Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Mol Gastroenterol Hepatol ; 18(2): 101350, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38704148

RESUMEN

BACKGROUND & AIMS: Gut bacterial sphingolipids, primarily produced by Bacteroidetes, have dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells. Patients with inflammatory bowel disease display altered sphingolipids profiles in fecal samples. However, how bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remains unclear. METHODS: We used dextran sodium sulfate (DSS)-induced colitis in mice monocolonized with Bacteroides fragilis strains expressing or lacking sphingolipids to assess the influence of bacterial sphingolipids on intestinal inflammation using transcriptional, protein, and cellular analyses. Colonic explant and organoid were used to study the function of bacterial sphingolipids. Host mucosal immune cells and cytokines were profiled and characterized using flow cytometry, enzyme-linked immunosorbent assay, and Western blot, and cytokine function in vivo was investigated by monoclonal antibody injection. RESULTS: B fragilis sphingolipids exacerbated intestinal inflammation. Mice monocolonized with B fragilis lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin (IL)-22 by ILC3. Mice colonized with B fragilis lacking sphingolipids following DSS treatment showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production. Protection against DSS colitis associated with B fragilis lacking sphingolipids was reversed on IL22 blockade. Furthermore, bacterial sphingolipids restricted epithelial IL18 production following DSS treatment and interfered with IL22 production by a subset of ILC3 cells expressing both IL18R and major histocompatibility complex class II. CONCLUSIONS: B fragilis-derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL18 expression and concomitantly suppressing the production of IL22 by ILC3 cells.


Asunto(s)
Bacteroides fragilis , Colitis , Sulfato de Dextran , Interleucina-22 , Interleucinas , Esfingolípidos , Animales , Esfingolípidos/metabolismo , Interleucinas/metabolismo , Ratones , Colitis/inmunología , Colitis/patología , Colitis/inducido químicamente , Colitis/microbiología , Colitis/metabolismo , Sulfato de Dextran/toxicidad , Bacteroides fragilis/inmunología , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Factor de Transcripción STAT3/metabolismo , Ratones Endogámicos C57BL
2.
Front Immunol ; 12: 657622, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33912185

RESUMEN

Gut immune cells have been increasingly appreciated as important players in the central nervous system (CNS) autoimmunity in animal models of multiple sclerosis (MS). Among the gut immune cells, innate lymphoid cell type 3 (ILC3) is of special interest in MS research, as they represent the innate cell counterpart of the major pathogenic cell population in MS, i.e. T helper (Th)17 cells. Importantly, these cells have been shown to stimulate regulatory T cells (Treg) and to counteract pathogenic Th17 cells in animal models of autoimmune diseases. Besides, they are also well known for their ability to stabilize the intestinal barrier and to shape the immune response to the gut microbiota. Thus, proper maintenance of the intestinal barrier and the establishment of the regulatory milieu in the gut performed by ILC3 may prevent activation of CNS antigen-specific Th17 cells by the molecular mimicry. Recent findings on the role of ILC3 in the gut-CNS axis and their relevance for MS pathogenesis will be discussed in this paper. Possibilities of ILC3 functional modulation for the benefit of MS patients will be addressed, as well.


Asunto(s)
Encéfalo/metabolismo , Tracto Gastrointestinal/metabolismo , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Esclerosis Múltiple/etiología , Esclerosis Múltiple/metabolismo , Animales , Autoinmunidad , Biomarcadores , Encéfalo/inmunología , Comunicación Celular/inmunología , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Retroalimentación Fisiológica , Tracto Gastrointestinal/inmunología , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunomodulación , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA