Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Vaccine ; 42(22): 126204, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39126830

RESUMEN

The ESKAPE family, comprising Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp., poses a significant global threat due to their heightened virulence and extensive antibiotic resistance. These pathogens contribute largely to the prevalence of nosocomial or hospital-acquired infections, resulting in high morbidity and mortality rates. To tackle this healthcare problem urgent measures are needed, including development of innovative vaccines and therapeutic strategies. Designing vaccines involves a complex and resource-intensive process of identifying protective antigens and potential vaccine candidates (PVCs) from pathogens. Reverse vaccinology (RV), an approach based on genomics, made this process more efficient by leveraging bioinformatics tools to identify potential vaccine candidates. In recent years, artificial intelligence and machine learning (ML) techniques has shown promise in enhancing the accuracy and efficiency of reverse vaccinology. This study introduces a supervised ML classification framework, to predict potential vaccine candidates specifically against ESKAPE pathogens. The model's training utilized biological and physicochemical properties from a dataset containing protective antigens and non-protective proteins of ESKAPE pathogens. Conventional autoencoders based strategy was employed for feature encoding and selection. During the training process, seven machine learning algorithms were trained and subjected to Stratified 5-fold Cross Validation. Random Forest and Logistic Regression exhibited best performance in various metrics including accuracy, precision, recall, WF1 score, and Area under the curve. An ensemble model was developed, to take collective strengths of both the algorithms. To assess efficacy of our final ensemble model, a high-quality benchmark dataset was employed. VacSol-ML(ESKAPE) demonstrated outstanding discrimination between protective vaccine candidates (PVCs) and non-protective antigens. VacSol-ML(ESKAPE), proves to be an invaluable tool in expediting vaccine development for these pathogens. Accessible to the public through both a web server and standalone version, it encourages collaborative research. The web-based and standalone tools are available at http://vacsolml.mgbio.tech/.


Asunto(s)
Antígenos Bacterianos , Vacunas Bacterianas , Aprendizaje Automático , Antígenos Bacterianos/inmunología , Humanos , Vacunas Bacterianas/inmunología , Klebsiella pneumoniae/inmunología , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/patogenicidad , Enterococcus faecium/inmunología , Enterococcus faecium/genética , Staphylococcus aureus/inmunología , Staphylococcus aureus/genética , Acinetobacter baumannii/inmunología , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Biología Computacional/métodos , Enterobacter/inmunología , Enterobacter/genética , Vacunología/métodos
2.
BMC Microbiol ; 24(1): 280, 2024 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-39068414

RESUMEN

BACKGROUND: Enterococcus faecium and Staphylococcus aureus are the Gram-positive pathogens of the ESKAPE group, known to represent a great threat to human health due to their high virulence and multiple resistances to antibiotics. Combined, enterococci and S. aureus account for 26% of healthcare-associated infections and are the most common organisms responsible for blood stream infections. We previously showed that the peptidyl-prolyl cis/trans isomerase (PPIase) PpiC of E. faecium elicits the production of specific, opsonic, and protective antibodies that are effective against several strains of E. faecium and E. faecalis. Due to the ubiquitous characteristics of PPIases and their essential function within Gram-positive cells, we hypothesized a potential cross-reactive effect of anti-PpiC antibodies. RESULTS: Opsonophagocytic assays combined with bioinformatics led to the identification of the foldase protein PrsA as a new potential vaccine antigen in S. aureus. We show that PrsA is a stable dimeric protein able to elicit opsonic antibodies against the S. aureus strain MW2, as well as cross-binding and cross-opsonic in several S. aureus, E. faecium and E. faecalis strains. CONCLUSIONS: Given the multiple antibiotic resistances S. aureus and enterococci present, finding preventive strategies is essential to fight those two nosocomial pathogens. The study shows the potential of PrsA as an antigen to use in vaccine formulation against the two dangerous Gram-positive ESKAPE bacteria. Our findings support the idea that PPIases should be further investigated as vaccine targets in the frame of pan-vaccinomics strategy.


Asunto(s)
Proteínas Bacterianas , Enterococcus faecalis , Enterococcus faecium , Isomerasa de Peptidilprolil , Staphylococcus aureus , Staphylococcus aureus/inmunología , Staphylococcus aureus/genética , Enterococcus faecium/inmunología , Enterococcus faecium/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética , Isomerasa de Peptidilprolil/inmunología , Isomerasa de Peptidilprolil/genética , Enterococcus faecalis/inmunología , Enterococcus faecalis/genética , Humanos , Infecciones por Bacterias Grampositivas/prevención & control , Infecciones por Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/microbiología , Vacunas Bacterianas/inmunología , Proteínas Opsoninas/inmunología , Anticuerpos Antibacterianos/inmunología , Anticuerpos Antibacterianos/sangre , Animales , Reacciones Cruzadas , Ratones , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/genética , Fagocitosis , Infecciones Estafilocócicas/prevención & control , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología
3.
Microbiol Spectr ; 12(6): e0190923, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38651859

RESUMEN

Acquired immunity is an important way to construct the intestinal immune barrier in mammals, which is almost dependent on suckling. To develop a new strategy for accelerating the construction of gut microbiome, newborn Holstein calves were continuously fed with 40 mL of compound probiotics (containing Lactobacillus plantarum T-14, Enterococcus faecium T-11, Saccharomyces cerevisiae T-209, and Bacillus licheniformis T-231) per day for 60 days. Through diarrhea rate monitoring, immune index testing, antioxidant capacity detection, and metagenome sequencing, the changes in diarrhea incidence, average daily gain, immune index, and gut microbiome of newborn calves within 60 days were investigated. Results indicated that feeding the compound probiotics reduced the average diarrhea rate of calves by 42.90%, increased the average daily gain by 43.40%, raised the antioxidant indexes of catalase, superoxide dismutase, total antioxidant capacity, and Glutathione peroxidase by 22.81%, 6.49%, 8.33%, and 13.67%, respectively, and increased the immune indexes of IgA, IgG, and IgM by 10.44%, 4.85%, and 6.12%, respectively. Moreover, metagenome sequencing data showed that feeding the compound probiotics increased the abundance of beneficial strains (e.g., Lactococcus lactis and Bacillus massionigeriensis) and decreased the abundance of some harmful strains (e.g., Escherichia sp. MOD1-EC5189 and Mycobacterium brisbane) in the gut microbiome of calves, thus contributing to accelerating the construction of healthy gut microbiome in newborn Holstein calves. IMPORTANCE: The unstable gut microbiome and incomplete intestinal function of newborn calves are important factors for the high incidence of early diarrhea. This study presents an effective strategy to improve the overall immunity and gut microbiome in calves and provides new insights into the application of compound probiotics in mammals.


Asunto(s)
Diarrea , Microbioma Gastrointestinal , Probióticos , Animales , Bovinos , Probióticos/administración & dosificación , Probióticos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Diarrea/veterinaria , Diarrea/microbiología , Diarrea/inmunología , Diarrea/prevención & control , Animales Recién Nacidos , Enfermedades de los Bovinos/microbiología , Enfermedades de los Bovinos/prevención & control , Enfermedades de los Bovinos/inmunología , Saccharomyces cerevisiae , Lactobacillus plantarum , Enterococcus faecium/inmunología
4.
Front Immunol ; 12: 793260, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069576

RESUMEN

Dietary probiotics may enhance gut health by directly competing with pathogenic agents and through immunostimulatory effects. These properties are recognized in the context of bacterial and viral pathogens, but less is known about interactions with eukaryotic pathogens such as parasitic worms (helminths). In this study we investigated whether two probiotic mixtures (comprised of Bacillus amyloliquefaciens, B. subtilis, and Enterococcus faecium [BBE], or Lactobacillus rhamnosus LGG and Bifidobacterium animalis subspecies Lactis Bb12 [LB]) could modulate helminth infection kinetics as well as the gut microbiome and intestinal immune responses in pigs infected with the nodular worm Oesophagostomum dentatum. We observed that neither probiotic mixture influenced helminth infection levels. BBE, and to a lesser extent LB, changed the alpha- and beta-diversity indices of the colon and fecal microbiota, notably including an enrichment of fecal Bifidobacterium spp. by BBE. However, these effects were muted by concurrent O. dentatum infection. BBE (but not LB) significantly attenuated the O. dentatum-induced upregulation of genes involved in type-2 inflammation and restored normal lymphocyte ratios in the ileo-caecal lymph nodes that were altered by infection. Moreover, inflammatory cytokine release from blood mononuclear cells and intestinal lymphocytes was diminished by BBE. Collectively, our data suggest that selected probiotic mixtures can play a role in maintaining immune homeostasis during type 2-biased inflammation. In addition, potentially beneficial changes in the microbiome induced by dietary probiotics may be counteracted by helminths, highlighting the complex inter-relationships that potentially exist between probiotic bacteria and intestinal parasites.


Asunto(s)
Bacillus/inmunología , Enterococcus faecium/inmunología , Microbioma Gastrointestinal/inmunología , Esofagostomiasis , Oesophagostomum/inmunología , Probióticos/farmacología , Enfermedades de los Porcinos , Animales , Femenino , Masculino , Esofagostomiasis/inmunología , Esofagostomiasis/microbiología , Esofagostomiasis/veterinaria , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/parasitología
5.
Invest Ophthalmol Vis Sci ; 61(8): 22, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32673387

RESUMEN

Purpose: Ocular surface microbiome changes can affect meibomian gland dysfunction (MGD) development. This study aimed to delineate differences among the microbiome of eyelid skin, conjunctiva, and meibum in healthy controls (HCs) and patients afflicted with MGD. Methods: Shotgun metagenomic analysis was used to determine if there are differences between the microbial communities in ocular sites surrounding the meibomian gland in healthy individuals and patients afflicted with MGD. Results: The meibum bacterial content of these microbiomes was dissimilar in these two different types of individuals. Almost all of the most significant taxonomic changes in the meibum microbiome of individuals with MGD were also present in their eyelid skin, but not in the conjunctiva. Such site-specific microbe pattern changes accompany increases in the gene expression levels controlling carbohydrate and lipid metabolism. Most of the microbiomes in patients with MGD possess a microbe population capable of metabolizing benzoate. Pathogens known to underlie ocular infection were evident in these individuals. MGD meibum contained an abundance of Campylobacter coli, Campylobacter jejuni, and Enterococcus faecium pathogens, which were almost absent from HCs. Functional annotation indicated that in the microbiomes of MGD meibum their capability to undergo chemotaxis, display immune evasive virulence, and mediate type IV secretion was different than that in the microbiomes of meibum isolated from HCs. Conclusions: MGD meibum contains distinct microbiota whose immune evasive virulence is much stronger than that in the HCs. Profiling differences between the meibum microbiome makeup in HCs and patients with MGD characterizes changes of microbial communities associated with the disease status.


Asunto(s)
Campylobacter coli , Campylobacter jejuni , Enterococcus faecium , Párpados/microbiología , Disfunción de la Glándula de Meibomio , Metagenómica/métodos , Microbiota/genética , Lágrimas , Adulto , Campylobacter coli/genética , Campylobacter coli/inmunología , Campylobacter coli/patogenicidad , Campylobacter jejuni/genética , Campylobacter jejuni/inmunología , Campylobacter jejuni/patogenicidad , Conjuntiva/microbiología , Enterococcus faecium/genética , Enterococcus faecium/inmunología , Enterococcus faecium/patogenicidad , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Evasión Inmune , Masculino , Disfunción de la Glándula de Meibomio/metabolismo , Disfunción de la Glándula de Meibomio/microbiología , Lágrimas/metabolismo , Lágrimas/microbiología
6.
BMC Res Notes ; 13(1): 159, 2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32178719

RESUMEN

OBJECTIVE: Probiotics are fed to improve enteric health, and they may also affect respiratory immunity through their exposure to the upper respiratory tract upon ingestion. However, their effect on the respiratory system is not known. Our aim was to determine how probiotics affect functions and markers of bronchoalveolar lung lavage cells (BAL) isolated from lungs of calves at slaughter. RESULTS: Treatments consisted of ten probiotic species and one control treatment. Probiotics and BAL were incubated 1:1 for 2 h at 37 °C and 5% CO2. The cell surface markers measured included CD14, CD205, and CD18, and E. coli bioparticles were used to measure phagocytosis and oxidative burst. Differences were considered significant at P ≤ 0.05 and were noted for percent cells fluorescing and mean fluorescence intensity for CD14 and CD205. Additionally, oxidative burst was different as measured by both percentage of cells fluorescing and mean fluorescence intensity, and phagocytosis differed among species as measured by mean fluorescence intensity. Overall, probiotic species differed in their ability to suppress or increase leukocyte function showing that probiotic bacteria differentially modulate BAL.


Asunto(s)
Células Epiteliales Alveolares/microbiología , Pulmón/microbiología , Probióticos/administración & dosificación , Células Epiteliales Alveolares/inmunología , Animales , Antígenos CD/inmunología , Antígenos CD18/inmunología , Bovinos , Enterococcus faecium/inmunología , Fluorescencia , Lectinas Tipo C/inmunología , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Receptores de Lipopolisacáridos/inmunología , Pulmón/metabolismo , Antígenos de Histocompatibilidad Menor/inmunología , Fagocitosis/efectos de los fármacos , Propionibacterium freudenreichii/inmunología , Receptores de Superficie Celular/inmunología
7.
Inflammation ; 43(2): 552-567, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31811548

RESUMEN

Intestinal epithelial cells (IEC) and immune cells, such as dendritic cells (DC), jointly control the immune response towards luminal pathogens in the intestinal mucosa. Crosstalk between IEC and DC is crucial for coordinating immune responses and occurs via soluble factors and direct cell-cell contacts. The present study aimed at establishing a direct-contact co-culture model of porcine IEC and DC to mimic these interactions. The effects of (1) co-cultivation of the two cell types and (2) bacterial infection on the inflammatory response patterns of each of the cell types were determined with a special focus on the canonical and non-canonical inflammasome signaling pathways. In infection experiments, in vitro cultures were exposed to either the probiotic Enterococcus (E.) faecium NCIMB 10415 or enterotoxigenic Escherichia coli (ETEC). In porcine IEC (IPEC-J2), co-cultivation with porcine monocyte-derived DC (MoDC) resulted in reduced basal NLRP3 (nucleotide oligomerization domain [NOD]-like receptor [NLR] family, pyrin domain containing 3) inflammasome mRNA levels in unstimulated conditions. In porcine MoDC, the presence of IPEC-J2 cells evoked a noticeable decrease of interleukin (IL)-8 and transforming growth factor-ß (TGF-ß) mRNA and protein expression. ETEC, in contrast to E. faecium, modulated the inflammasome pathway in IPEC-J2 cells and porcine MoDC. Co-cultured IPEC-J2 cells showed an augmented inflammasome response to ETEC infection. By contrast, MoDC revealed a weakened ETEC response under such co-culture conditions as indicated by a reduction of inflammasome-related IL-1ß protein release. Our data indicate that the close contact between IEC and resident immune cells has a major effect on their immunological behavior.


Asunto(s)
Células Dendríticas/microbiología , Enterococcus faecium , Mediadores de Inflamación , Mucosa Intestinal/microbiología , Monocitos/microbiología , Animales , Línea Celular , Técnicas de Cocultivo/métodos , Células Dendríticas/inmunología , Enterococcus faecium/inmunología , Mediadores de Inflamación/inmunología , Mucosa Intestinal/inmunología , Monocitos/inmunología , Porcinos
8.
Infect Immun ; 87(9)2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31285252

RESUMEN

Multidrug-resistant enterococci are major causes of hospital-acquired infections. Immunotherapy with monoclonal antibodies (MAbs) targeting bacterial antigens would be a valuable treatment option in this setting. Here, we describe the development of two MAbs through hybridoma technology that target antigens from the most clinically relevant enterococcal species. Diheteroglycan (DHG), a well-characterized capsular polysaccharide of Enterococcus faecalis, and the secreted antigen A (SagA), an immunogenic protein from Enterococcus faecium, are both immunogens that have been proven to raise opsonic and cross-reactive antibodies against enterococcal strains. For this purpose, a conjugated form of the native DHG with SagA was used to raise the antibodies in mice, while enzyme-linked immunosorbent assay and opsonophagocytic assay were combined in the selection process of hybridoma cells producing immunoreactive and opsonic antibodies targeting the selected antigens. From this process, two highly specific IgG1(κ) MAbs were obtained, one against the polysaccharide (DHG.01) and one against the protein (SagA.01). Both MAbs exhibited good opsonic killing against the target bacterial strains: DHG.01 showed 90% killing against E. faecalis type 2, and SagA.01 showed 40% killing against E. faecium 11231/6. In addition, both MAbs showed cross-reactivity toward other E. faecalis and E. faecium strains. The sequences from the variable regions of the heavy and light chains were reconstructed in expression vectors, and the activity of the MAbs upon expression in eukaryotic cells was confirmed with the same immunological assays. In summary, we identified two opsonic MAbs against enterococci which could be used for therapeutic or prophylactic approaches against enterococcal infections.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Farmacorresistencia Microbiana , Enterococcus faecalis/inmunología , Enterococcus faecium/inmunología , Inmunoterapia/métodos , Proteínas Opsoninas/inmunología , Animales , Antígenos Bacterianos/inmunología , Cápsulas Bacterianas/química , Ratones , Polisacáridos/inmunología
9.
J Infect Dis ; 220(10): 1589-1598, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31289829

RESUMEN

Enterococci have emerged as important nosocomial pathogens due to their resistance to the most commonly used antibiotics. Alternative treatments or prevention options are aimed at polysaccharides and surface-related proteins that play important roles in pathogenesis. Previously, we have shown that 2 Enterococcus faecium proteins, the secreted antigen A and the peptidyl-prolyl cis-trans isomerase, as well as the Enterococcus faecalis polysaccharide diheteroglycan, are able to induce opsonic and cross-protective antibodies. Here, we evaluate the use of glycoconjugates consisting of these proteins and an enterococcal polysaccharide to develop a vaccine with broader strain coverage. Diheteroglycan was conjugated to these 2 enterococcal proteins. Rabbit sera raised against these glycoconjugates showed Immunoglobulin G titers against the corresponding conjugate, as well as against the respective protein and carbohydrate antigens. Effective opsonophagocytic killing for the 2 sera was observed against different E. faecalis and E. faecium strains. Enzyme-linked immunosorbent assays against whole bacterial cells showed immune recognition of 22 enterococcal strains by the sera. Moreover, the sera conferred protection against E. faecalis and E. faecium strains in a mouse infection model. Our results suggest that these glycoconjugates are promising candidates for vaccine formulations with a broader coverage against these nosocomial pathogens and that the evaluated proteins are potential carrier proteins.


Asunto(s)
Antígenos Bacterianos/inmunología , Vacunas Bacterianas/inmunología , Enterococcus faecalis/inmunología , Enterococcus faecium/inmunología , Infecciones por Bacterias Grampositivas/prevención & control , Animales , Anticuerpos Antibacterianos/sangre , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Actividad Bactericida de la Sangre , Modelos Animales de Enfermedad , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/patología , Inmunidad Heteróloga , Inmunoglobulina G/sangre , Masculino , Ratones Endogámicos BALB C , Viabilidad Microbiana , Proteínas Opsoninas/sangre , Fagocitosis , Polisacáridos Bacterianos/inmunología , Conejos , Resultado del Tratamiento , Vacunas Conjugadas/administración & dosificación , Vacunas Conjugadas/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
10.
Cell Host Microbe ; 26(1): 114-122.e8, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31278040

RESUMEN

Quorum-sensing molecules (QSMs) are secreted by bacteria to signal population density. Upon reaching a critical concentration, QSMs induce transcriptional alterations in bacteria, which enable virulence factor expression and biofilm formation. It is unclear whether mammalian hosts can recognize QSMs to trigger responsive antibacterial immunity. We report that mouse mast-cell-specific G-protein-coupled receptor Mrgprb2 and its human homolog MRGPRX2 are receptors for Gram-positive QSMs, including competence-stimulating peptide (CSP)-1. CSP-1 activates Mrgprb2 and MRGPRX2, triggering mast cell degranulation, which inhibits bacterial growth and prevents biofilm formation. Such antibacterial functions are reduced in Mrgprb2-deficient mast cells, while wild-type mast cells fail to inhibit the growth of bacterial strains lacking CSP-1. Mrgprb2-knockout mice exhibit reduced bacterial clearance, while pharmacologically activating Mrgprb2 in vivo eliminates bacteria and improves disease score. These findings identify a host defense mechanism that uses QSMs as an "Achilles heel" and suggest MRGPRX2 as a potential therapeutic target for controlling bacterial infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Tejido Conectivo/inmunología , Inmunidad Innata , Mastocitos/inmunología , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Streptococcus pneumoniae/inmunología , Animales , Bacteriocinas/metabolismo , Enterococcus faecium/inmunología , Humanos , Ratones , Ratones Noqueados , Streptococcus pyogenes/inmunología
11.
Glycoconj J ; 36(5): 429-438, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31230165

RESUMEN

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.


Asunto(s)
Anticuerpos Antibacterianos/farmacología , Antígenos Bacterianos/aislamiento & purificación , Pared Celular/química , Enterococcus faecium/química , Lipopolisacáridos/aislamiento & purificación , Polisacáridos Bacterianos/aislamiento & purificación , Ácidos Teicoicos/aislamiento & purificación , Animales , Antibacterianos/farmacología , Anticuerpos Antibacterianos/biosíntesis , Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Cápsulas Bacterianas/química , Cápsulas Bacterianas/inmunología , Pared Celular/inmunología , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Endocarditis Bacteriana/tratamiento farmacológico , Endocarditis Bacteriana/microbiología , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/inmunología , Enterococcus faecium/patogenicidad , Femenino , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Lipopolisacáridos/química , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos BALB C , Proteínas Opsoninas/farmacología , Proteínas de Unión a las Penicilinas/química , Proteínas de Unión a las Penicilinas/inmunología , Proteínas de Unión a las Penicilinas/aislamiento & purificación , Proteínas de Unión a las Penicilinas/farmacología , Fagocitosis/efectos de los fármacos , Polisacáridos Bacterianos/química , Polisacáridos Bacterianos/inmunología , Cultivo Primario de Células , Ratas , Ratas Wistar , Ácidos Teicoicos/química , Ácidos Teicoicos/inmunología , Ácidos Teicoicos/farmacología , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología
12.
Elife ; 82019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30969170

RESUMEN

We discovered that Enterococcus faecium (E. faecium), a ubiquitous commensal bacterium, and its secreted peptidoglycan hydrolase (SagA) were sufficient to enhance intestinal barrier function and pathogen tolerance, but the precise biochemical mechanism was unknown. Here we show E. faecium has unique peptidoglycan composition and remodeling activity through SagA, which generates smaller muropeptides that more effectively activates nucleotide-binding oligomerization domain-containing protein 2 (NOD2) in mammalian cells. Our structural and biochemical studies show that SagA is a NlpC/p60-endopeptidase that preferentially hydrolyzes crosslinked Lys-type peptidoglycan fragments. SagA secretion and NlpC/p60-endopeptidase activity was required for enhancing probiotic bacteria activity against Clostridium difficile pathogenesis in vivo. Our results demonstrate that the peptidoglycan composition and hydrolase activity of specific microbiota species can activate host immune pathways and enhance tolerance to pathogens.


Asunto(s)
Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Enterococcus faecium/enzimología , Enterococcus faecium/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/química , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Cristalografía por Rayos X , Células HEK293 , Humanos , Proteína Adaptadora de Señalización NOD2/metabolismo , Peptidoglicano/metabolismo , Conformación Proteica
13.
Infect Immun ; 87(6)2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30936157

RESUMEN

Enterococcus faecalis is a human intestinal pathobiont with intrinsic and acquired resistance to many antibiotics, including vancomycin. Nature provides a diverse and virtually untapped repertoire of bacterial viruses, or bacteriophages (phages), that could be harnessed to combat multidrug-resistant enterococcal infections. Bacterial phage resistance represents a potential barrier to the implementation of phage therapy, emphasizing the importance of investigating the molecular mechanisms underlying the emergence of phage resistance. Using a cohort of 19 environmental lytic phages with tropism against E. faecalis, we found that these phages require the enterococcal polysaccharide antigen (Epa) for productive infection. Epa is a surface-exposed heteroglycan synthesized by enzymes encoded by both conserved and strain-specific genes. We discovered that exposure to phage selective pressure favors mutation in nonconserved epa genes both in culture and in a mouse model of intestinal colonization. Despite gaining phage resistance, epa mutant strains exhibited a loss of resistance to cell wall-targeting antibiotics. Finally, we show that an E. faecalisepa mutant strain is deficient in intestinal colonization, cannot expand its population upon antibiotic-driven intestinal dysbiosis, and fails to be efficiently transmitted to juvenile mice following birth. This study demonstrates that phage therapy could be used in combination with antibiotics to target enterococci within a dysbiotic microbiota. Enterococci that evade phage therapy by developing resistance may be less fit at colonizing the intestine and sensitized to vancomycin, preventing their overgrowth during antibiotic treatment.


Asunto(s)
Antibacterianos/farmacología , Bacteriófagos/fisiología , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/virología , Enterococcus faecium/virología , Infecciones por Bacterias Grampositivas/terapia , Intestinos/microbiología , Animales , Terapia Biológica , Enterococcus faecalis/inmunología , Enterococcus faecalis/fisiología , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/inmunología , Enterococcus faecium/fisiología , Femenino , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Vancomicina/farmacología
14.
Vet Immunol Immunopathol ; 203: 78-87, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30143242

RESUMEN

Dendritic cells (DC) are crucial for maintaining intestinal homeostasis and generating proper immune responses to bacteria occurring in the gut. Microbial stimuli can be recognized by intracellular receptors called inflammasomes, e.g., nucleotide oligomerization domain (NOD)-like receptor protein 3 (NLRP3). The aim of the present study was to unravel the inflammasome response of porcine monocyte-derived DC (MoDC). We investigated the capacity of probiotic Enterococcus faecium NCIMB 10415 (E. faecium) and enterotoxigenic Escherichia coli (ETEC) to elicit inflammasome activation. Since inflammasome activation normally requires a two-step process, MoDC were initially incubated with lipopolysaccharide (LPS) in order to prime cells. Primed and unprimed cells were then stimulated with the aforementioned bacterial strains. We also assessed whether preincubation with the probiotic prior to ETEC infection modified the immune response via the inflammasome pathway. Phenotypical analysis by flow cytometry showed that monocytes and MoDC expressed the surface markers CD14, CD16, and CD1 continuously, whereas swine leucocyte antigen (SLA) II was upregulated during differentiation. Following LPS priming, NLRP3, interleukin (IL)-1ß and IL-18 mRNA expression, and IL-1ß protein release increased. In unprimed cells, ETEC upregulated the expression of inflammasome components at later time points than in LPS-primed MoDC. Preincubation with the probiotic did not influence NLRP3 inflammasome activation in comparison with cells infected with ETEC alone. We conclude that ETEC, but not E. faecium, was able to stimulate inflammasome components in porcine MoDC. The present experimental conditions revealed no NLRP3 inflammasome-dependent protective effects of E. faecium during a pathogenic ETEC challenge.


Asunto(s)
Células Dendríticas/inmunología , Enterococcus faecium/inmunología , Escherichia coli Enterotoxigénica/inmunología , Inflamasomas/inmunología , Probióticos/farmacología , Animales , Células Dendríticas/microbiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/veterinaria , Infecciones por Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/veterinaria , Porcinos/inmunología , Porcinos/microbiología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología
15.
Benef Microbes ; 9(6): 937-949, 2018 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-30099893

RESUMEN

The biological effects of three probiotic strains Lactobacillus rhamnosus K32, Bifidobacterium longum GT15, Enterococcus faecium L3 and their mixture were studied using a model of dysbiosis induced in rats by antibiotics. It was found that after taking different probiotics intestinal microbiota changed in a strain-specific manner. The maximal activity against pathogens was revealed after the administration of a mixture of bacterial strains under study or a single strain of enterococci. The strain E. faecium L3 showed the most activity against both Klebsiella spp. and Bacteroides fragilis. It helped to restore the original content of Faecalibacterium prausnitzii. The number of Klebsiella spp. was the same in the group receiving L. rhamnosus K32 and the group of animals, which was not consuming probiotics. Different probiotic strains included in the composition had various immunological effects. Probiotic bifidobacteria, enterococci and the mixture of three probiotics stimulated of mRNA expression of interleukin (IL)-10 in mesenteric lymph nodes. The changes in microbiota after consuming an enterococcal probiotic correlated with an increase in transforming growth factor (TGF)-ß and IL-10 content in blood serum and an increase of the intestinal mucus layer. Consumption of L. rhamnosus K32 led to the stimulation of IL-8 expression in mesenteric lymph nodes. Control group not receiving probiotics was characterised by expression of pro-inflammatory cytokines, damage of epithelial cells and the destruction of their tight junctions. The damage to the ultrastructure of the mucosa was prevented in all the groups taking probiotics.


Asunto(s)
Bifidobacterium longum/inmunología , Disbiosis/terapia , Enterococcus faecium/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/inmunología , Lacticaseibacillus rhamnosus/inmunología , Probióticos/administración & dosificación , Animales , Bifidobacterium longum/crecimiento & desarrollo , Terapia Biológica/métodos , Modelos Animales de Enfermedad , Disbiosis/inducido químicamente , Enterococcus faecium/crecimiento & desarrollo , Inmunidad Innata , Factores Inmunológicos/sangre , Lacticaseibacillus rhamnosus/crecimiento & desarrollo , Ratas , Resultado del Tratamiento
16.
PLoS One ; 13(5): e0196564, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29727446

RESUMEN

Streptococcus agalactiae, or group B streptococcus (GBS), is an important pathogen as it is the leading cause of neonatal deaths due to sepsis, meningitis or bacterial pneumonia. Although the development of an effective and safe GBS vaccine is on the agenda of many research labs, there is no GBS vaccine on the market yet. In the present study we attempted to engineer a live vaccine strain based on Bac, a surface protein of GBS, incorporated into a surface fimbrial protein of probiotic Enterococcus. The resulting strain induced specific systemic and local immune responses in mice and provided protection against GBS when administered via the intranasal, oral or intravaginal immunization routes.


Asunto(s)
Inmunidad Mucosa , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Streptococcus agalactiae/patogenicidad , Administración Intranasal , Administración Intravaginal , Administración Oral , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/sangre , Carga Bacteriana , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Enterococcus faecium/genética , Enterococcus faecium/inmunología , Femenino , Proteínas Fimbrias/genética , Proteínas Fimbrias/inmunología , Ratones , Probióticos , Infecciones Estreptocócicas/microbiología , Vacunas Estreptocócicas/administración & dosificación , Vacunas Estreptocócicas/genética , Streptococcus agalactiae/genética , Vacunas Conjugadas/administración & dosificación , Vacunas Conjugadas/genética , Vacunas Conjugadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
17.
Avian Pathol ; 47(3): 325-333, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29534604

RESUMEN

Probiotics and immunization are being widely adopted by the poultry industry with the goal of controlling Salmonella enterica. However, the interaction between these two management protocols has been sparsely studied. The present study aimed to understand the role of an Enterococcus faecium probiotic in the production of salmonella-specific IgA in layers immunized with a live vaccine. Four groups were used: "Control" (no vaccine or probiotic); "Probiotic" (which received an E. faecium product); "Vaccine" (immunized with two doses of a live attenuated S. Enteritidis vaccine); and "Vaccine + probiotic". Faecal salmonella-specific IgA was analysed 7 and 20 days post-vaccination (dpv) boost. At 7 dpv, the "Vaccine" and "Vaccine + probiotic" groups had similar IgA levels. However, at 20 dpv, IgA levels were two times higher in the "Vaccine + probiotic" group compared to the "Vaccine" group. To understand the role of the intestinal microbiota in this finding, bacterial diversity in faeces was analysed by 16S rRNA gene sequencing. The improvement in IgA production in probiotic-treated birds was accompanied by marked changes in the faecal microbiome. Some of the main differences between the "Vaccine" and "Vaccine + probiotic" groups included reduction of Escherichia-Shigella and increases in Blautia, Anaerotruncus and Lactobacillus in the latter group. Although no direct causal link can be established from this study design, it is possible that the E. faecium probiotic induces improved antibody production following vaccination via modulation of the intestinal microbiota.


Asunto(s)
Pollos/inmunología , Enterococcus faecium/inmunología , Inmunoglobulina A/inmunología , Enfermedades de las Aves de Corral/prevención & control , Probióticos/farmacología , Salmonelosis Animal/prevención & control , Salmonella enteritidis/inmunología , Animales , Pollos/microbiología , Heces/microbiología , Femenino , Microbiota , Enfermedades de las Aves de Corral/microbiología , Salmonelosis Animal/microbiología , Vacunación/veterinaria , Vacunas Atenuadas/inmunología
18.
Virulence ; 9(1): 683-699, 2018 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-29436902

RESUMEN

The innate immune response of the nematode Caenorhabditis elegans has been extensively studied and a variety of Toll-independent immune response pathways have been identified. Surprisingly little, however, is known about how pathogens activate the C. elegans immune response. Enterococcus faecalis and Enterococcus faecium are closely related enterococcal species that exhibit significantly different levels of virulence in C. elegans infection models. Previous work has shown that activation of the C. elegans immune response by Pseudomonas aeruginosa involves P. aeruginosa-mediated host damage. Through ultrastructural imaging, we report that infection with either E. faecalis or E. faecium causes the worm intestine to become distended with proliferating bacteria in the absence of extensive morphological changes and apparent physical damage. Genetic analysis, whole-genome transcriptional profiling, and multiplexed gene expression analysis demonstrate that both enterococcal species, whether live or dead, induce a rapid and similar transcriptional defense response dependent upon previously described immune signaling pathways. The host response to E. faecium shows a stricter dependence upon stress response signaling pathways than the response to E. faecalis. Unexpectedly, we find that E. faecium is a C. elegans pathogen and that an active wild-type host defense response is required to keep an E. faecium infection at bay. These results provide new insights into the mechanisms underlying the C. elegans immune response to pathogen infection.


Asunto(s)
Caenorhabditis elegans/inmunología , Caenorhabditis elegans/fisiología , Enterococcus faecalis/inmunología , Enterococcus faecium/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Inmunidad Innata , Estrés Fisiológico , Animales , Caenorhabditis elegans/microbiología , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Enterococcus faecalis/crecimiento & desarrollo , Enterococcus faecium/crecimiento & desarrollo , Perfilación de la Expresión Génica , Infecciones por Bacterias Grampositivas/patología , Intestinos/microbiología , Intestinos/patología , Análisis por Micromatrices , Microscopía Electrónica de Transmisión , Análisis de Supervivencia
19.
Microbiologyopen ; 7(5): e00602, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29484836

RESUMEN

Hospitalized patients are often administered antibiotics that perturb the gastrointestinal commensal microbiota, leading to outgrowth of antibiotic-resistant bacteria, like multidrug-resistant Enterococcus faecium, subsequent spread, and eventually infections. However, the events that occur at the initial stage of intestinal colonization and outgrowth by multidrug-resistant E. faecium within the antibiotic-treated host have not been thoroughly studied. Here, we describe and visualize that only 6 hr after cephalosporin treatment of mice, the Muc-2 mucus layer is reduced and E-cadherin junctions were altered. In contrast, the cadherin-17 junctions were unaffected in antibiotic treated mice during E. faecium colonization or in untreated animals. E. faecium was capable to colonize the mouse colon already within 6 hr after inoculation, and agglutinated at the apical side of the intestinal epithelium. During the primary stage of gastrointestinal colonization the number of IgA+ cells and CD11b+ IgA+ cells increased in the lamina propria of the colon and mediated an elevated IgA response upon E. faecium colonization.


Asunto(s)
Antibacterianos/administración & dosificación , Cefalosporinas/administración & dosificación , Colon/microbiología , Enterococcus faecium/crecimiento & desarrollo , Inmunidad Mucosa , Mucosa Intestinal/microbiología , Inmunidad Adaptativa , Animales , Colon/inmunología , Enterococcus faecium/inmunología , Inmunidad Innata , Mucosa Intestinal/inmunología , Ratones
20.
Science ; 359(6371): 104-108, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29302014

RESUMEN

Anti-PD-1-based immunotherapy has had a major impact on cancer treatment but has only benefited a subset of patients. Among the variables that could contribute to interpatient heterogeneity is differential composition of the patients' microbiome, which has been shown to affect antitumor immunity and immunotherapy efficacy in preclinical mouse models. We analyzed baseline stool samples from metastatic melanoma patients before immunotherapy treatment, through an integration of 16S ribosomal RNA gene sequencing, metagenomic shotgun sequencing, and quantitative polymerase chain reaction for selected bacteria. A significant association was observed between commensal microbial composition and clinical response. Bacterial species more abundant in responders included Bifidobacterium longum, Collinsella aerofaciens, and Enterococcus faecium. Reconstitution of germ-free mice with fecal material from responding patients could lead to improved tumor control, augmented T cell responses, and greater efficacy of anti-PD-L1 therapy. Our results suggest that the commensal microbiome may have a mechanistic impact on antitumor immunity in human cancer patients.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Inmunoterapia/métodos , Melanoma/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Neoplasias Cutáneas/terapia , Animales , Anticuerpos Monoclonales/uso terapéutico , Bifidobacterium longum/clasificación , Bifidobacterium longum/genética , Bifidobacterium longum/inmunología , Bifidobacterium longum/aislamiento & purificación , Enterococcus faecium/clasificación , Enterococcus faecium/genética , Enterococcus faecium/inmunología , Enterococcus faecium/aislamiento & purificación , Heces/microbiología , Microbioma Gastrointestinal/genética , Humanos , Melanoma/inmunología , Ratones , ARN Ribosómico 16S/genética , Neoplasias Cutáneas/inmunología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA