constituents of the cell wall of many Gram-positive bacteria. The chemical structures of LTA vary among organisms, albeit in the majority of Gram-positive bacteria the LTAs feature a common poly-1,3-(glycerolphosphate) backbone. Previously, the specificity of opsonic antibodies for this backbone present in some Gram-positive bacteria has been demonstrated, suggesting that this minimal structure may be sufficient for vaccine development. In the present work, we studied a well-defined synthetic LTA-fragment, which is able to inhibit opsonic killing of polyclonal rabbit sera raised against native LTA from Enterococcus faecalis 12030. This promising compound was conjugated with BSA and used to raise rabbit polyclonal antibodies. Subsequently, the opsonic activity of this serum was tested in an opsonophagocytic assay and specificity was confirmed by an opsonophagocytic inhibition assay. The conjugated LTA-fragment was able to induce specific opsonic antibodies that mediate killing of the clinical strains E. faecalis 12030, Enterococcus faecium E1162, and community-acquired Staphylococcus aureus strain MW2 (USA400). Prophylactic immunization with the teichoic acid conjugate and with the rabbit serum raised against this compound was evaluated in active and passive immunization studies in mice, and in an enterococcal endocarditis rat model. In all animal models, a statistically significant reduction of colony counts was observed indicating that the novel synthetic LTA-fragment conjugate is a promising vaccine candidate for active or passive immunotherapy against E. faecalis and other Gram-positive bacteria.


Assuntos
Infecção Hospitalar/imunologia , Lipopolissacarídeos/imunologia , Ácidos Teicoicos/imunologia , Vacinas Conjugadas/imunologia , Vacinas Sintéticas/administração & dosagem , Animais , Anticorpos Antibacterianos/administração & dosagem , Anticorpos Antibacterianos/imunologia , Infecção Hospitalar/microbiologia , Infecção Hospitalar/prevenção & controle , Enterococcus faecium/imunologia , Enterococcus faecium/patogenicidade , Soros Imunes/imunologia , Imunização Passiva , Camundongos , Proteínas Opsonizantes/imunologia , Coelhos , Ratos , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Vacinas Conjugadas/química , Vacinas Sintéticas/imunologia
12.
PLoS One ; 9(10): e107827, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25296179

RESUMO

BACKGROUND: Enterococci are the third most common cause of healthcare-associated infections, which include urinary tract infections, bacteremia and endocarditis. Cell-surface structures such as lipoteichoic acid (LTA) have been poorly examined in E. faecalis, especially with respect to urinary tract infections (UTIs). The dlt operon is responsible for the D-alanylation of LTA and includes the gene dltA, which encodes the D-alanyl carrier protein ligase (Dcl). The involvement of LTA in UTI infection by E. faecalis has not been studied so far. Here, we examined the role of teichoic acid alanylation in the adhesion of enterococci to uroepithelial cells. RESULTS: In a mouse model of urinary tract infection, we showed that E. faecalis 12030ΔdltA mutant colonizes uroepithelial surfaces more efficiently than wild type bacteria. We also demonstrated that this mutant adhered four fold better to human bladder carcinoma cell line T24 compared to the wild type strain. Bacterial adherence could be significantly inhibited by purified lipoteichoic acid (LTA) and inhibition was specific. CONCLUSION: In contrast to bacteraemia model and adherence to colon surfaces, E. faecalis 12030ΔdltA mutant colonized uroepithelial surfaces more efficiently than wild-type bacteria. In the case of the uroepithelial surface the adherence to specific host cells could be prevented by purified LTA. Our results therefore suggest a novel function of alanylation of LTA in E. faecalis.


Assuntos
Enterococcus faecalis/metabolismo , Enterococcus faecalis/patogenicidade , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Ácidos Teicoicos/metabolismo , Ácidos Teicoicos/farmacologia , Infecções Urinárias/metabolismo , Animais , Aderência Bacteriana/efeitos dos fármacos , Linhagem Celular Tumoral , Enterococcus faecalis/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C
13.
PLoS One ; 9(5): e96295, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24806450

RESUMO

BACKGROUND: After uropathogenic Escherichia coli (UPEC), Enterococcus faecalis is the second most common pathogen causing urinary tract infections. Monoglucosyl-diacylglycerol (MGlcDAG) and diglucosyl-diacylglycerol (DGlcDAG) are the main glycolipids of the E. faecalis cell membrane. Examination of two mutants in genes bgsB and bgsA (both glycosyltransferases) showed that these genes are involved in cell membrane glycolipid biosynthesis, and that their inactivation leads to loss of glycolipids DGlcDAG (bgsA) or both MGlcDAG and DGlcDAG (bgsB). Here we investigate the function of bgsB and bgsA regarding their role in the pathogenesis in a mouse model of urinary tract infection and in bacterial adhesion to T24 bladder epithelial cells. RESULTS: In a mouse model of urinary tract infection, we showed that E. faecalis 12030ΔbgsB and E. faecalis 12030ΔbgsA mutants, colonize uroepithelial surfaces more efficiently than wild-type bacteria. We also demonstrated that these mutants showed a more than three-fold increased binding to human bladder carcinoma cells line T24 compared to the wild-type strain. Bacterial binding could be specifically inhibited by purified glycolipids. Lipoteichoic acid (LTA), wall-teichoic acid (WTA), and glycosaminoglycans (GAGs) were not significantly involved in binding of E. faecalis to the bladder epithelial cell line. CONCLUSIONS: Our data show that the deletion of bgsB and bgsA and the absence of the major glycolipid diglucosyl-diacylglycerol increases colonization and binding to uroepithelial cells. We hypothesize that secreted diglucosyl-diacylglycerol blocks host binding sites, thereby preventing bacterial adhesion. Further experiments will be needed to clarify the exact mechanism underlying the adhesion through glycolipids and their cognate receptors.


Assuntos
Enterococcus faecalis/metabolismo , Enterococcus faecalis/fisiologia , Glicolipídeos/metabolismo , Glicolipídeos/farmacologia , Infecções Urinárias/microbiologia , Animais , Aderência Bacteriana/efeitos dos fármacos , Linhagem Celular Tumoral , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/genética , Feminino , Glicolipídeos/genética , Glicosaminoglicanos/farmacologia , Humanos , Lipopolissacarídeos/farmacologia , Camundongos , Ácidos Teicoicos/farmacologia
14.
PLoS One ; 9(3): e91863, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24637922

RESUMO

Enterococci are among the major pathogens implicated in cardiac infections and biofilm formation. E. faecalis has been shown to play an important role in infectious endocarditis. Several distinct mechanisms for biofilm formation have been identified in E. faecalis. Our group has previously characterized two distinct bacterial glucosyltransferases playing key roles in the production of the major cell wall glycolipids and leading to reduced biofilm production. To assess if this mechanism is involved in the pathogenesis of enterococcal endocarditis we compared the wild-type strain of E. faecalis 12030 with two mutants in gene EF2891 and EF2890 respectively in a rat model of infective endocarditis. The results showed less endocarditic lesions and reduced colony counts per vegetation in the two mutants. indicating that the modification of bacterial surface lipids results in significantly reduced virulence in infective endocarditis. These results underscore the important role of biofilm formation in the pathogenicity of enterococcal endocarditis and may indicate an interesting target for novel therapeutic strategies.


Assuntos
Parede Celular/metabolismo , Endocardite Bacteriana/microbiologia , Enterococcus faecalis/genética , Enterococcus faecalis/metabolismo , Glicolipídeos/metabolismo , Mutação , Animais , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Enterococcus faecalis/patogenicidade , Feminino , Ratos , Virulência
15.
PLoS One ; 7(6): e38458, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22723861

RESUMO

BACKGROUND: Enterococcus faecalis is one of the leading causes of nosocomial infections. Due to its innate and acquired resistance to most antibiotics, identification of new targets for antimicrobial treatment of E. faecalis is a high priority. The multiple peptide resistance factor MprF, which was first described in Staphylococcus aureus, modifies phosphatidylglycerol with lysin and reduces the negative charge of the membrane, thus increasing resistance to cationic antimicrobial peptides. We studied the effect of mprF in E. faecalis regarding influence on bacterial physiology and virulence. RESULTS: Two putative mprF paralogs (mprF1 and mprF2) were identified in E. faecalis by BLAST search using the well-described S. aureus gene as a lead. Two deletion mutants in E. faecalis 12030 were created by homologous recombination. Analysis of both mutants by thin-layer chromatography showed that inactivation of mprF2 abolishes the synthesis of three distinct amino-phosphatidylglycerols (PGs). In contrast, deletion of mprF1 did not interfere with the biosynthesis of amino-PG. Inactivation of mprF2 increased susceptibility against several antimicrobial peptides and resulted in a 42% increased biofilm formation compared to wild-type mprF. However, resistance to opsonic killing was increased in the mutant, while virulence in a mouse bacteremia model was unchanged. CONCLUSION: Our data suggest that only mprF2 is involved in the aminoacylation of PG in enterococci, and is probably responsible for synthesis of Lys-PG, Ala-PG, and Arg-PG, while mprF1 does not seem to have a role in aminoacylation. As in other Gram-positive pathogens, aminoacylation through MprF2 increases resistance against cationic antimicrobial peptides. Unlike mprF found in other bacteria, mprF2 does not seem to be a major virulence factor in enterococci.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Enterococcus faecalis/patogenicidade , Infecções por Bactérias Gram-Positivas/microbiologia , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Biofilmes/crescimento & desenvolvimento , Farmacorresistência Bacteriana/genética , Enterococcus faecalis/genética , Enterococcus faecalis/crescimento & desenvolvimento , Deleção de Genes , Camundongos , Fosfolipídeos/metabolismo , Análise de Sequência de DNA , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
16.
J Infect Dis ; 205(7): 1076-85, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22362863

RESUMO

Type 1 lipoteichoic acid (LTA) is present in many clinically important gram-positive bacteria, including enterococci, streptococci, and staphylococci, and antibodies against LTA have been shown to opsonize nonencapsulated Enterococcus faecalis strains. In the present study, we show that antibodies against E. faecalis LTA also bind to type 1 LTA from other gram-positive species and opsonized Staphylocccus epidermidis and Staphylcoccus aureus strains as well as group B streptococci. Inhibition studies using teichoic acid oligomers indicated that cross-reactive opsonic antibodies bind to the teichoic acid backbone. Passive immunization with rabbit antibodies against E. faecalis LTA promoted the clearance of bacteremia by E. faecalis and S. epidermidis in mice. Furthermore, passive protection also reduced mortality in a murine S. aureus peritonitis model. The effectiveness of rabbit antibody against LTA suggests that this conserved bacterial structure could function as a single vaccine antigen that targets multiple gram-positive pathogens.


Assuntos
Anticorpos Antibacterianos/administração & dosagem , Glicerofosfatos/imunologia , Imunização Passiva/métodos , Lipopolissacarídeos/imunologia , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/imunologia , Ácidos Teicoicos/imunologia , Adulto , Animais , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/imunologia , Bacteriemia/prevenção & controle , Modelos Animais de Doenças , Enterococcus faecalis/imunologia , Feminino , Experimentação Humana , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Opsonizantes/imunologia , Peritonite/prevenção & controle , Fagocitose , Coelhos , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus epidermidis/imunologia , Streptococcus agalactiae/imunologia , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...