Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Glycoconj J ; 36(5): 429-438, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31230165

RESUMO

Enterococcus faecium (E. faecium) has emerged as one of today's leading causes of health care-associated infections that is difficult to treat with the available antibiotics. These pathogens produce capsular polysaccharides on the cell surface which play a significant role in adhesion, virulence and evasion. Therefore, we aimed at the identification and characterization of bacterial polysaccharide antigens which are central for the development of vaccine-based prophylactic approaches. The crude cell wall-associated polysaccharides from E. faecium, its mutant and complemented strains were purified and analyzed by a primary antibody raised against lipoteichoic acid (LTA) and diheteroglycan (DHG). The resistant E. faecium strains presumably possess novel capsular polysaccharides that allow them to avoid the evasion from opsonic killing. The E. faecium U0317 strain was very well opsonized by anti-U0317 (~95%), an antibody against the whole bacterial cell. The deletion mutant showed a significantly increased susceptibility to opsonophagocytic killing (90-95%) against the penicillin binding protein (anti-PBP-5). By comparison, in a mouse urinary tract and rat endocarditis infection model, respectively, there were no significant differences in virulence. In this study we explored the biological role of the capsule of E. faecium. Our findings showed that the U0317 strain is not only sensitive to anti-LTA but also to antibodies against other enterococcal surface proteins. Our findings demonstrate that polysaccharides capsule mediated-resistance to opsonophagocytosis. We also found that the capsular polysaccharides do not play an important role in bacterial virulence in urinary tract and infective endocarditis in vivo models.


Assuntos
Anticorpos Antibacterianos/farmacologia , Antígenos de Bactérias/isolamento & purificação , Parede Celular/química , Enterococcus faecium/química , Lipopolissacarídeos/isolamento & purificação , Polissacarídeos Bacterianos/isolamento & purificação , Ácidos Teicoicos/isolamento & purificação , Animais , Antibacterianos/farmacologia , Anticorpos Antibacterianos/biossíntese , Antígenos de Bactérias/química , Antígenos de Bactérias/imunologia , Cápsulas Bacterianas/química , Cápsulas Bacterianas/imunologia , Parede Celular/imunologia , Modelos Animais de Doenças , Farmacorresistência Bacteriana , Endocardite Bacteriana/tratamento farmacológico , Endocardite Bacteriana/microbiologia , Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/imunologia , Enterococcus faecium/patogenicidade , Feminino , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Lipopolissacarídeos/química , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Opsonizantes/farmacologia , Proteínas de Ligação às Penicilinas/química , Proteínas de Ligação às Penicilinas/imunologia , Proteínas de Ligação às Penicilinas/isolamento & purificação , Proteínas de Ligação às Penicilinas/farmacologia , Fagocitose/efeitos dos fármacos , Polissacarídeos Bacterianos/química , Polissacarídeos Bacterianos/imunologia , Cultura Primária de Células , Ratos , Ratos Wistar , Ácidos Teicoicos/química , Ácidos Teicoicos/imunologia , Ácidos Teicoicos/farmacologia , Infecções Urinárias/tratamento farmacológico , Infecções Urinárias/microbiologia
2.
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
3.
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
4.
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
5.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...