Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
J Proteome Res ; 14(4): 1911-9, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25751005

RESUMEN

Inflammatory bowel diseases are acute and chronic disabling inflammatory disorders with multiple complex etiologies that are not well-defined. Chronic intestinal inflammation has been linked to an energy-deficient state of gut epithelium with alterations in oxidative metabolism. Plasma-, urine-, stool-, and liver-specific metabonomic analyses are reported in a naïve T cell adoptive transfer (AT) experimental model of colitis, which evaluated the impact of long-chain n-3 polyunsaturated fatty acid (PUFA)-enriched diet. Metabolic profiles of AT animals and their controls under chow diet or fish oil supplementation were compared to describe the (i) consequences of inflammatory processes and (ii) the differential impact of n-3 fatty acids. Inflammation was associated with higher glycoprotein levels (related to acute-phase response) and remodeling of PUFAs. Low triglyceride levels and enhanced PUFA levels in the liver suggest activation of lipolytic pathways that could lead to the observed increase of phospholipids in the liver (including plasmalogens and sphingomyelins). In parallel, the increase in stool excretion of most amino acids may indicate a protein-losing enteropathy. Fecal content of glutamine was lower in AT mice, a feature exacerbated under fish oil intervention that may reflect a functional relationship between intestinal inflammatory status and glutamine metabolism. The decrease in Krebs cycle intermediates in urine (succinate, α-ketoglutarate) also suggests a reduction in the glutaminolytic pathway at a systemic level. Our data indicate that inflammatory status is related to this overall loss of energy homeostasis.


Asunto(s)
Traslado Adoptivo/métodos , Colitis/metabolismo , Colitis/prevención & control , Aceites de Pescado/farmacología , Metaboloma/fisiología , Metabolómica/métodos , Animales , Suplementos Dietéticos , Heces/química , Glutamina/análisis , Ácidos Cetoglutáricos/análisis , Hígado/metabolismo , Espectroscopía de Resonancia Magnética , Metaboloma/efectos de los fármacos , Ratones , Ácido Succínico/análisis , Orina/química
2.
Lipids Health Dis ; 12: 81, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23725086

RESUMEN

BACKGROUND: Inflammatory bowel diseases (IBD) are chronic intestinal inflammatory diseases affecting about 1% of western populations. New eating behaviors might contribute to the global emergence of IBD. Although the immunoregulatory effects of omega-3 fatty acids have been well characterized in vitro, their role in IBD is controversial. METHODS: The aim of this study was to assess the impact of increased fish oil intake on colonic gene expression, eicosanoid metabolism and development of colitis in a mouse model of IBD. Rag-2 deficient mice were fed fish oil (FO) enriched in omega-3 fatty acids i.e. EPA and DHA or control diet for 4 weeks before colitis induction by adoptive transfer of naïve T cells and maintained in the same diet for 4 additional weeks. Onset of colitis was monitored by colonoscopy and further confirmed by immunological examinations. Whole genome expression profiling was made and eicosanoids were measured by HPLC-MS/MS in colonic samples. RESULTS: A significant reduction of colonic proinflammatory eicosanoids in FO fed mice compared to control was observed. However, neither alteration of colonic gene expression signature nor reduction in IBD scores was observed under FO diet. CONCLUSION: Thus, increased intake of dietary FO did not prevent experimental colitis.


Asunto(s)
Colitis/dietoterapia , Colitis/metabolismo , Eicosanoides/metabolismo , Aceites de Pescado/farmacología , Intestinos/efectos de los fármacos , Animales , Colitis/genética , Colon/fisiopatología , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Ácidos Grasos Omega-3/farmacología , Aceites de Pescado/química , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/etiología , Mucosa Intestinal/metabolismo , Ratones Endogámicos C57BL , Ratones Mutantes , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Colaboradores-Inductores/patología
3.
J Clin Gastroenterol ; 46(9): 735-47, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22941427

RESUMEN

Inflammatory bowel diseases (IBD) including ulcerative colitis and Crohn's disease are chronically relapsing, immune-mediated disorders of the gastrointestinal tract. A major challenge in the treatment of IBD is the heterogenous nature of these pathologies. Both, ulcerative colitis and Crohn's disease are of multifactorial etiology and feature a complex interaction of host genetic susceptibility and environmental factors such as diet and gut microbiota. Genome-wide association studies identified disease-relevant single-nucleotide polymorphisms in approximately 100 genes, but at the same time twin studies also clearly indicated a strong environmental impact in disease development. However, attempts to link dietary factors to the risk of developing IBD, based on epidemiological observations showed controversial outcomes. Yet, emerging high-throughput technologies implying complete biological systems might allow taking nutrient-gene interactions into account for a better classification of patient subsets in the future. In this context, 2 new scientific fields, "nutrigenetics" and "nutrigenomics" have been established. "Nutrigenetics," studying the effect of genetic variations on nutrient-gene interactions and "Nutrigenomics," describing the impact of nutrition on physiology and health status on the level of gene transcription, protein expression, and metabolism. It is hoped that the integration of both research areas will promote the understanding of the complex gene-environment interaction in IBD etiology and in the long-term will lead to personalized nutrition for disease prevention and treatment. This review briefly summarizes data on the impact of nutrients on intestinal inflammation, highlights nutrient-gene interactions, and addresses the potential of applying "omic" technologies in the context of IBD.


Asunto(s)
Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Enfermedades Inflamatorias del Intestino/genética , Nutrigenómica , Dieta , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/terapia , Intestinos/microbiología
4.
PLoS One ; 8(8): e71661, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977107

RESUMEN

BACKGROUND: Obesity has been associated with a more severe disease course in inflammatory bowel disease (IBD) and epidemiological data identified dietary fats but not obesity as risk factors for the development of IBD. Crohn's disease is one of the two major IBD phenotypes and mostly affects the terminal ileum. Despite recent observations that high fat diets (HFD) impair intestinal barrier functions and drive pathobiont selection relevant for chronic inflammation in the colon, mechanisms of high fat diets in the pathogenesis of Crohn's disease are not known. The aim of this study was to characterize the effect of HFD on the development of chronic ileal inflammation in a murine model of Crohn's disease-like ileitis. METHODS: TNF(ΔARE/WT) mice and wildtype C57BL/6 littermates were fed a HFD compared to control diet for different durations. Intestinal pathology and metabolic parameters (glucose tolerance, mesenteric tissue characteristics) were assessed. Intestinal barrier integrity was characterized at different levels including polyethylene glycol (PEG) translocation, endotoxin in portal vein plasma and cellular markers of barrier function. Inflammatory activation of epithelial cells as well as immune cell infiltration into ileal tissue were determined and related to luminal factors. RESULTS: HFD aggravated ileal inflammation but did not induce significant overweight or typical metabolic disorders in TNF(ΔARE/WT). Expression of the tight junction protein Occludin was markedly reduced in the ileal epithelium of HFD mice independently of inflammation, and translocation of endotoxin was increased. Epithelial cells showed enhanced expression of inflammation-related activation markers, along with enhanced luminal factors-driven recruitment of dendritic cells and Th17-biased lymphocyte infiltration into the lamina propria. CONCLUSIONS: HFD feeding, independently of obesity, accelerated disease onset of small intestinal inflammation in Crohn's disease-relevant mouse model through mechanisms that involve increased intestinal permeability and altered luminal factors, leading to enhanced dendritic cell recruitment and promoted Th17 immune responses.


Asunto(s)
Enfermedad de Crohn/etiología , Enfermedad de Crohn/patología , Dieta Alta en Grasa/efectos adversos , Ileítis/etiología , Ileítis/patología , Obesidad/complicaciones , Tejido Adiposo/patología , Animales , Polaridad Celular , Enfermedad de Crohn/sangre , Enfermedad de Crohn/inmunología , Células Dendríticas/metabolismo , Endotoxinas/metabolismo , Enterocitos/metabolismo , Enterocitos/patología , Regulación de la Expresión Génica , Glucosa/metabolismo , Ileítis/sangre , Ileítis/inmunología , Íleon/metabolismo , Íleon/patología , Inflamación/complicaciones , Inflamación/genética , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Obesidad/sangre , Obesidad/inmunología , Obesidad/patología , Ocludina/metabolismo , Células Th17/citología , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA