Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Vet Res ; 49(1): 35, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29636083

RESUMEN

The minimum inhibitory concentration of bambermycin on three porcine Helicobacter suis strains was shown to be 8 µg/mL. The effect of in-feed medication with this antibiotic on the course of a gastric infection with one of these strains, the host response and the gastric microbiota was determined in mice, as all of these parameters may be involved in gastric pathology. In H. suis infected mice which were not treated with bambermycin, an increased number of infiltrating B-cells, T-cells and macrophages in combination with a Th2 response was demonstrated, as well as a decreased parietal cell mass. Compared to this non-treated, infected group, in H. suis infected mice medicated with bambermycin, gastric H. suis colonization was not altered, but a decreased number of infiltrating T-cells, B-cells and macrophages as well as downregulated expressions of IL-1ß, IL-8M, IL-10 and IFN-γ were demonstrated and the parietal cell mass was not affected. In bambermycin treated mice that were not infected with H. suis, the number of infiltrating T-cells and expression of IL-1ß were lower than in non-infected mice that did not receive bambermycin. Gastric microbiota analysis indicated that the relative abundance of bacteria that might exert unfavorable effects on the host was decreased during bambermycin supplementation. In conclusion, bambermycin did not affect H. suis colonization, but decreased gastric inflammation and inhibited the effects of a H. suis infection on parietal cell loss. Not only direct interaction of H. suis with parietal cells, but also inflammation may play a role in death of these gastric acid producing cells.


Asunto(s)
Antibacterianos/farmacología , Bambermicinas/farmacología , Infecciones por Helicobacter/veterinaria , Helicobacter heilmannii/fisiología , Enfermedades de los Porcinos/tratamiento farmacológico , Alimentación Animal/análisis , Animales , Dieta/veterinaria , Suplementos Dietéticos/análisis , Modelos Animales de Enfermedad , Femenino , Infecciones por Helicobacter/tratamiento farmacológico , Inflamación/inmunología , Inflamación/microbiología , Inflamación/veterinaria , Ratones , Ratones Endogámicos BALB C , Células Parietales Gástricas/inmunología , Organismos Libres de Patógenos Específicos , Estómago/inmunología , Porcinos
2.
Sci Rep ; 6: 20169, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26833404

RESUMEN

Helicobacter (H.) suis causes gastric pathologies in both pigs and humans. Very little is known on the metabolism of this bacterium and its impact on the host. In this study, we have revealed the importance of the glutamate-generating metabolism, as shown by a complete depletion of glutamine (Gln) in the medium during H. suis culture. Besides Gln, H. suis can also convert glutathione (GSH) to glutamate, and both reactions are catalyzed by the H. suis γ-glutamyltranspeptidase (GGT). Both for H. pylori and H. suis, it has been hypothesized that the degradation of Gln and GSH may lead to a deficiency for the host, possibly initiating or promoting several pathologies. Therefore the in vivo effect of oral supplementation with Gln and GSH was assessed. Oral supplementation with Gln was shown to temper H. suis induced gastritis and epithelial (hyper)proliferation in Mongolian gerbils. Astonishingly, supplementation of the feed with GSH, another GGT substrate, resulted in inflammation and epithelial proliferation levels returning to baseline levels of uninfected controls. This indicates that Gln and GSH supplementation may help reducing tissue damage caused by Helicobacter infection in both humans and pigs, highlighting their potential as a supportive therapy during and after Helicobacter eradication therapy.


Asunto(s)
Suplementos Dietéticos , Glutatión/administración & dosificación , Glutatión/uso terapéutico , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/microbiología , Helicobacter/fisiología , Estómago/microbiología , Estómago/patología , Administración Oral , Aminoácidos/análisis , Amoníaco/metabolismo , Animales , Carbohidratos/análisis , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Gerbillinae , Glutamina/metabolismo , Glutatión/farmacología , Helicobacter/efectos de los fármacos , Helicobacter/crecimiento & desarrollo , Inflamación/patología , Antígeno Ki-67/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/patología , Viabilidad Microbiana/efectos de los fármacos , gamma-Glutamiltransferasa/metabolismo
3.
PLoS One ; 8(10): e77966, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24147103

RESUMEN

Helicobacter (H.) suis colonizes the stomach of the majority of pigs as well as a minority of humans worldwide. Infection causes chronic inflammation in the stomach of the host, however without an effective clearance of the bacteria. Currently, no information is available about possible mechanisms H. suis utilizes to interfere with the host immune response. This study describes the effect on various lymphocytes of the γ-glutamyl transpeptidase (GGT) from H. suis. Compared to whole cell lysate from wild-type H. suis, lysate from a H. suis ggt mutant strain showed a decrease of the capacity to inhibit Jurkat T cell proliferation. Incubation of Jurkat T cells with recombinantly expressed H. suis GGT resulted in an impaired proliferation, and cell death was shown to be involved. A similar but more pronounced inhibitory effect was also seen on primary murine CD4(+) T cells, CD8(+) T cells, and CD19(+) B cells. Supplementation with known GGT substrates was able to modulate the observed effects. Glutamine restored normal proliferation of the cells, whereas supplementation with reduced glutathione strengthened the H. suis GGT-mediated inhibition of proliferation. H. suis GGT treatment abolished secretion of IL-4 and IL-17 by CD4(+) T cells, without affecting secretion of IFN-γ. Finally, H. suis outer membrane vesicles (OMV) were identified as a possible delivery route of H. suis GGT to lymphocytes residing in the deeper mucosal layers. Thus far, this study is the first to report that the effects on lymphocytes of this enzyme, not only important for H. suis metabolism but also for that of other Helicobacter species, depend on the degradation of two specific substrates: glutamine and reduced glutatione. This will provide new insights into the pathogenic mechanisms of H. suis infection in particular and infection with gastric helicobacters in general.


Asunto(s)
Membrana Celular/metabolismo , Glutamina/farmacología , Glutatión/farmacología , Helicobacter heilmannii/enzimología , Linfocitos/metabolismo , gamma-Glutamiltransferasa/metabolismo , Animales , Antígenos CD19/metabolismo , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Interleucina-17/metabolismo , Interleucina-4/metabolismo , Ratones , Transporte de Proteínas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA