Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Infect Immun ; 89(12): e0022521, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34460289

RESUMEN

Heligmosomoides polygyrus is a helminth which naturally infects mice and is widely used as a laboratory model of chronic small intestinal helminth infection. While it is known that infection with H. polygyrus alters the composition of the host's bacterial microbiota, the functional implications of this alteration are unclear. We investigated the impact of H. polygyrus infection on short-chain fatty acid (SCFA) levels in the mouse intestine and sera. We found that helminth infection resulted in significantly upregulated levels of the branched SCFA isovaleric acid, exclusively in the proximal small intestine, which is the site of H. polygyrus colonization. We next set out to test the hypothesis that elevating local levels of isovaleric acid was a strategy used by H. polygyrus to promote its own fitness within the mammalian host. To test this, we supplemented the drinking water of mice with isovalerate during H. polygyrus infection and examined whether this affected helminth fecundity or chronicity. We did not find that isovaleric acid supplementation affected helminth chronicity; however, we found that it did promote helminth fecundity, as measured by helminth egg output in the feces of mice. Through antibiotic treatment of helminth-infected mice, we found that the bacterial microbiota was required in order to support elevated levels of isovaleric acid in the proximal small intestine during helminth infection. Overall, our data reveal that during H. polygyrus infection there is a microbiota-dependent localized increase in the production of isovaleric acid in the proximal small intestine and that this supports helminth fecundity in the murine host.


Asunto(s)
Ácidos Grasos Volátiles/metabolismo , Interacciones Huésped-Parásitos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitología , Nematospiroides dubius/fisiología , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/parasitología , Animales , Modelos Animales de Enfermedad , Metabolismo de los Lípidos , Ratones
2.
J Infect Dis ; 215(8): 1245-1254, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28368463

RESUMEN

Intestinal helminth infections occur predominantly in regions where exposure to enteric bacterial pathogens is also common. Helminth infections inhibit host immunity against microbial pathogens, which has largely been attributed to the induction of regulatory or type 2 (Th2) immune responses. Here we demonstrate an additional 3-way interaction in which helminth infection alters the metabolic environment of the host intestine to enhance bacterial pathogenicity. We show that an ongoing helminth infection increased colonization by Salmonella independently of T regulatory or Th2 cells. Instead, helminth infection altered the metabolic profile of the intestine, which directly enhanced bacterial expression of Salmonella pathogenicity island 1 (SPI-1) genes and increased intracellular invasion. These data reveal a novel mechanism by which a helminth-modified metabolome promotes susceptibility to bacterial coinfection.


Asunto(s)
Coinfección/inmunología , Helmintiasis/inmunología , Parasitosis Intestinales/inmunología , Mucosa Intestinal/metabolismo , Metaboloma , Infecciones por Salmonella/inmunología , Células Th2/inmunología , Animales , Coinfección/microbiología , Coinfección/parasitología , Células HeLa , Humanos , Intestinos/microbiología , Intestinos/parasitología , Ratones , Ratones Endogámicos C57BL , Salmonella typhimurium/genética
3.
PLoS Negl Trop Dis ; 15(1): e0009052, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33471793

RESUMEN

Intestinal helminth infection can impair host resistance to co-infection with enteric bacterial pathogens. However, it is not known whether helminth drug-clearance can restore host resistance to bacterial infection. Using a mouse helminth-Salmonella co-infection system, we show that anthelmintic treatment prior to Salmonella challenge is sufficient to restore host resistance to Salmonella. The presence of the small intestine-dwelling helminth Heligmosomoides polygyrus at the point of Salmonella infection supports the initial establishment of Salmonella in the small intestinal lumen. Interestingly, if helminth drug-clearance is delayed until Salmonella has already established in the small intestinal lumen, anthelmintic treatment does not result in complete clearance of Salmonella. This suggests that while the presence of helminths supports initial Salmonella colonization, helminths are dispensable for Salmonella persistence in the host small intestine. These data contribute to the mechanistic understanding of how an ongoing or prior helminth infection can affect pathogenic bacterial colonization and persistence in the mammalian intestine.


Asunto(s)
Coinfección/microbiología , Coinfección/parasitología , Resistencia a la Enfermedad/fisiología , Helmintiasis/complicaciones , Parasitosis Intestinales/complicaciones , Nematospiroides dubius/fisiología , Salmonella/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal , Intestinos/microbiología , Intestinos/parasitología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Salmonella/complicaciones , Salmonella typhi
4.
Mucosal Immunol ; 11(4): 1039-1046, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29453411

RESUMEN

Intestinal helminths have well-characterized modulatory effects on mammalian immune pathways. Ongoing helminth infection has been associated with both the suppression of allergies and an altered susceptibility to microbial infections. Enteric helminths share a niche with the intestinal microbiota, and the presence of helminths alters the microbiota composition and the metabolic signature of the host. Recent studies have demonstrated that the helminth-modified intestinal microbiome has the capacity to modify host immune responses even in the absence of live helminth infection. This article discusses the mechanisms by which helminths modify the intestinal microbiome of mammals, and reviews the evidence for a helminth-modified microbiome directly influencing host immunity during infectious and inflammatory diseases. Understanding the multifaceted mechanisms that underpin helminth immunomodulation will pave the way for novel therapies to combat infectious and inflammatory diseases.


Asunto(s)
Helmintiasis/inmunología , Helmintos/inmunología , Infecciones/inmunología , Inflamación/inmunología , Intestinos/inmunología , Microbiota , Animales , Terapia Biológica/tendencias , Inmunidad Innata , Inmunomodulación , Intestinos/microbiología , Intestinos/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA