Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Vaccine ; 41(34): 4927-4932, 2023 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-37423800

RESUMEN

Carriage of Neisseria meningitidisis an accepted endpoint in monitoring meningococcal vaccine effects. We applied molecular methods to assess the impact of menACWY vaccine implementation on meningococcal carriage and genogroup-specific prevalence in young adults in Fall of 2022, four years after the introduction of the tetravalent vaccine in the Netherlands. The overall carriage rate of genogroupable meningococci was not significantly different compared to a pre-menACWY cohort investigated in 2018 (20.8 % or 125 of 601 versus 17.4 % or 52 of 299 individuals, p = 0.25). Of 125 carriers of genogroupable meningococci, 122 (97.6 %) were positive for either vaccine-types menC, menW, menY or genogroups, menB, menE, and menX, which are not targeted by the menACWY vaccine. Compared with a pre-vaccine-implementation cohort, there was 3.8-fold reduction (p < 0.001) in vaccine-type carriage rates and 9.0-fold increase (p < 0.0001) in non-vaccine type menE prevalence. We observe a reduction in menW and menY and an increase in menE, which suggest that implementation of menACWY vaccine affected carriage.


Asunto(s)
Infecciones Meningocócicas , Vacunas Meningococicas , Neisseria meningitidis , Adulto Joven , Humanos , Neisseria meningitidis/genética , Países Bajos/epidemiología , Infecciones Meningocócicas/epidemiología , Infecciones Meningocócicas/prevención & control , Genotipo , Vacunas Combinadas
2.
BMC Microbiol ; 12: 299, 2012 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-23253484

RESUMEN

BACKGROUND: The use of food-grade Lactic Acid Bacteria (LAB) as DNA delivery vehicles represents an attractive strategy to deliver DNA vaccines at the mucosal surfaces as they are generally regarded as safe (GRAS). We previously showed that either native Lactococcus lactis (LL) or recombinant invasive LL expressing Fibronectin Binding Protein A of Staphylococcus aureus (LL-FnBPA+) or Internalin A of Listeria monocytogenes (LL-InlA+), were able to deliver and trigger DNA expression by epithelial cells, either in vitro or in vivo. InlA does not bind to its receptor, the murine E-cadherin, thus limiting the use of LL-InlA+ in in vivo murine models. Moreover, FnBPA binds to its receptors, integrins, via fibronectin introducing another limiting factor. In order to avoid the limitations of LL-InlA+ and LL-FnBPA+, a new L. lactis strain was engineered to produce a previously described mutated form of InlA (LL-mInlA+) allowing the binding of mInlA on murine E-cadherin. RESULTS: After showing the expression of mInLA at the surface of LL-mInlA+ strain, in vitro gentamycin survival assay in Caco-2 cells showed that LL-mInlA+ is 1000 times more invasive than LL. LL-mInlA+ invasivity was also validated by fluorescence microscopy. LL and LL-mInlA+ were transformed with pValacBLG, a plasmid containing the cDNA of bovine ß-Lactoglobulin (BLG), resulting in strains LL-BLG and LL-mInlA+BLG. The plasmid transfer in vitro using LL-mInlA+BLG was increased 10 times compared to LL-BLG. Moreover, the number of mice producing BLG in isolated enterocytes after oral administration of LL-mInlA+BLG in vivo was slightly higher than after oral administration of LL-BLG. CONCLUSIONS: We confirmed in this study that the production of mInlA at the surface of L. lactis is a promising strategy for plasmid transfer in vitro and in vivo.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Endocitosis , Técnicas de Transferencia de Gen , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Vacunas de ADN/metabolismo , Administración Oral , Animales , Proteínas Bacterianas/genética , Células CACO-2 , Enterocitos/metabolismo , Femenino , Expresión Génica , Genes Reporteros , Humanos , Lactoglobulinas/metabolismo , Listeria monocytogenes/genética , Ratones , Ratones Endogámicos BALB C , Plásmidos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Vacunas de ADN/administración & dosificación
3.
Am J Physiol Gastrointest Liver Physiol ; 298(6): G851-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20224007

RESUMEN

Lactobacillus plantarum, a commensal bacterium of humans, has been proposed to enhance the intestinal barrier, which is compromised in a number of intestinal disorders. To study the effect of L. plantarum strain WCFS1 on human barrier function, healthy subjects were administered L. plantarum or placebo in the duodenum for 6 h by means of a feeding catheter. The scaffold protein zonula occludens (ZO)-1 and transmembrane protein occludin were found to be significantly increased in the vicinity of the tight-junction (TJ) structures, which form the paracellular seal between cells of the epithelium. In an in vitro model of the human epithelium, L. plantarum induced translocation of ZO-1 to the TJ region; however, the effects on occludin were minor compared with those seen in vivo. L. plantarum was shown to activate Toll-like receptor 2 (TLR2) signaling, and treatment of Caco-2 monolayers with the TLR2 agonist Pam(3)-Cys-SK4(PCSK) significantly increased fluorescent staining of occludin in the TJ. Pretreatment of Caco-2 monolayers with L. plantarum or PCSK significantly attenuated the effects of phorbol ester-induced dislocation of ZO-1 and occludin and the associated increase in epithelial permeability. Our results identifying commensal bacterial stimulation of TLR2 in the gut epithelium as a regulator of epithelial integrity have important implications for understanding probiotic mechanisms and the control of intestinal homeostasis.


Asunto(s)
Células Epiteliales/metabolismo , Lactobacillus plantarum/fisiología , Proteínas de la Membrana/metabolismo , Uniones Estrechas/metabolismo , Adulto , Células CACO-2 , Estudios Cruzados , Citoprotección , Duodeno/citología , Duodeno/metabolismo , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/genética , Microscopía Confocal , Ocludina , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transducción de Señal , Uniones Estrechas/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Proteína de la Zonula Occludens-1
4.
Int J Med Microbiol ; 300(1): 41-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19783476

RESUMEN

In the intestine innate recognition of microbes is achieved through pattern recognition receptor (PRR) families expressed in immune cells and different cell lineages of the intestinal epithelium. Toll-like receptor (TLR) and nucleotide-binding and oligomerization domain-like receptor (NLR) families are emerging as key mediators of immunity through their role as maturation factors of immune cells and triggers for the production of cytokines and chemokines and antimicrobial factors. At the mucosal surface chronic activation of the immune system is avoided through the epithelial production of a glycocalyx, steady-state production of antimicrobial factors as well as the selective expression and localization of PRRs. Additionally, the polarization of epithelial TLR signaling and suppression of NF-kappaB activation by luminal commensals appears to contribute to the homeostasis of tolerance and immunity. Several studies have demonstrated that TLR signaling in epithelial cells contributes to a range of homeostatic mechanisms including proliferation, wound healing, epithelial integrity, and regulation of mucosal immune functions. The intestinal epithelium appears to have uniquely evolved to maintain mucosal tolerance and immunity, and future efforts to further understand the molecular mechanisms of intestinal homeostasis may have a major impact on human health.


Asunto(s)
Homeostasis , Tolerancia Inmunológica , Inmunidad , Intestinos/inmunología , Intestinos/microbiología , Transducción de Señal , Humanos
5.
Clin Transl Gastroenterol ; 7: e149, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26938480

RESUMEN

OBJECTIVES: The potential of Escherichia coli (E. coli) isolated from inflammatory bowel disease (IBD) patients to damage the integrity of the intestinal epithelium was investigated. METHODS: E. coli strains isolated from patients with ulcerative colitis (UC) and healthy controls were tested for virulence capacity by molecular techniques and cytotoxic assays and transepithelial electric resistance (TER). E. coli isolate p19A was selected, and deletion mutants were created for alpha-hemolysin (α-hemolysin) (hly) clusters and cytotoxic necrotizing factor type 1 (cnf1). Probiotic E. coli Nissle and pathogenic E. coli LF82 were used as controls. RESULTS: E. coli strains from patients with active UC completely disrupted epithelial cell tight junctions shortly after inoculation. These strains belong to phylogenetic group B2 and are all α-hemolysin positive. In contrast, probiotic E. coli Nissle, pathogenic E. coli LF82, four E. coli from patients with inactive UC and three E. coli strains from healthy controls did not disrupt tight junctions. E. coli p19A WT as well as cnf1, and single loci of hly mutants from cluster I and II were all able to damage Caco-2 (Heterogeneous human epithelial colorectal adenocarcinoma) cell tight junctions. However, this phenotype was lost in a mutant with knockout (Δ) of both hly loci (P<0.001). CONCLUSIONS: UC-associated E. coli producing α-hemolysin can cause rapid loss of tight junction integrity in differentiated Caco-2 cell monolayers. This effect was abolished in a mutant unable to express α-hemolysin. These results suggest that high Hly expression may be a mechanism by which specific strains of E. coli pathobionts can contribute to epithelial barrier dysfunction and pathophysiology of disease in IBD.

6.
BMC Res Notes ; 6: 431, 2013 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-24164922

RESUMEN

BACKGROUND: In the intestinal mucosa, several adaptations of TLR signalling have evolved to avoid chronic inflammatory responses to the presence of commensal microbes. Here we investigated whether polarized monolayers of intestinal epithelial cells might regulate inflammatory responses by secreting IL-8 in a vectorial fashion (i.e. apical versus basolateral) depending on the location of the TLR stimulus. RESULTS: In the Caco-2 BBE model of polarized villus-like epithelium, apical stimulation with TLR2 and TLR5 ligands resulted in the apical secretion of IL-8. The CXCR1 receptor for IL-8 was expressed only on the apical membrane of Caco-2 BBE cells and differentiated epithelial cells in the human small intestine and colon. Transcriptome analyses revealed that Caco-2 BBE cells respond to stimulation with IL-8 supporting the hypothesis that IL-8 induces G protein-coupled receptor signalling. CONCLUSIONS: These results show that IL-8 induces autocrine signalling via an apical CXCR1 in Caco-2 BBE intestinal epithelial cells and that this receptor is also expressed on the apical surface of differentiated human intestinal epithelial cells in vivo, suggesting an autocrine function for IL-8 secreted in the lumen.


Asunto(s)
Comunicación Autocrina/genética , Interleucina-8/metabolismo , Intestino Delgado/metabolismo , Receptores de Interleucina-8A/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 5/genética , Células CACO-2 , Polaridad Celular , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Interleucina-8/genética , Intestino Delgado/citología , Intestino Delgado/efectos de los fármacos , Lipopéptidos/farmacología , Mapeo de Interacción de Proteínas , Receptores de Interleucina-8A/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 5/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA