Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
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
2.
mSphere ; 4(4)2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31292230

RESUMEN

Commensal and generally harmless in healthy individuals, Enterococcus faecalis causes opportunistic infections in immunocompromised patients. Plasmid-cured E. faecalis strain VE14089, derived from sequenced reference strain V583, is widely used for functional studies due to its improved genetic amenability. Although strain VE14089 has no major DNA rearrangements, with the exception of an ∼20-kb integrated region of pTEF1 plasmid, the strain presented significant growth differences from the V583 reference strain of our collection (renamed VE14002). In the present study, genome sequencing of strain VE14089 identified additional point mutations. Excision of the integrated pTEF1 plasmid region and sequential restoration of wild-type alleles showing nonsilent mutations were performed to obtain the VE18379 reference-derivative strain. Recovery of the growth ability of the restored VE18379 strain at a level similar to that seen with the reference strain points to GreA and Spx as bacterial fitness determinants. Virulence potential in Galleria mellonella and intestinal colonization in mouse demonstrated host adaptation of the VE18379 strain equivalent to VE14002 host adaptation. We further demonstrated that deletion of the 16.8-kb variable region of the epa locus recapitulates the key role of Epa decoration in host adaptation, providing a genetic system to study the role of specific epa-variable regions in host adaptation independently of other genetic variations.IMPORTANCEE. faecalis strain VE14089 was derived from V583 cured of its plasmids. Although VE14089 had no major DNA rearrangements, it presented significant growth and host adaptation differences from the reference strain V583 of our collection. To construct a strain with better fitness, we sequenced the genome of VE14089, identified single nucleotide polymorphisms (SNPs), and repaired the genes that could account for these changes. Using this reference-derivative strain, we provide a novel genetic system to understand the role of the variable region of epa in the enterococcal lifestyle.


Asunto(s)
Proteínas Bacterianas/genética , Enterococcus faecalis/genética , Aptitud Genética , Polisacáridos Bacterianos/genética , Animales , Enterococcus faecalis/patogenicidad , Genoma Bacteriano , Larva/microbiología , Ratones , Mariposas Nocturnas/microbiología , Fenotipo , Mutación Puntual , Polimorfismo de Nucleótido Simple , Virulencia , Factores de Virulencia/genética , Secuenciación Completa del Genoma
3.
Sci Rep ; 9(1): 8926, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31222056

RESUMEN

Enterococci are subdominant members of the human gastrointestinal microbiota. Enterococcus faecalis is generally harmless for healthy individuals, but it can cause a diverse range of infections in immunodeficient or elderly patients with severe underlying diseases. In this study, we analysed the levels of intestinal translocation of indigenous enterococci in C57BL/6, CF-1 and CX3CR1-/- mice upon clindamycin antibiotic-induced dysbiosis. We found that C57BL/6 was the most permissive model for enterococcal translocation and that initiation of E. faecalis translocation coincided with a threshold of enterococcal colonisation in the gut lumen, which once reached, triggered E. faecalis dissemination to deeper organs. We showed that the extent to which E. faecalis clinical strain VE14821 competed with indigenous enterococci differed between the C57BL/6 and CX3CR1-/- models. Finally, using a simplified gnotobiotic model, we observed E. faecalis crossing an intact intestinal tract using intestinal epithelial cells as one route to reach the lamina propria. Our study opens new perspectives for assessing the effect of various immunodeficiencies and for investigating mechanisms underlying enterococcal translocation.


Asunto(s)
Enterococcus/crecimiento & desarrollo , Microbioma Gastrointestinal , Animales , Transporte Biológico , Receptor 1 de Quimiocinas CX3C/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
World J Microbiol Biotechnol ; 35(6): 85, 2019 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-31134456

RESUMEN

Surface properties like hydrophobicity, aggregation ability, adhesion to mucosal surfaces and epithelial cells and transit time are key features for the characterization of probiotic strains. In this study, we used two Lactobacillus paracasei subsp. paracasei strains (BGNJ1-64 and BGSJ2-8) strains which were previously described with very strong aggregation capacity. The aggregation promoting factor (AggLb) expressed in these strains showed high level of binding to collagen and fibronectin, components of extracellular matrix. The working hypothesis was that strains able to aggregate have an advantage to resist in intestinal tract. So, we assessed whether these strains and their derivatives (without aggLb gene) are able to bind or not to intestinal components and we compared the transit time of each strains in mice. In that purpose parental strains (BGNJ1-64 and BGSJ2-8) and their aggregation negative derivatives (BGNJ1-641 and BGSJ2-83) were marked with double antibiotic resistance in order to be tracked in in vivo experiments in mice. Comparative analysis of binding ability of WT and aggregation negative strains to different human intestinal cell lines and mucin revealed no significant difference among them, excluding involvement of AggLb in interaction with surface of intestinal cells and mucin. In vivo experiments showed that surviving and transit time of marked strains in mice did not drastically depend on the presence of the AggLb aggregation factor.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/microbiología , Intestinos/microbiología , Lacticaseibacillus paracasei/crecimiento & desarrollo , Lacticaseibacillus paracasei/fisiología , Unión Proteica , Animales , Adhesión Bacteriana/fisiología , Células CACO-2 , Moléculas de Adhesión Celular/fisiología , Colágeno/metabolismo , Fibronectinas/metabolismo , Células HT29 , Interacciones Microbiota-Huesped/fisiología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Masculino , Ratones , Ratones Endogámicos C57BL , Mucinas/metabolismo , Probióticos , Análisis de la Onda del Pulso , Propiedades de Superficie
5.
Sci Rep ; 8(1): 5098, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29572473

RESUMEN

Enterococci, in particular vancomycin-resistant enterococci (VRE), are a leading cause of hospital-acquired infections. Promoting intestinal resistance against enterococci could reduce the risk of VRE infections. We investigated the effects of two Lactobacillus strains to prevent intestinal VRE. We used an intestinal colonisation mouse model based on an antibiotic-induced microbiota dysbiosis to mimic enterococci overgrowth and VRE persistence. Each Lactobacillus spp. was administered daily to mice starting one week before antibiotic treatment until two weeks after antibiotic and VRE inoculation. Of the two strains, Lactobacillus paracasei CNCM I-3689 decreased significantly VRE numbers in the feces demonstrating an improvement of the reduction of VRE. Longitudinal microbiota analysis showed that supplementation with L. paracasei CNCM I-3689 was associated with a better recovery of members of the phylum Bacteroidetes. Bile salt analysis and expression analysis of selected host genes revealed increased level of lithocholate and of ileal expression of camp (human LL-37) upon L. paracasei CNCM I-3689 supplementation. Although a direct effect of L. paracasei CNCM I-3689 on the VRE reduction was not ruled out, our data provide clues to possible anti-VRE mechanisms supporting an indirect anti-VRE effect through the gut microbiota. This work sustains non-antibiotic strategies against opportunistic enterococci after antibiotic-induced dysbiosis.


Asunto(s)
Bacteroidetes/fisiología , Lacticaseibacillus paracasei/fisiología , Probióticos/administración & dosificación , Enterococos Resistentes a la Vancomicina/fisiología , Animales , Antibacterianos/farmacología , Bacteroidetes/efectos de los fármacos , Clindamicina/farmacología , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Intestinos/microbiología , Masculino , Ratones , Probióticos/farmacología , Enterococos Resistentes a la Vancomicina/efectos de los fármacos
6.
Environ Microbiol ; 19(9): 3579-3594, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28695648

RESUMEN

Bacterial adhesion is a critical step for colonization of the host. The pioneer colonizer and commensal bacterium of the human gastrointestinal tract, Streptococcus salivarius, has strong adhesive properties but the molecular determinants of this adhesion remain uncharacterized. Serine-rich repeat (SRR) glycoproteins are a family of adhesins that fulfil an important role in adhesion. In general, Gram-positive bacterial genomes have a unique SRR glycoprotein-encoding gene. We demonstrate that S. salivarius expresses three large and glycosylated surface-exposed proteins - SrpA, SrpB and SrpC - that show characteristics of SRR glycoproteins and are secreted through the accessory SecA2/Y2 system. Two glycosyltransferases - GtfE/F - encoded outside of the secA2/Y2 locus, unusually, perform the first step of the sequential glycosylation process, which is crucial for SRR activity. We show that SrpB and SrpC play complementary adhesive roles involved in several steps of the colonization process: auto-aggregation, biofilm formation and adhesion to a variety of host epithelial cells and components. We also show that at least one of the S. salivarius SRR glycoproteins is important for colonization in mice. SrpA, SrpB and SrpC are the main factors underlying the multifaceted adhesion of S. salivarius and, therefore, play a major role in host colonization.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana/fisiología , Proteínas Bacterianas/metabolismo , Mucosa Intestinal/microbiología , Glicoproteínas de Membrana/metabolismo , Streptococcus salivarius/patogenicidad , Animales , Adhesión Bacteriana/genética , Células Epiteliales/microbiología , Tracto Gastrointestinal/microbiología , Glucosiltransferasas/genética , Glicosilación , Humanos , Masculino , Ratones , Modelos Animales , Streptococcus salivarius/genética , Streptococcus salivarius/metabolismo
7.
BMC Microbiol ; 15: 112, 2015 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-26003173

RESUMEN

BACKGROUND: Mechanisms underlying the transition from commensalism to virulence in Enterococcus faecalis are not fully understood. We previously identified the enterococcal leucine-rich protein A (ElrA) as a virulence factor of E. faecalis. The elrA gene is part of an operon that comprises four other ORFs encoding putative surface proteins of unknown function. RESULTS: In this work, we compared the susceptibility to phagocytosis of three E. faecalis strains, including a wild-type (WT), a ΔelrA strain, and a strain overexpressing the whole elr operon in order to understand the role of this operon in E. faecalis virulence. While both WT and ΔelrA strains were efficiently phagocytized by RAW 264.7 mouse macrophages, the elr operon-overexpressing strain showed a decreased capability to be internalized by the phagocytic cells. Consistently, the strain overexpressing elr operon was less adherent to macrophages than the WT strain, suggesting that overexpression of the elr operon could confer E. faecalis with additional anti-adhesion properties. In addition, increased virulence of the elr operon-overexpressing strain was shown in a mouse peritonitis model. CONCLUSIONS: Altogether, our results indicate that overexpression of the elr operon facilitates the E. faecalis escape from host immune defenses.


Asunto(s)
Proteínas Bacterianas/genética , Enterococcus faecalis/fisiología , Operón , Peritonitis/microbiología , Fagocitosis , Animales , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Enterococcus faecalis/genética , Enterococcus faecalis/patogenicidad , Regulación Bacteriana de la Expresión Génica , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/veterinaria , Macrófagos/metabolismo , Ratones , Virulencia
8.
FEMS Microbiol Lett ; 362(8): fnv012, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25667231

RESUMEN

Vancomycin-resistant enterococci (VRE) represent major opportunistic pathogens in immunocompromised populations. Particularly adapted to the hospital environment, VRE efficiently colonize the gastrointestinal (GI) tract of patients. Furthermore, certain circumstances of antibiotic-induced dysbiosis of the gut microbiota contribute to colonization, overgrowth and persistence of VRE in the GI tract, increasing the risk of infection in critically ill and/or severally immunocompromised patients. VRE treatment with antibiotics remains challenging due to the robustness and ability of enterococci to adapt to harsh conditions and to acquire novel resistance genes. Reducing VRE intestinal colonization, overgrowth and carriage has thus become an important issue to reduce the risk of infection and dissemination. This review summarizes the knowledge of the conditions favoring VRE colonization and persistence in the GI tract and focuses on the strategies to reduce overgrowth or persistence of VRE in the GI tract based on the oral administration of probiotic or commensal bacteria in human studies and in animal models, and on the potential underlying mechanisms of the anti-VRE effect.


Asunto(s)
Bacterias/crecimiento & desarrollo , Tracto Gastrointestinal/microbiología , Probióticos/administración & dosificación , Simbiosis , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Animales , Antibacterianos/efectos adversos , Modelos Animales de Enfermedad , Humanos , Enterococos Resistentes a la Vancomicina/patogenicidad
9.
J Infect Dis ; 211(1): 62-71, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25035517

RESUMEN

Enterococcus faecalis is a commensal bacterium of the human intestine and a major opportunistic pathogen in immunocompromised and elderly patients. The pathogenesis of E. faecalis infection relies in part on its capacity to colonize the gut. Following disruption of intestinal homeostasis, E. faecalis can overgrow, cross the intestinal barrier, and enter the lymph and bloodstream. To identify and characterize E. faecalis genes that are key to intestinal colonization, our strategy consisted in screening mutants for the following phenotypes related to intestinal lifestyle: antibiotic resistance, overgrowth, and competition against microbiota. From the identified colonization genes, epaX encodes a glycosyltransferase located in a variable region of the enterococcal polysaccharide antigen (epa) locus. We demonstrated that EpaX acts on sugar composition, promoting resistance to bile salts and cell wall integrity. Given that EpaX is enriched in hospital-adapted isolates, this study points to the importance of the epa variability as a key determinant for enterococcal intestinal colonization.


Asunto(s)
Antígenos de Superficie/metabolismo , Enterococcus faecalis/crecimiento & desarrollo , Enterococcus faecalis/metabolismo , Intestinos/microbiología , Polisacáridos/metabolismo , Animales , Antígenos de Superficie/genética , Ácidos y Sales Biliares/genética , Ácidos y Sales Biliares/metabolismo , Pared Celular/genética , Pared Celular/metabolismo , Farmacorresistencia Microbiana , Enterococcus faecalis/genética , Enterococcus faecalis/patogenicidad , Genes Bacterianos , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Infecciones por Bacterias Grampositivas/microbiología , Masculino , Ratones , Microbiota/genética , Polisacáridos/genética , Ramnosa/metabolismo
10.
PLoS Genet ; 9(6): e1003539, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23754962

RESUMEN

Polylysogeny is frequently considered to be the result of an adaptive evolutionary process in which prophages confer fitness and/or virulence factors, thus making them important for evolution of both bacterial populations and infectious diseases. The Enterococcus faecalis V583 isolate belongs to the high-risk clonal complex 2 that is particularly well adapted to the hospital environment. Its genome carries 7 prophage-like elements (V583-pp1 to -pp7), one of which is ubiquitous in the species. In this study, we investigated the activity of the V583 prophages and their contribution to E. faecalis biological traits. We systematically analyzed the ability of each prophage to excise from the bacterial chromosome, to replicate and to package its DNA. We also created a set of E. faecalis isogenic strains that lack from one to all six non-ubiquitous prophages by mimicking natural excision. Our work reveals that prophages of E. faecalis V583 excise from the bacterial chromosome in the presence of a fluoroquinolone, and are able to produce active phage progeny. Intricate interactions between V583 prophages were also unveiled: i) pp7, coined EfCIV583 for E. faecalis chromosomal island of V583, hijacks capsids from helper phage 1, leading to the formation of distinct virions, and ii) pp1, pp3 and pp5 inhibit excision of pp4 and pp6. The hijacking exerted by EfCIV583 on helper phage 1 capsids is the first example of molecular piracy in Gram positive bacteria other than staphylococci. Furthermore, prophages encoding platelet-binding-like proteins were found to be involved in adhesion to human platelets, considered as a first step towards the development of infective endocarditis. Our findings reveal not only a role of E. faecalis V583 prophages in pathogenicity, but also provide an explanation for the correlation between antibiotic usage and E. faecalis success as a nosocomial pathogen, as fluoriquinolone may provoke release of prophages and promote gene dissemination among isolates.


Asunto(s)
Enterococcus faecalis/genética , Interacciones Huésped-Patógeno/genética , Profagos/genética , Factores de Virulencia/genética , Activación Viral/genética , Cromosomas Bacterianos/genética , Infección Hospitalaria/genética , Enterococcus faecalis/patogenicidad , Genoma Bacteriano , Humanos , Profagos/metabolismo , Profagos/patogenicidad , Factores de Virulencia/metabolismo
11.
ISME J ; 7(7): 1256-61, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23677008

RESUMEN

The healthy intestine is characterized by a low level of oxygen and by the presence of large bacterial communities of obligate anaerobes. Dysbiosis of the gut microbiota has been reported in patients suffering from inflammatory bowel diseases (IBDs), but the mechanisms causing this imbalance remain unknown. Observations have included a decrease in obligate anaerobes of the phylum Firmicutes and an increase in facultative anaerobes, including members of the family Enterobacteriaceae. The shift of bacterial communities from obligate to facultative anaerobes strongly suggests a disruption in anaerobiosis and points to a role for oxygen in intestinal dysbiosis. Proposals to evaluate this hypothesis of a role for oxygen in IBD dysbiosis are provided. If this hypothesis is confirmed, decreasing oxygen in the intestine could open novel means to rebalance the microbiota and could provide novel preventative or therapeutic strategies for IBD patients in whom current treatments are ineffective.


Asunto(s)
Disbiosis/microbiología , Enfermedades Inflamatorias del Intestino/microbiología , Intestinos/microbiología , Bacterias Anaerobias/fisiología , Disbiosis/patología , Enterobacteriaceae/crecimiento & desarrollo , Enterobacteriaceae/metabolismo , Enterobacteriaceae/fisiología , Bacterias Grampositivas/fisiología , Humanos , Enfermedades Inflamatorias del Intestino/patología , Intestinos/química , Oxígeno/metabolismo
12.
PLoS One ; 6(12): e29023, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22194979

RESUMEN

Spread of antibiotic resistance among bacteria responsible for nosocomial and community-acquired infections urges for novel therapeutic or prophylactic targets and for innovative pathogen-specific antibacterial compounds. Major challenges are posed by opportunistic pathogens belonging to the low GC% gram-positive bacteria. Among those, Enterococcus faecalis is a leading cause of hospital-acquired infections associated with life-threatening issues and increased hospital costs. To better understand the molecular properties of enterococci that may be required for virulence, and that may explain the emergence of these bacteria in nosocomial infections, we performed the first large-scale functional analysis of E. faecalis V583, the first vancomycin-resistant isolate from a human bloodstream infection. E. faecalis V583 is within the high-risk clonal complex 2 group, which comprises mostly isolates derived from hospital infections worldwide. We conducted broad-range screenings of candidate genes likely involved in host adaptation (e.g., colonization and/or virulence). For this purpose, a library was constructed of targeted insertion mutations in 177 genes encoding putative surface or stress-response factors. Individual mutants were subsequently tested for their i) resistance to oxidative stress, ii) antibiotic resistance, iii) resistance to opsonophagocytosis, iv) adherence to the human colon carcinoma Caco-2 epithelial cells and v) virulence in a surrogate insect model. Our results identified a number of factors that are involved in the interaction between enterococci and their host environments. Their predicted functions highlight the importance of cell envelope glycopolymers in E. faecalis host adaptation. This study provides a valuable genetic database for understanding the steps leading E. faecalis to opportunistic virulence.


Asunto(s)
Membrana Celular/metabolismo , Enterococcus faecalis/genética , Biblioteca de Genes , Marcación de Gen , Pruebas Genéticas , Mutación/genética , Factores de Virulencia/metabolismo , Animales , Antibacterianos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Células CACO-2 , Membrana Celular/efectos de los fármacos , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/crecimiento & desarrollo , Enterococcus faecalis/patogenicidad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Genes Bacterianos/genética , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Modelos Animales , Modelos Biológicos , Mariposas Nocturnas/efectos de los fármacos , Mariposas Nocturnas/microbiología , Proteínas Opsoninas/metabolismo , Fagocitosis/efectos de los fármacos , Fenotipo , Plásmidos/genética , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética , Virulencia/efectos de los fármacos , Virulencia/genética , Factores de Virulencia/genética
13.
Microbiology (Reading) ; 155(Pt 11): 3564-3571, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19696101

RESUMEN

Despite the existence of various virulence factors in the Enterococcus genus, enterococcal virulence is still a debated issue. A main consideration is the detection of the same virulence genes in strains isolated from nosocomial or community-acquired infections, and from food products. The goal of this study was to evaluate the roles of two well-characterized enterococcal virulence factors, Fsr and gelatinase, in the potential virulence of Enterococcus faecalis food strains. Virulence of unrelated Enterococcus isolates, including dairy strains carrying fsr and gelE operons, was compared in the Galleria mellonella insect model. E. faecalis dairy strains were able to kill larvae and were as virulent as strain OG1RF, one of the most widely used for virulence studies. In contrast, Enterococcus durans and Enterococcus faecium strains were avirulent or poorly virulent for G. mellonella. To evaluate the role of fsrB and gelE in virulence of E. faecalis dairy strains, both genes were deleted independently in two strains. The Delta fsrB and Delta gelE deletion mutants both produced a gelatinase-negative phenotype. Although both mutations significantly attenuated virulence in G. mellonella, the Delta fsrB strains were more strongly attenuated. These results agree with previous findings suggesting the involvement of fsrB in the control of other cell functions relevant to virulence. Our work demonstrates that the presence of functional fsrB, and to a lesser extent gelE, in dairy enterococci should be considered with caution.


Asunto(s)
Proteínas Bacterianas/genética , Enterococcus faecalis/patogenicidad , Gelatinasas/genética , Factores de Virulencia/genética , Animales , ADN Bacteriano/genética , Enterococcus faecalis/enzimología , Enterococcus faecalis/genética , Eliminación de Gen , Mariposas Nocturnas/microbiología , Operón , Análisis de Secuencia de ADN , Virulencia
14.
Can J Microbiol ; 54(8): 660-7, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18772928

RESUMEN

Lactobacillus casei DN-114 001 is a probiotic strain able to interact with the immune system and to interfere with gastrointestinal pathogens. The derived strain DN-114 001Rif was studied during its transit through the upper and distal intestine of human volunteers. Seven volunteers participated in the study, which involved intestinal intubation to sample ileal contents and collection of fecal samples, with a wash-out period of 8 days between the 2 steps. The retrieval of the probiotic was analyzed in the ileum every 2 h for 8 h following the ingestion of one dose of the test product and in the feces prior to, during, and after daily consumption of the test product for 8 days. Persistence of the probiotic amplifiable DNA was assessed using temporal temperature gradient gel electrophoresis and real-time PCR. Fluorescent in situ hybridization allowed analysis of the composition of the dominant digestive microbiota. The ingestion of L. casei DN-114 001Rif led to a significant and transient increase of its amplifiable DNA in ileal and fecal samples. This is related to a high stability in the composition of dominant groups of the gut microbiota. Data from ileal samples are scarce and our study confirms the potentiality for interaction between probiotics and the human immune system.


Asunto(s)
Productos Lácteos Cultivados/metabolismo , Heces/microbiología , Microbiología de Alimentos , Íleon/microbiología , Lacticaseibacillus casei/aislamiento & purificación , Probióticos/administración & dosificación , Adulto , Productos Lácteos Cultivados/microbiología , ADN Bacteriano/genética , Heces/química , Femenino , Humanos , Lacticaseibacillus casei/genética , Masculino , Reacción en Cadena de la Polimerasa
15.
Int J Food Microbiol ; 125(2): 176-81, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18554738

RESUMEN

The objective of this study was to determine i) if Camembert cheese micro-organisms could be detected in fecal samples after regular consumption by human subjects and ii) the consequence of this consumption on global metabolic activities of the host colonic microbiota. An open human protocol was designed where 12 healthy volunteers were included: a 2-week period of fermented products exclusion followed by a 4-weeks Camembert ingestion period where 2x40 g/day of Camembert cheese was consumed. Stools were collected from the volunteers before consumption, twice during the ingestion period (2nd and 4th week) and once after a wash out period of 2 weeks. During the consumption of Camembert cheese, high levels of Lactococcus lactis and Leuconostoc mesenteroides were measured in fecal samples using real-time quantitative PCR, reaching median values of 8.2 and 7.5 Log(10) genome equivalents/g of stool. For Ln. mesenteroides, persistence was observed 15 days after the end of Camembert consumption. The survival of Geotrichum candidum was also assessed and the fecal concentration reached a median level of 7.1 Log(10) CFU/g in stools. Except a decreasing trend of the nitrate reductase activity, no significant modification was shown in the metabolic activities during this study.


Asunto(s)
Queso/microbiología , Colon/microbiología , Heces/microbiología , Lactobacillus/crecimiento & desarrollo , Streptococcus thermophilus/crecimiento & desarrollo , Adulto , Recuento de Colonia Microbiana , Estudios Cruzados , ADN Bacteriano/química , ADN Bacteriano/genética , ADN de Hongos/química , ADN de Hongos/genética , Femenino , Microbiología de Alimentos , Geotrichum/crecimiento & desarrollo , Geotrichum/aislamiento & purificación , Geotrichum/metabolismo , Humanos , Lactobacillus/aislamiento & purificación , Lactobacillus/metabolismo , Masculino , Nitrato-Reductasa/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Streptococcus thermophilus/aislamiento & purificación , Streptococcus thermophilus/metabolismo , Factores de Tiempo
16.
J Pediatr Gastroenterol Nutr ; 46(5): 580-8, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18493215

RESUMEN

OBJECTIVE: To test the safety and effect on faecal microbiota of a formula with prebiotic oligosaccharides alone or in combination with acidic oligosaccharides in infants at the age of partial formula feeding. PATIENTS AND METHODS: The study was a double-blind, placebo-controlled, randomised intervention trial in which 82 healthy, full-term, partially breast-fed children, from 1 week to 3 months old, were given 1 of the following formulae: whey-based formula (control group), whey-based formula with galacto- and long-chain fructo-oligosaccharides (scGOS/lcFOS group), or whey-based formula with galacto- and long-chain fructo-oligosaccharides added with pectin-derived acidic oligosaccharides (scGOS/lcFOS/pAOS group). Children were studied for the duration of the partial formula feeding period and every 2 weeks for 2 months after breast-feeding cessation. The total bacteria count and the proportion of 7 bacterial families were determined using in situ hybridisation coupled to flow cytometry. RESULTS: The total bacterial count did not alter with time or type of feeding (9.9 +/- 0.1 log10 cells per gram wet weight). Compared with the control group, there was an increase of the Bifidobacterium genus (P = 0.0001), and a decrease of proportions for the Bacteroides group (P = 0.02) and the Clostridium coccoides group (P = 0.01) in both oligosaccharide groups. The proportion of bifidobacteria was significantly higher in the scGOS/lcFOS/pAOS compared with the scGOS/lcFOS group (P < 0.01). CONCLUSIONS: Infant formulae appear to be clinically safe and effective on infant microbiota. They minimize the alteration of faecal microbiota after cessation of breast-feeding and promote bifidobacteria proportions, with a stronger effect when acidic oligosaccharides are present.


Asunto(s)
Bifidobacterium/crecimiento & desarrollo , Heces/microbiología , Fórmulas Infantiles , Fenómenos Fisiológicos Nutricionales del Lactante/fisiología , Oligosacáridos/administración & dosificación , Probióticos/administración & dosificación , Bacteroides/crecimiento & desarrollo , Clostridium/crecimiento & desarrollo , Recuento de Colonia Microbiana , Método Doble Ciego , Femenino , Citometría de Flujo , Humanos , Concentración de Iones de Hidrógeno , Hibridación in Situ , Lactante , Recién Nacido , Intestinos/microbiología , Masculino , Oligosacáridos/química
17.
J Mol Microbiol Biotechnol ; 14(1-3): 128-36, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17957120

RESUMEN

The survival of Bifidobacterium animalis strain DN-173 010 was assessed after its ingestion in a fermented product or in a lyophilised form. Twelve healthy subjects were included in a randomised, open study with 2 parallel groups. The composition and activities of the faecal microbiota were monitored before (10-day baseline step), during (1-week product administration step) and after (10-day follow-up step) the ingestion of 1 of the 2 products. A colony immunoblotting method, fluorescent in situ hybridisation with group-specific DNA probes, and temporal temperature gradient gel electrophoresis using group-specific primers were carried out to compare survival of B. animalis strain DN-173 010 after ingestion of the 2 products, together with analyses of enzyme activities and faecal metabolites. At the end of the supplementation step, the mean number of B. animalis DN-173 010 quantified by immunodetection in the faeces of 5 of 6 subjects in each treatment group was >/=10(8) colony-forming units/g faeces. These numbers corresponded to an average survival of 22% for the lyophilised form and 20% for the fermented product. At the same step, the PCR temporal temperature gradient gel electrophoresis profiles showed a double band corresponding to the B. animalis DN-173 010 pattern for 11 subjects. No major modification was observed during the trial in either the dominant members of the faecal microbiota assessed by fluorescent in situ hybridisation or their activities. In conclusion, we show that the lyophilised form of B. animalis DN-173 010 survives transit and could represent a more convenient form to administer for long-term clinical trials.


Asunto(s)
Bifidobacterium/crecimiento & desarrollo , Productos Lácteos Cultivados/microbiología , Heces/microbiología , Liofilización/métodos , Probióticos/administración & dosificación , Adulto , Bifidobacterium/aislamiento & purificación , Recuento de Colonia Microbiana , Femenino , Citometría de Flujo , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
18.
J Mol Microbiol Biotechnol ; 14(1-3): 137-44, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17957121

RESUMEN

Lactococcus lactis is a model of food-grade lactic acid bacterium, which can durably colonize the digestive tract of germ-free mice. To study in vivo the bacterial adaptation to a novel nutritional resource brought by alimentation, the lactose-catabolizing strain IL2661 of L. lactis was established in monoxeny in mice. Half of the mice then received a lactose-rich diet. The mouse has no efficient intestinal lactase and is well adapted to a follow-up of the metabolic activity of microbial origin. The analysis of lactose and lactate in the feces suggested that L. lactis was able to use lactose in vivo. We developed a proteomic approach to evaluate in deeper details the metabolic response of the bacterium. We observed that L. lactis switched its metabolism to use the novel carbon source and reduced the level of proteins involved in an alternative mode of ATP production. In parallel, we also found that the amount of proteins involved in transcriptional regulation, transport and catabolism decreased in the presence of lactose. The proteome analysis informed us about the resources used by the bacteria in absence of lactose. In competition experiments, we found that the metabolic adaptation gives a strong ecological advantage to the bacteria able to efficiently utilize lactose.


Asunto(s)
Adaptación Fisiológica , Proteínas Bacterianas/metabolismo , Tracto Gastrointestinal/microbiología , Regulación Bacteriana de la Expresión Génica , Lactococcus lactis/metabolismo , Lactosa/metabolismo , Proteoma , Animales , Proteínas Bacterianas/genética , Vida Libre de Gérmenes , Lactococcus lactis/crecimiento & desarrollo , Ratones , Ratones Endogámicos C3H
19.
Carcinogenesis ; 28(11): 2419-25, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17660508

RESUMEN

2-amino-3-methylimidazo[4,5-f]quinoline (IQ) is a genotoxic/carcinogenic compound formed in meat and fish during cooking. Following absorption in the upper part of the gastrointestinal tract, IQ is mainly metabolized in the liver by xenobiotic-metabolizing enzymes. Among them, UDP-glucuronosyl transferases lead to harmless glucuronidated derivatives that are partly excreted via the bile into the digestive lumen, where they come into contact with the resident microbiota. The purpose of this study is to investigate if microbial beta-glucuronidase could contribute to IQ genotoxicity by releasing reactive intermediates from IQ glucuronides. We constructed a beta-glucuronidase-deficient isogenic mutant from a wild-type Escherichia coli strain carrying the gene uidA encoding this enzyme and compared the genotoxicity of IQ in gnotobiotic rats monoassociated with the wild-type or the mutant strain. The Comet assay performed on colonocytes and hepatocytes showed that the presence of beta-glucuronidase in the digestive lumen dramatically increased (3-fold) the genotoxicity of IQ in the colon. This deleterious effect was paralleled by slight modifications of the pharmacokinetics of IQ. The urinary and faecal excretion of the parent compound and its conjugated derivatives reached a maximum 24-48 h after gavage in rats harbouring the beta-glucuronidase-deficient strain. In rats associated with the wild-type strain, the kinetics of urinary excretion showed a biphasic curve with a second, smaller peak after 144 h. This is the first in vivo demonstration that bacterial beta-glucuronidase plays a pivotal role in the genotoxicity of a common food-borne carcinogen.


Asunto(s)
Bacterias/enzimología , Colon/efectos de los fármacos , Glucuronidasa/metabolismo , Intestinos/enzimología , Mutágenos/toxicidad , Quinolinas/toxicidad , Animales , Cromatografía Líquida de Alta Presión , Ensayo Cometa , Heces , Humanos , Masculino , Espectrometría de Masas , Quinolinas/orina , Ratas , Ratas Endogámicas F344
20.
FEMS Microbiol Ecol ; 60(3): 513-20, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17428302

RESUMEN

Molecular methods based on 16S rRNA gene sequence analyses have shown that bacteria of the Clostridium leptum subgroup are predominant in the colonic microbiota of healthy humans; this subgroup includes bacteria that produce butyrate, a source of energy for intestinal epithelial cells. To improve our understanding of the species within this important group, separation methods using fluorescence-activated cell sorting (FACS) and specific PCR were combined with 16S rRNA gene sequence analyses. FACS was developed for bacteria labelled in situ with two rRNA oligonucleotide probes, namely EUB338-FITC for total bacteria and Clep866-CY5/cp or Fprau645-CY5 for bacteria of the C. leptum subgroup. Bacterial cell sorting allowed a selective recovery of members of the C. leptum subgroup from the human microbiota with efficiencies as high as 95%. Group-specific PCR amplification of the C. leptum subgroup was developed, and temporal thermal gradient gel electrophoresis showed host-specific profiles with low complexity, with a sharing of common bands between individuals and bands stable over 2 months for the same individual. A library of 16S rRNA gene cloned sequences (106 sequences) was prepared with DNA obtained from both separation methods, and 15 distinct phylotypes were identified, among which 10 have no cultivable or currently cultivated representative in reference collections.


Asunto(s)
Clostridium/aislamiento & purificación , Colon/microbiología , Citometría de Flujo , Reacción en Cadena de la Polimerasa , Clostridium/clasificación , Clostridium/genética , ADN Bacteriano/genética , Heces/microbiología , Genes de ARNr , Humanos , Hibridación Fluorescente in Situ , Datos de Secuencia Molecular , Oligonucleótidos , Filogenia , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA