Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nutrients ; 12(7)2020 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-32708852

RESUMEN

The early life period is crucial for the maturation of the intestinal barrier, its immune system, and a life-long beneficial host-microbiota interaction. The study aims to assess the impact of a beneficial dietary (short-chain fructooligosaccharides, scFOS) supplementation vs. a detrimental dietary environment (such as mycotoxin deoxynivalenol, DON) on offspring intestinal immune system developmental profiles. Sows were given scFOS-supplemented or DON-contaminated diets during the last 4 weeks of gestation, whereas force-feeding piglets with DON was performed during the first week of offspring life. Intestinal antigen-presenting cell (APC) subset frequency was analyzed by flow cytometry in the Peyer's patches and in lamina propria and the responsiveness of intestinal explants to toll-like receptor (TLR) ligands was performed using ELISA and qRT-PCR from post-natal day (PND) 10 until PND90. Perinatal exposure with scFOS did not affect the ontogenesis of APC. While it early induced inflammatory responses in piglets, scFOS further promoted the T regulatory response after TLR activation. Sow and piglet DON contamination decreased CD16+ MHCII+ APC at PND10 in lamina propria associated with IFNγ inflammation and impairment of Treg response. Our study demonstrated that maternal prebiotic supplementation and mycotoxin contamination can modulate the mucosal immune system responsiveness of offspring through different pathways.


Asunto(s)
Contaminación de Alimentos/análisis , Sistema Inmunológico/metabolismo , Membrana Mucosa/metabolismo , Micotoxinas/toxicidad , Prebióticos/administración & dosificación , Alimentación Animal/análisis , Alimentación Animal/toxicidad , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Animales Recién Nacidos , Citocinas/metabolismo , Dieta/veterinaria , Suplementos Dietéticos , Femenino , Interferón gamma/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/efectos de los fármacos , Micotoxinas/administración & dosificación , Oligosacáridos/administración & dosificación , Embarazo , Preñez/efectos de los fármacos , Preñez/inmunología , Receptores de IgG/metabolismo , Porcinos , Tricotecenos/administración & dosificación , Tricotecenos/toxicidad
2.
FASEB J ; 33(1): 301-313, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29975568

RESUMEN

Perinatal nutrition programs physiologic and metabolic functions, with consequences on the susceptibility to develop metabolic diseases in adulthood. The microbiota represents a key factor of such programming. We investigated whether perinatal prebiotic [short-chain fructooligosaccharides (scFOS)] supplementation improved adult metabolic health in association with microbiota changes in pigs used as human model. Sows were supplemented with scFOS or not during the end of gestation and the entire lactation, and offspring received scFOS accordingly during 1 mo after weaning. Pigs were then fed a standard diet for 5 mo, followed by a high-fat diet for 3 mo once adults. Perinatal scFOS supplementation induced a persistent modulation of the composition of the fecal microbiota in adulthood, notably by increasing the Prevotella genus. Meanwhile, scFOS animals displayed improved capacity to secrete glucagon-like peptide-1 and improved pancreas sensitivity to glucose without any changes in peripheral insulin sensitivity. Perinatal scFOS supplementation also increased ileal secretory IgA secretion and alkaline phosphatase activity and decreased TNF-α expression in adipose tissue. In conclusion, perinatal scFOS supplementation induced long-lasting modulation of intestinal microbiota and had beneficial consequences on the host physiology in adulthood. Our results highlight the key role of perinatal nutrition on later microbiota and host metabolic adaptation to an unbalanced diet.-Le Bourgot, C., Ferret-Bernard, S., Apper, E., Taminiau, B., Cahu, A., Le Normand, L., Respondek, F., Le Huërou-Luron, I., Blat, S. Perinatal short-chain fructooligosaccharides program intestinal microbiota and improve enteroinsular axis function and inflammatory status in high-fat diet-fed adult pigs.


Asunto(s)
Alimentación Animal/análisis , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Inflamación/veterinaria , Enfermedades Intestinales/veterinaria , Oligosacáridos/administración & dosificación , Enfermedades de los Porcinos/prevención & control , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Animales Recién Nacidos , Suplementos Dietéticos , Heces/microbiología , Femenino , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Inflamación/tratamiento farmacológico , Inflamación/etiología , Insulina/metabolismo , Enfermedades Intestinales/tratamiento farmacológico , Enfermedades Intestinales/etiología , Embarazo , Porcinos , Enfermedades de los Porcinos/etiología
3.
Sci Rep ; 8(1): 11656, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-30076313

RESUMEN

Clinical and animal studies have demonstrated beneficial effects of early consumption of dairy lipids and a probiotic, Lactobacillus fermentum (Lf), on infant gut physiology. The objective of this study was to investigate their long-term effects on gut microbiota and host entero-insular axis and metabolism. Piglets were suckled with a milk formula containing only plant lipids (PL), a half-half mixture of plant lipids and dairy lipids (DL), or this mixture supplemented with Lf (DL + Lf). They were weaned on a standard diet and challenged with a high-energy diet until postnatal day 140. DL and DL + Lf modulated gut microbiota composition and metabolism, increasing abundance of several Clostridia genera. Moreover, DL + Lf specifically decreased the faecal content of 2-oxoglutarate and lysine compared to PL and 5-aminovalerate compared to PL and DL. It also increased short-chain fatty acid concentrations like propionate compared to DL. Furthermore, DL + Lf had a beneficial effect on the endocrine function, enhancing caecal GLP-1 and GLP-1 meal-stimulated secretion. Correlations highlighted the consistent relationship between microbiota and gut physiology. Together, our results evidence a beneficial programming effect of DL + Lf in infant formula composition on faecal microbiota and entero-insular axis function.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Fórmulas Infantiles/química , Lípidos/administración & dosificación , Probióticos/administración & dosificación , Animales , Suplementos Dietéticos , Heces/microbiología , Humanos , Lactante , Limosilactobacillus fermentum/química , Lípidos/química , Leche/química , Probióticos/química , Porcinos , Porcinos Enanos
4.
FASEB J ; 32(4): 2160-2171, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29242276

RESUMEN

Butyrate can improve gut functions, whereas histone deacetylase inhibitors might alleviate neurocognitive alterations. Our aim was to assess whether oral butyrate could modulate brain metabolism and plasticity and if this would relate to gut function. Sixteen pigs were subjected to sodium butyrate (SB) supplementation via beverage water or water only [control (C)]. All pigs had blood sampled after 2 and 3 wk of treatment, and were subjected to a brain positron emission tomography after 3 wk. Animals were euthanized after 4 wk to sample pancreas, intestine, and brain for gut physiology and anatomy measurements, as well as hippocampal histology, Ki67, and doublecortin (DCX) immunohistochemistry. SB compared with C treatment triggered basal brain glucose metabolism changes in the nucleus accumbens and hippocampus ( P = 0.003), increased hippocampal granular cell layer volume ( P = 0.006), and neurogenesis (Ki67: P = 0.026; DCX: P = 0.029). After 2 wk of treatment, plasma levels of glucose, insulin, lactate, glucagon-like peptide 1, and peptide tyrosine tyrosine remained unchanged. After 3 wk, plasma levels of lactate were lower in SB compared with C animals ( P = 0.028), with no difference for glucose and insulin. Butyrate intake impacted very little gut anatomy and function. These results demonstrate that oral SB impacted brain functions with little effects on the gut.-Val-Laillet, D., Guérin, S., Coquery, N., Nogret, I., Formal, M., Romé, V., Le Normand, L., Meurice, P., Randuineau, G., Guilloteau, P., Malbert, C.-H., Parnet, P., Lallès, J.-P., Segain, J.-P. Oral sodium butyrate impacts brain metabolism and hippocampal neurogenesis, with limited effects on gut anatomy and function in pigs.


Asunto(s)
Ácido Butírico/farmacología , Hipocampo/efectos de los fármacos , Antagonistas de los Receptores Histamínicos/farmacología , Intestinos/efectos de los fármacos , Neurogénesis , Administración Oral , Animales , Glucemia/metabolismo , Ácido Butírico/administración & dosificación , Ácido Butírico/efectos adversos , Femenino , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Antagonistas de los Receptores Histamínicos/administración & dosificación , Antagonistas de los Receptores Histamínicos/efectos adversos , Insulina/sangre , Intestinos/fisiología , Ácido Láctico/sangre , Porcinos
5.
Eur J Nutr ; 57(2): 463-476, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27744547

RESUMEN

PURPOSE: Although composition of infant formula has been significantly improved during the last decade, major differences with the composition and structure of breast milk still remain and might affect nutrient digestion and gut biology. We hypothesized that the incorporation of dairy fat in infant formulas could modify their physiological impacts by making their composition closer to that of human milk. The effect of milk fat and milk fat globule membrane (MFGM) fragments in infant formulas on gut digestion, mucosal immunity and microbiota composition was evaluated. METHODS: Three formulas containing either (1) vegetable lipids stabilized only by proteins (V-P), (2) vegetable lipids stabilized by a mixture of proteins and MFGM fragments (V-M) and (3) a mixture of milk and vegetable lipids stabilized by a mixture of proteins and MFGM fragments (M-M) were automatically distributed to 42 newborn piglets until slaughter at postnatal day (PND) 7 or 28, and compared to a fourth group of sow's suckling piglets (SM) used as a breast-fed reference. RESULTS: At both PND, casein and ß-lactoglobulin digestion was reduced in M-M proximal jejunum and ileum contents compared to V-P and V-M ones leading to more numerous ß-Cn peptides in M-M contents. The IFNγ cytokine secretion of ConA-stimulated MLN cells from M-M piglets tended to be higher than in V-P ones at PND 7 and PND 28 and was closer to that of SM piglets. No dietary treatment effect was observed on IL-10 MLN cell secretion. Changes in faecal microbiota in M-M piglets resulted in an increase in Proteobacteria and Bacteroidetes and a decrease in Firmicutes phyla compared to V-P ones. M-M piglets showed higher abundances of Parabacteroides, Escherichia/Shigella and Klebsiella genus. CONCLUSIONS: The incorporation of both milk fat and MFGM fragments in infant formula modifies protein digestion, the dynamic of the immune system maturation and the faecal microbiota composition.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa , Inmunomodulación , Leche/química , Modelos Inmunológicos , Aceites de Plantas/administración & dosificación , Animales , Animales Recién Nacidos , Caseínas/administración & dosificación , Caseínas/metabolismo , Citocinas/metabolismo , Digestión , Heces/microbiología , Contenido Digestivo/química , Contenido Digestivo/microbiología , Glucolípidos/administración & dosificación , Glucolípidos/metabolismo , Glicoproteínas/administración & dosificación , Glicoproteínas/metabolismo , Humanos , Fórmulas Infantiles , Fenómenos Fisiológicos Nutricionales del Lactante , Recién Nacido , Lactoglobulinas/administración & dosificación , Lactoglobulinas/metabolismo , Gotas Lipídicas , Ganglios Linfáticos/crecimiento & desarrollo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Leche/metabolismo , Aceites de Plantas/metabolismo , Proteínas de Vegetales Comestibles/administración & dosificación , Proteínas de Vegetales Comestibles/metabolismo , Sus scrofa/crecimiento & desarrollo
6.
Br J Nutr ; 117(1): 83-92, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28115029

RESUMEN

Prebiotic supplementation modulates immune system development and function. However, less is known about the effects of maternal prebiotic consumption on offspring intestinal defences and immune system responsiveness. We investigated the effects of maternal short-chain fructo-oligosaccharide (scFOS) supplementation on mucin-secreting cells, ileal secretory IgA and cytokine secretion of weaned offspring and their humoral response to an oral vaccine against obligate intracellular Lawsonia intracellularis. Sows were fed a control diet (CTRL) or scFOS-supplemented diet during the last third of gestation and throughout lactation. At weaning, each litter was divided into two groups receiving a post-weaning CTRL or scFOS diet for a month. Pigs from the four groups were either non-vaccinated (n 16) or vaccinated (n 117) at day 33. Biomarkers related to intestinal defences and immune parameters were analysed 3 weeks later. SCFA production was assessed over time in suckling and weaned pigs. Maternal scFOS supplementation improved ileal cytokine secretions (interferon (IFN)-γ, P<0·05; IL-4, P=0·07) and tended to increase caecal goblet cell number (P=0·06). It increased IgA vaccine response in the serum (P<0·01) and ileal mucosa (P=0·08). Higher bacterial fermentative activity was observed during lactation (total faecal SCFA, P<0·001) and after weaning (colonic butyrate, P=0·10) in pigs from scFOS-supplemented mothers. No synergistic effect between maternal and post-weaning scFOS supplementation was observed. Therefore, maternal scFOS supplementation has long-lasting consequences by strengthening gut defences and immune response to a vaccine against an intestinal obligate intracellular pathogen. Prebiotic consumption by gestating and lactating mothers is decisive in modulating offspring intestinal immunity.


Asunto(s)
Vacunas Bacterianas/inmunología , Butiratos/sangre , Citocinas/metabolismo , Células Caliciformes/fisiología , Lawsonia (Bacteria) , Oligosacáridos/administración & dosificación , Alimentación Animal/análisis , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Citocinas/genética , Infecciones por Desulfovibrionaceae/microbiología , Infecciones por Desulfovibrionaceae/veterinaria , Dieta/veterinaria , Suplementos Dietéticos , Femenino , Fenómenos Fisiologicos Nutricionales Maternos , Oligosacáridos/química , Prebióticos , Porcinos , Enfermedades de los Porcinos/prevención & control
7.
PLoS One ; 9(9): e107508, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25238157

RESUMEN

Peripartum nutrition is crucial for developing the immune system of neonates. We hypothesized that maternal short-chain fructooligosaccharide (scFOS) supplementation could accelerate the development of intestinal immunity in offspring. Thirty-four sows received a standard or a scFOS supplemented diet (10 g scFOS/d) for the last 4 weeks of gestation and the 4 weeks of lactation. Colostrum and milk immunoglobulins (Ig) and TGFß1 concentrations were evaluated on the day of delivery and at d 6 and d 21 postpartum. Piglet intestinal structure, the immunologic features of jejunal and ileal Peyer's patches, and mesenteric lymph node cells were analysed at postnatal d 21. Short-chain fatty acid concentrations were measured over time in the intestinal contents of suckling and weaned piglets. Colostral IgA (P<0.05) significantly increased because of scFOS and TGFß1 concentrations tended to improve (P<0.1). IFNγ secretion by stimulated Peyer's patch and mesenteric lymph node cells, and secretory IgA production by unstimulated Peyer's patch cells were increased (P<0.05) in postnatal d 21 scFOS piglets. These differences were associated with a higher proportion of activated CD25+CD4α+ T cells among the CD4+ helper T lymphocytes (P<0.05) as assessed by flow cytometry. IFNγ secretion was positively correlated with the population of activated T lymphocytes (P<0.05). Total short-chain fatty acids were unchanged between groups during lactation but were higher in caecal contents of d 90 scFOS piglets (P<0.05); specifically propionate, butyrate and valerate. In conclusion, we demonstrated that maternal scFOS supplementation modified the intestinal immune functions in piglets in association with increased colostral immunity. Such results underline the key role of maternal nutrition in supporting the postnatal development of mucosal immunity.


Asunto(s)
Calostro/inmunología , Suplementos Dietéticos , Intestinos/inmunología , Oligosacáridos/farmacología , Porcinos/inmunología , Animales , Femenino , Intestinos/crecimiento & desarrollo , Embarazo , Fenómenos Fisiologicos de la Nutrición Prenatal , Porcinos/metabolismo
8.
J Nutr Biochem ; 25(10): 1090-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25087993

RESUMEN

We recently observed that maternal 18:3n-3 increases piglet jejunal permeability. We hypothesized that this would favor intestinal lipopolysaccharide (LPS) passage and alter gut immune system education toward this bacterial ligand. Sows were fed 18:3n-3 or 18:2n-6 diets throughout gestation and lactation. In each litter, two piglets were given oral Gram-negative spectrum antibiotic from post-natal day (PND) 14 to 28. All piglets were weaned on a regular diet at PND28. 18:3n-3 piglets exhibited greater jejunal permeability to FITC-LPS at PND28. Levels of 18:3n-3 but neither 20:5n-3 nor 20:4n-6 were greater in mesenteric lymph nodes (MLN) of 18:3n-3 piglets. Jejunal explant or MLN cell cytokine responses to LPS were not influenced by the maternal diet. Antibiotic increased jejunal permeability to FITC-LPS and lowered the level of 20:5n-3 in MLN, irrespective of the maternal diet. At PND52, no long-lasting effect of the maternal diet or antibiotic treatment on jejunal permeability was noticed. 18:3n-3 and 20:4n-6 levels were greater and lower, respectively, in MLN of 18:3n-3 compared to 18:2n-6 piglets. IL-10 production by MLN cells in response to LPS was greater in the 18:3n-3 group, irrespective of the neonatal antibiotic treatment. IL-8 secretion by jejunal explants in response to LPS was lower in antibiotic-treated 18:3n-3 compared to 18:2n-6 piglets. Finally, proportion of MHC class II(+) antigen-presenting cells was greater in 18:3n-3 than 18:2n-6 MLN cells. In conclusion, maternal 18:3n-3 directs the intestinal immune response to LPS toward an anti-inflammatory profile beyond the breastfeeding period; microbiota involvement seems dependent of the immune cells considered.


Asunto(s)
Ácidos Grasos Omega-3/farmacología , Yeyuno/efectos de los fármacos , Lipopolisacáridos/efectos adversos , Animales , Animales Recién Nacidos , Células Cultivadas , Citocinas/metabolismo , ADN Bacteriano/genética , Dieta/veterinaria , Ácidos Grasos Omega-6/farmacología , Femenino , Inflamación/patología , Yeyuno/inmunología , Yeyuno/microbiología , Fenómenos Fisiologicos Nutricionales Maternos , Venas Mesentéricas/efectos de los fármacos , Venas Mesentéricas/metabolismo , Microbiota , Permeabilidad , Embarazo , Resultado del Embarazo , Porcinos , Destete
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA