Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Sci Rep ; 13(1): 21931, 2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-38081984

RESUMEN

Brown seaweeds have a rich bioactive content known to modulate biological processes, including the mucosal immune response and microbiota function, and may therefore have the potential to control enteric pathogens. Here, we tested if dietary seaweed (Saccharina latissima) supplementation could modulate pig gut health with a specific focus on parasitic helminth burdens, gut microbiota composition, and host immune response during a five week feeding period in pigs co-infected with the helminths Ascaris suum and Oesophagostomum dentatum. We found that inclusion of fermented S. latissima (Fer-SL) at 8% of the diet increased gut microbiota α-diversity with higher relative abundances of Firmicutes, Tenericutes, Verrucomicrobia, Spirochaetes and Elusimicrobia, and lower abundance of Prevotella copri. In the absence of helminth infection, transcription of immune-related genes in the intestine was only moderately influenced by dietary seaweed. However, Fer-SL modulated the transcriptional response to infection in a site-specific manner in the gut, with an attenuation of infection-induced gene expression in the jejunum and an amplification of gene expression in the colon. Effects on systemic immune parameters (e.g. blood lymphocyte populations) were limited, indicating the effects of Fer-SL were mainly localized to the intestinal tissues. Despite previously documented in vitro anti-parasitic activity against pig helminths, Fer-SL inclusion did not significantly affect parasite egg excretion or worm establishment. Collectively, our results show that although Fer-SL inclusion did not reduce parasite burdens, it may modify the gut environment during enteric parasite infection, which encourages continued investigations into the use of seaweeds or related products as novel tools to improve gut health.


Asunto(s)
Microbioma Gastrointestinal , Animales , Porcinos , Dieta , Oesophagostomum , Suplementos Dietéticos , Inmunidad
2.
BMC Biol ; 21(1): 138, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37316905

RESUMEN

BACKGROUND: The influence of diet on immune function and resistance to enteric infection and disease is becoming ever more established. Highly processed, refined diets can lead to inflammation and gut microbiome dysbiosis, whilst health-promoting dietary components such as phytonutrients and fermentable fibres are thought to promote a healthy microbiome and balanced mucosal immunity. Chicory (Cichorium intybus) is a leafy green vegetable rich in fibres and bioactive compounds that may promote gut health. RESULTS: Unexpectedly, we here show that incorporation of chicory into semisynthetic AIN93G diets renders mice susceptible to infection with enteric helminths. Mice fed a high level of chicory leaves (10% dry matter) had a more diverse gut microbiota, but a diminished type-2 immune response to infection with the intestinal roundworm Heligmosomoides polygyrus. Furthermore, the chicory-supplemented diet significantly increased burdens of the caecum-dwelling whipworm Trichuris muris, concomitant with a highly skewed type-1 immune environment in caecal tissue. The chicory-supplemented diet was rich in non-starch polysaccharides, particularly uronic acids (the monomeric constituents of pectin). In accordance, mice fed pectin-supplemented AIN93G diets had higher T. muris burdens and reduced IgE production and expression of genes involved in type-2 immunity. Importantly, treatment of pectin-fed mice with exogenous IL-25 restored type-2 responses and was sufficient to allow T. muris expulsion. CONCLUSIONS: Collectively, our data suggest that increasing levels of fermentable, non-starch polysaccharides in refined diets compromises immunity to helminth infection in mice. This diet-infection interaction may inform new strategies for manipulating the gut environment to promote resistance to enteric parasites.


Asunto(s)
Dieta , Infecciones por Nematodos , Animales , Ratones , Polisacáridos , Suplementos Dietéticos , Pectinas
3.
FASEB J ; 36(4): e22256, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35333423

RESUMEN

Proanthocyanidins (PAC) are dietary polyphenols with putative anti-inflammatory and immunomodulatory effects. However, whether dietary PAC can regulate type-2 immune function and inflammation at mucosal surfaces remains unclear. Here, we investigated if diets supplemented with purified PAC modulated pulmonary and intestinal mucosal immune responses during infection with the helminth parasite Ascaris suum in pigs. A. suum infection induced a type-2 biased immune response in lung and intestinal tissues, characterized by pulmonary granulocytosis, increased Th2/Th1 T cell ratios in tracheal-bronchial lymph nodes, intestinal eosinophilia, and modulation of genes involved in mucosal barrier function and immunity. Whilst PAC had only minor effects on pulmonary immune responses, RNA-sequencing of intestinal tissues revealed that dietary PAC significantly enhanced transcriptional responses related to immune function and antioxidant responses in the gut of both naïve and A. suum-infected animals. A. suum infection and dietary PAC induced distinct changes in gut microbiota composition, primarily in the jejunum and colon, respectively. Notably, PAC consumption substantially increased the abundance of Limosilactobacillus reuteri. In vitro experiments with porcine macrophages and intestinal epithelial cells supported a role for both PAC polymers and PAC-derived microbial metabolites in regulating oxidative stress responses in host tissues. Thus, dietary PAC may have distinct beneficial effects on intestinal health during infection with mucosal pathogens, while having a limited activity to modulate naturally-induced type-2 pulmonary inflammation. Our results shed further light on the mechanisms underlying the health-promoting properties of PAC-rich foods, and may aid in the design of novel dietary supplements to regulate mucosal inflammatory responses in the gastrointestinal tract.


Asunto(s)
Ascaris suum , Proantocianidinas , Animales , Antioxidantes , Ascaris suum/fisiología , Colon , Dieta , Inflamación , Pulmón , Proantocianidinas/farmacología , Porcinos
4.
J Nutr Biochem ; 100: 108887, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34655757

RESUMEN

Phytonutrients such as cinnamaldehyde (CA) have been studied for their effects on metabolic diseases, but their influence on mucosal inflammation and immunity to enteric infection are not well documented. Here, we show that consumption of CA in mice significantly down-regulates transcriptional pathways connected to inflammation in the small intestine, and alters T-cell populations in mesenteric lymph nodes. During infection with the enteric helminth Heligomosomoides polygyrus, CA treatment attenuated infection-induced changes in biological pathways connected to cell cycle and mitotic activity, and tended to reduce worm burdens. Mechanistically, CA did not appear to exert activity through a prebiotic effect, as CA treatment did not significantly change the composition of the gut microbiota. Instead, in vitro experiments showed that CA directly induced xenobiotic metabolizing pathways in intestinal epithelial cells and suppressed endotoxin-induced inflammatory responses in macrophages. Collectively, our results show that CA down-regulates inflammatory pathways in the intestinal mucosa and can limit the pathological response to enteric infection. These properties appear to be largely independent of the gut microbiota, and instead connected to the ability of CA to induce antioxidant pathways in intestinal cells. Our results encourage further investigation into the use of CA and related phytonutrients as functional food components to promote intestinal health in humans and animals.


Asunto(s)
Acroleína/análogos & derivados , Suplementos Dietéticos , Inflamación/inmunología , Intestino Delgado/metabolismo , Fitoquímicos/administración & dosificación , Infecciones por Strongylida/inmunología , Acroleína/administración & dosificación , Acroleína/farmacología , Animales , Células Cultivadas , Femenino , Microbioma Gastrointestinal , Inmunidad Mucosa , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/inmunología , Ganglios Linfáticos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Nematospiroides dubius , Fitoquímicos/farmacología , Linfocitos T/inmunología , Transcripción Genética , Transcriptoma , Xenobióticos/metabolismo
5.
JPEN J Parenter Enteral Nutr ; 46(3): 592-599, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33988859

RESUMEN

BACKGROUND: In the first weeks after birth, enteral feeding and bacterial colonization interact to influence gut maturation in preterm infants. Bovine colostrum (BC) has been suggested as a relevant supplementary diet when own mother's milk (MM) is insufficient or absent. This pilot trial tests whether the supplement type, BC or donor human milk (DM), affects gut colonization in preterm infants during the first week of life. METHODS: On day 7, fecal samples were collected from preterm infants (n = 24) fed BC or DM as a supplement to MM. The gut microbiome (GM) was analyzed by 16S ribosomal RNA amplicon sequencing. Correlations between the relative abundance of specific bacterial taxa and blood chemistry variables, including amino acids, were explored. RESULTS: BC-supplemented infants showed a lower relative abundance of the families Lactobacillaceae and Enterococcaceae than DM infants. Planococcaceae were more abundant in infants delivered by cesarean birth vs vaginally. The relative abundance of bacterial families, specifically Enterobacteriaceae, correlated negatively with plasma levels of multiple essential and nonessential amino acids (valine, isoleucine, lysine, histidine, and arginine). CONCLUSION: The nature of nutrition supplements (BC or DM) just after birth may affect GM development and nutrient metabolism in the neonatal period of preterm infants. The exploratory nature of our study calls for confirmation of these results and their possible long-term clinical implications for preterm infants.


Asunto(s)
Calostro , Microbioma Gastrointestinal , Animales , Bovinos , Femenino , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Leche Humana , Proyectos Piloto , Embarazo
6.
J Anim Sci Biotechnol ; 12(1): 85, 2021 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-34281627

RESUMEN

BACKGROUND: The direct use of medical zinc oxide in feed will be abandoned after 2022 in Europe, leaving an urgent need for substitutes to prevent post-weaning disorders. RESULTS: This study investigated the effect of using rapeseed-seaweed blend (rapeseed meal added two brown macroalgae species Ascophylum nodosum and Saccharina latissima) fermented by lactobacilli (FRS) as feed ingredients in piglet weaning. From d 28 of life to d 85, the piglets were fed one of three different feeding regimens (n = 230 each) with inclusion of 0%, 2.5% and 5% FRS. In this period, no significant difference of piglet performance was found among the three groups. From a subset of piglets (n = 10 from each treatment), blood samples for hematology, biochemistry and immunoglobulin analysis, colon digesta for microbiome analysis, and jejunum and colon tissues for histopathological analyses were collected. The piglets fed with 2.5% FRS manifested alleviated intraepithelial and stromal lymphocytes infiltration in the gut, enhanced colon mucosa barrier relative to the 0% FRS group. The colon microbiota composition was determined using V3 and V1-V8 region 16S rRNA gene amplicon sequencing by Illumina NextSeq and Oxford Nanopore MinION, respectively. The two amplicon sequencing strategies showed high consistency between the detected bacteria. Both sequencing strategies indicated that inclusion of FRS reshaped the colon microbiome of weaned piglets with increased Shannon diversity. Prevotella stercorea was verified by both methods to be more abundant in the piglets supplied with FRS feed, and its abundance was positively correlated with colonic mucosa thickness but negatively correlated with blood concentrations of leucocytes and IgG. CONCLUSIONS: FRS supplementation relieved the gut lymphocyte infiltration of the weaned piglets, improved the colon mucosa barrier with altered microbiota composition. Increasing the dietary inclusion of FRS from 2.5% to 5% did not lead to further improvements.

7.
Sci Rep ; 10(1): 21204, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33273678

RESUMEN

Atopic dermatitis is a chronic eczema commonly observed among children in Western countries. The gut microbiota is a significant factor in the pathogenesis, and ways to promote intestinal colonizers with anti-inflammatory capabilities are therefore favorable. The present study addressed the effects of a prebiotic, xylooligosaccharide (XOS), on the gut microbiota and ear inflammation in an oxazolone-induced dermatitis model in BALB/c mice. Mice were fed a XOS supplemented or a control diet throughout the experiment. Ear thickness and clinical skin inflammation were scored blindly after three weeks topical challenge with 0.4% oxazolone. The mice were divided into high and low responders to oxazolone-induced dermatitis based on clinical inflammation and histological evaluation of ear biopsies, and significantly fewer high responders were present in the XOS fed group. In addition, XOS fed mice had higher abundance of Prevotella spp. in their gut microbiota compared to the control fed mice. Serum IgE and ear tissue cytokine levels correlated significantly with the clinical scores, and with the abundance of Prevotella spp. The strong association between the low-responding phenotype and high abundance of Prevotella spp., indicates an alleviating effect of this intestinal colonizer in allergic sensitization. Prevotella should be considered as a relevant target for future microbiota-directed treatment strategies in atopic patients.


Asunto(s)
Dermatitis Atópica/terapia , Suplementos Dietéticos , Microbioma Gastrointestinal , Oxazolona/toxicidad , Prebióticos , Prevotella/crecimiento & desarrollo , Animales , Dermatitis Atópica/sangre , Dermatitis Atópica/inducido químicamente , Modelos Animales de Enfermedad , Oído , Femenino , Inmunoglobulina E/sangre , Ratones , Ratones Endogámicos BALB C
8.
Animals (Basel) ; 10(1)2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31952154

RESUMEN

The feeding of medicinal zinc oxide (ZnO) to weaner piglets will be phased out after 2022 in Europe, leaving pig producers without options to manage post-weaning disorders. This study assessed whether rapeseed meal, fermented alone (FRM) or co-fermented with a single (Ascophylum nodosum; FRMA), or two (A. nodossum and Saccharina latissima; FRMAS) brown macroalagae species, could improve weaner piglet performance and stimulate intestinal development as well as maturation of gut microbiota in the absence of in-feed zinc. Weaned piglets (n = 1240) were fed, during 28-85 days of age, a basal diet with no additives (negative control; NC), 2500 ppm in-feed ZnO (positive control; PC), FRM, FRMA or FRMAS. Piglets fed FRM and FRMA had a similar or numerically improved, respectively, production performance compared to PC piglets. Jejunal villus development was stimulated over NC in PC, FRM and FRMAS (gender-specific). FRM enhanced colon mucosal development and reduced signs of intestinal inflammation. All fermented feeds and PC induced similar changes in the composition and diversity of colon microbiota compared to NC. In conclusion, piglet performance, intestinal development and health indicators were sustained or numerically improved when in-feed zinc was replaced by FRM.

9.
BMC Microbiol ; 19(1): 277, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31823731

RESUMEN

BACKGROUND: Childhood malnutrition is a global health challenge associated with multiple adverse consequences, including delayed maturation of the gut microbiota (GM) which might induce long-term immune dysfunction and stunting. To understand GM dynamics during malnutrition and subsequent re-feeding, we used a piglet model with a malnutrition-induced phenotype similar to humans. Piglets were weaned at the age of 4 weeks, fed a nutritionally optimal diet for 1 week post-weaning before being fed a pure maize diet for 7 weeks to induce symptoms of malnutrition. After malnourishment, the piglets were re-fed using different regimes all based on general food aid products, namely Corn-Soy blend (CSB) fortified with phosphorus (CSB+), CSB fortified with phosphorus and skim milk powder (CSB++) and CSB fortified with phosphorus and added whey permeate (CSB + P). RESULTS: Malnourishment had profound impact on the GM of the piglets leading to a less diverse GM dominated especially by Akkermansia spp. as determined by 16S rRNA gene amplicon sequencing. All three re-feeding regimes partly restored GM, leading to a more diverse GM compositionally closer to that of well-nourished piglets. This effect was even more pronounced for CSB++ compared to CSB+ and CSB + P. CONCLUSION: The GM of piglets were profoundly disturbed by malnourishment resulting in significantly increased abundance of Akkermansia spp. CSB++ may have superior effect on recovering GM diversity compared to the two other food aid products used in this study.


Asunto(s)
Alimentación Animal/análisis , Disbiosis , Microbioma Gastrointestinal , Desnutrición/complicaciones , Factores de Edad , Animales , Bacterias/clasificación , Niño , Modelos Animales de Enfermedad , Femenino , Humanos , Desnutrición/microbiología , Leche , Fósforo/administración & dosificación , ARN Ribosómico 16S/genética , Glycine max , Porcinos , Destete , Proteína de Suero de Leche/administración & dosificación , Zea mays
10.
Food Res Int ; 125: 108644, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31554129

RESUMEN

Potato fiber is a side product in starch manufacturing rich in dietary fibers such as pectin, cellulose, hemicellulose and resistant starch. So far, the beneficial properties of potato fiber have been poorly characterized. This study investigated the effect of FiberBind 400, a commercial potato fiber product, on survival of probiotic Lactobacillus strains at simulated gastric conditions and on the composition and metabolic activity of the gut microbiota, using the TIM-2 colon model. Resistant starch and native starch from potato were used as reference substrates. FiberBind 400 had an ability to improve survival of the four tested strains, Lactobacillus fermentum PCC®, L. rhamnosus LGG®, L. reuteri RC-14® and L. paracasei F-19® in a strain-dependent way. The highest effect was observed for L. fermentum PCC® and L. rhamnosus LGG®. The effect of starches on bacterial survival was insignificant. Composition of the fecal microbiota in TIM-2 fermentations was assessed by high-throughput sequencing of 16S rRNA gene amplicon. Fermentation of FiberBind 400 resulted in more diverse microbial communities compared to starches. Changes in microbial abundances specifically mediated by FiberBind 400, included increases in the genera Lachnospira, Butyrivibrio, Mogibacterium, Parabacteroides, Prevotella and Desulfovibrio, and the species B. ovatus, as well as decreases in Ruminococcus torques and unassigned Ruminococcus spp. Shifts in other bacterial populations, such as increased abundances of Oscillospira, Enterococcus, Bacteroidales, Citrobacter, along with reduction of Roseburia, Ruminococcus, and Faecalibacterium prausnitzii were not significantly different between the substrates. Cumulative production of individual short-chain fatty acids was similar between potato fiber and starches. The study demonstrated that FiberBind 400 had a potential to protect probiotic Lactobacillus strains during the passage through the gastrointestinal tract and selectively modulate the gut bacterial populations. This knowledge can support application of potato fiber as a functional food ingredient with added biological benefits.


Asunto(s)
Fibras de la Dieta/administración & dosificación , Microbioma Gastrointestinal/fisiología , Lactobacillus/fisiología , Probióticos , Solanum tuberosum/química , Digestión , Ácidos Grasos/biosíntesis , Ácidos Grasos Volátiles , Heces/microbiología , Fermentación , Alimentos Funcionales , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Lactobacillus/efectos de los fármacos , Lactobacillus/crecimiento & desarrollo , Especificidad de la Especie , Almidón/administración & dosificación
11.
Diabetologia ; 62(9): 1689-1700, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31139852

RESUMEN

AIMS/HYPOTHESIS: Adopting a diet containing indigestible fibre compounds such as prebiotics to fuel advantageous bacteria has proven beneficial for alleviating inflammation. The effect of the microbial changes on autoimmunity, however, remains unknown. We studied the effects of prebiotic xylooligosaccharides (XOS) on pancreatic islet and salivary gland inflammation in NOD mice and tested whether these were mediated by the gut microbiota. METHODS: Mother and offspring mice were fed an XOS-supplemented diet until diabetes onset or weaning and were compared with a control-fed group. Diabetes incidence was monitored, insulitis and sialadenitis were scored in histological sections from adult mice, and several metabolic and immune variables were analysed in mice before the development of diabetes. Gut barrier function was assessed using an in vivo FITC-dextran permeability test. The importance of XOS-mediated gut microbial changes were evaluated in antibiotic-treated mice fed either XOS or control diet or given a faecal microbiota transplant from test animals. RESULTS: Diabetes onset was delayed in the XOS-fed mice, which also had fewer cellular infiltrations in their pancreatic islets and salivary glands. Interestingly, insulitis was most reduced in the XOS-fed groups when the mice were also treated with an antibiotic cocktail. There was no difference in sialadenitis between the dietary groups treated with antibiotics; the mice were protected by microbiota depletion regardless of diet. Faecal microbiota transplantation was not able to transfer protection. No major differences in glucose-insulin regulation, glucagon-like peptide-1, or short-chain fatty acid production were related to the XOS diet. The XOS diet did, however, reduce gut permeability markers in the small and large intestine. This was accompanied by a more anti-inflammatory environment locally and systemically, dominated by a shift from M1 to M2 macrophages, a higher abundance of activated regulatory T cells, and lower levels of induction of natural killer T cells and cytotoxic T cells. CONCLUSIONS/INTERPRETATION: Prebiotic XOS have microbiota-dependent effects on salivary gland inflammation and microbiota-independent effects on pancreatic islet pathology that are accompanied by an improved gut barrier that seems able to heighten control of intestinal diabetogenic antigens that have the potential to penetrate the mucosa to activate autoreactive immune responses.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Prebióticos , Animales , Autoinmunidad/fisiología , Suplementos Dietéticos , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Glucuronatos/uso terapéutico , Ratones , Ratones Endogámicos NOD , Oligosacáridos/uso terapéutico
12.
Sci Rep ; 8(1): 4318, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29531228

RESUMEN

Consumption of fermentable dietary fibers (DFs), which can induce growth and/or activity of specific beneficial populations, is suggested a promising strategy to modulate the gut microbiota and restore health in microbiota-linked diseases. Until today, inulin and fructo-oligosaccharides (FOS) are the best studied DFs, while little is known about the gut microbiota-modulating effects of ß-glucan, α-galactooligosaccharide (α-GOS) and xylo-oligosaccharide (XOS). Here, we used three continuous in vitro fermentation PolyFermS model to study the modulating effect of these DFs on two distinct human adult proximal colon microbiota, independently from the host. Supplementation of DFs, equivalent to a 9 g daily intake, induced a consistent metabolic response depending on the donor microbiota. Irrespective to the DF supplemented, the Bacteroidaceae-Ruminococcaceae dominated microbiota produced more butyrate (up to 96%), while the Prevotellaceae-Ruminococcaceae dominated microbiota produced more propionate (up to 40%). Changes in abundance of specific bacterial taxa upon DF supplementation explained the observed changes in short-chain fatty acid profiles. Our data suggest that the metabolic profile of SCFA profile may be the most suitable and robust read-out to characterize microbiota-modulating effects of a DF and highlights importance to understand the inter-individual response to a prebiotic treatment for mechanistic understanding and human application.


Asunto(s)
Fibras de la Dieta/microbiología , Fermentación , Microbioma Gastrointestinal , Prebióticos/microbiología , Adulto , Bacteroidaceae/metabolismo , Butiratos/metabolismo , Fibras de la Dieta/análisis , Ácidos Grasos/metabolismo , Humanos , Metaboloma , Prebióticos/análisis , Propionatos/metabolismo , Ruminococcus/metabolismo
13.
PLoS One ; 12(10): e0186546, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29028844

RESUMEN

Polyphenols are a class of bioactive plant secondary metabolites that are thought to have beneficial effects on gut health, such as modulation of mucosal immune and inflammatory responses and regulation of parasite burdens. Here, we examined the interactions between a polyphenol-rich diet supplement and infection with the enteric nematode Ascaris suum in pigs. Pigs were fed either a basal diet or the same diet supplemented with grape pomace (GP), an industrial by-product rich in polyphenols such as oligomeric proanthocyanidins. Half of the animals in each group were then inoculated with A. suum for 14 days to assess parasite establishment, acquisition of local and systemic immune responses and effects on the gut microbiome. Despite in vitro anthelmintic activity of GP-extracts, numbers of parasite larvae in the intestine were not altered by GP-supplementation. However, the bioactive diet significantly increased numbers of eosinophils induced by A. suum infection in the duodenum, jejunum and ileum, and modulated gene expression in the jejunal mucosa of infected pigs. Both GP-supplementation and A. suum infection induced significant and apparently similar changes in the composition of the prokaryotic gut microbiota, and both also decreased concentrations of isobutyric and isovaleric acid (branched-chain short chain fatty acids) in the colon. Our results demonstrate that while a polyphenol-enriched diet in pigs may not directly influence A. suum establishment, it significantly modulates the subsequent host response to helminth infection. Our results suggest an influence of diet on immune function which may potentially be exploited to enhance immunity to helminths.


Asunto(s)
Ascaris suum/fisiología , Dieta , Microbioma Gastrointestinal/efectos de los fármacos , Inmunidad Mucosa/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Polifenoles/farmacología , Animales , Antihelmínticos/farmacología , Especificidad de Anticuerpos , Colon/efectos de los fármacos , Colon/inmunología , Colon/metabolismo , Colon/microbiología , Suplementos Dietéticos , Ácidos Grasos/biosíntesis , Ácidos Grasos/química , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Vitis/química
14.
Vet Immunol Immunopathol ; 189: 43-52, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28669386

RESUMEN

Dietary phytonutrients such as cinnamaldehyde (CA) may contribute to immune function during pathogen infections, and CA has been reported to have positive effects on gut health when used as feed additive for livestock. Here, we investigated whether CA could enhance antibody production and specific immune responses during infection with an enteric pathogen. We examined the effect of dietary CA on plasma antibody levels in parasite-naïve pigs, and subsequently acquisition of humoral immune responses during infection with the parasitic nematode Ascaris suum. Parasite-naïve pigs fed diets supplemented with CA had higher levels of total IgA and IgG in plasma, and A. suum-infected pigs fed CA had higher levels of parasite-specific IgM and IgA in plasma 14days post-infection. Moreover, dietary CA increased expression of genes encoding the B-cell marker CD19, sodium/glucose co-transporter1 (SCA5L1) and glucose transporter 2 (SLC2A2) in the jejunal mucosa of A.suum-infected pigs. Dietary CA induced only limited changes in the composition of the prokaryotic gut microbiota of A. suum-infected pigs, and in vitro experiments showed that CA did not directly induce proliferation or increase secretion of IgG and IgA from lymphocytes. Our results demonstrate that dietary CA can significantly enhance acquisition of specific immune responses in pigs. The underlying mechanism remains obscure, but apparently does not derive simply from direct contact between CA and host lymphocytes and appears to be independent of the gut microbiota.


Asunto(s)
Acroleína/análogos & derivados , Anticuerpos Antihelmínticos/inmunología , Ascariasis/veterinaria , Ascaris suum/inmunología , Inmunidad Humoral/efectos de los fármacos , Síndrome Respiratorio y de la Reproducción Porcina/parasitología , Acroleína/uso terapéutico , Animales , Anticuerpos Antihelmínticos/sangre , Ascariasis/inmunología , Suplementos Dietéticos , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Inmunidad Humoral/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Masculino , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Porcinos/inmunología , Porcinos/parasitología
15.
Am J Physiol Gastrointest Liver Physiol ; 309(5): G310-23, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26138468

RESUMEN

It is unclear when and how to start enteral feeding for preterm infants when mother's milk is not available. We hypothesized that early and slow advancement with either formula or bovine colostrum stimulates gut maturation and prevents necrotizing enterocolitis (NEC) in preterm pigs, used as models for preterm infants. Pigs were given either total parenteral nutrition (TPN, n = 14) or slowly advancing volumes (16-64 ml·kg(-1)·day(-1)) of preterm infant formula (IF, n = 15) or bovine colostrum (BC, n = 13), both given as adjunct to parenteral nutrition. On day 5, both enteral diets increased intestinal mass (27 ± 1 vs. 22 ± 1 g/kg) and glucagon-like peptide 2 release, relative to TPN (P < 0.05). The incidence of mild NEC lesions was higher in IF than BC and TPN pigs (60 vs. 0 and 15%, respectively, P < 0.05). Only the IF pigs showed reduced gastric emptying and gastric inhibitory polypeptide release, and increased tissue proinflammatory cytokine levels (IL-1ß and IL-8, P < 0.05) and expression of immune-related genes (AOAH, LBP, CXCL10, TLR2), relative to TPN. The IF pigs also showed reduced intestinal villus-to-crypt ratio, lactose digestion, and some plasma amino acids (Arg, Cit, Gln, Tyr, Val), and higher intestinal permeability, compared with BC pigs (all P < 0.05). Colonic microbiota analyses showed limited differences among groups. Early feeding with formula induces intestinal dysfunction whereas bovine colostrum supports gut maturation when mother's milk is absent during the first week after preterm birth. A diet-dependent feeding guideline may be required for newborn preterm infants.


Asunto(s)
Alimentación con Biberón , Calostro/metabolismo , Enterocolitis Necrotizante/veterinaria , Mucosa Intestinal/metabolismo , Aminoácidos/sangre , Animales , Bovinos , Citocinas/metabolismo , Femenino , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 2 Similar al Glucagón/metabolismo , Intestinos/patología , Embarazo , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA