Littérature scientifique sur le sujet « Α-Galacto-Oligosaccharides (α-GOS) »

Créez une référence correcte selon les styles APA, MLA, Chicago, Harvard et plusieurs autres

Choisissez une source :

Consultez les listes thématiques d’articles de revues, de livres, de thèses, de rapports de conférences et d’autres sources académiques sur le sujet « Α-Galacto-Oligosaccharides (α-GOS) ».

À côté de chaque source dans la liste de références il y a un bouton « Ajouter à la bibliographie ». Cliquez sur ce bouton, et nous générerons automatiquement la référence bibliographique pour la source choisie selon votre style de citation préféré : APA, MLA, Harvard, Vancouver, Chicago, etc.

Vous pouvez aussi télécharger le texte intégral de la publication scolaire au format pdf et consulter son résumé en ligne lorsque ces informations sont inclues dans les métadonnées.

Articles de revues sur le sujet "Α-Galacto-Oligosaccharides (α-GOS)"

1

Kruger, Claire, Yuting Zhou, Bjorn A. Thorsrud, Fanny Morel-Despeisse et Eric Chappuis. « Safety evaluation of α-galacto-oligosaccharides for use in infant formulas investigated in neonatal piglets ». Toxicology Research and Application 1 (1 janvier 2017) : 239784731772282. http://dx.doi.org/10.1177/2397847317722828.

Texte intégral
Résumé :
Galacto-oligosaccharide (GOS), comprising galactoses with a glucose or sucrose, is a family of nondigestible oligosaccharides. The present study evaluates the safety of an α-GOS product (P-GOS® P) in a neonatal piglet model for 3 weeks. Three days after birth, neonatal piglets were divided into control and treated groups and provided with swine milk replacers in the absence and presence of 8 mg/mL—of the α-GOS product, respectively. An increase in the weight of the large intestines in treated males was noted, which is a common finding in studies of animals fed nondigestible oligosaccharides. There were no α-GOS product-related adverse effects in the piglets in terms of clinical signs, body weights, feed consumption, clinical chemistry, hematology, organ weights, or histopathology. The study demonstrated that formula supplemented with 8 mg/mL of P-GOS P is safe and well tolerated in neonatal piglets and supports the safe use of P-GOS P in infant formulas.
Styles APA, Harvard, Vancouver, ISO, etc.
2

Djouzi, Zakia, et Claude Andlueux. « Compared effects of three oligosaccharides on metabolism of intestinal microflora in rats inoculated with a human faecal flora ». British Journal of Nutrition 78, no 2 (août 1997) : 313–24. http://dx.doi.org/10.1079/bjn19970149.

Texte intégral
Résumé :
Using germ-free rats inoculated with a human faecal flora (gnotobiotic rats), the effects of three oligosaccharides (β-fructo-oligosaccharides (FOS), β-galacto-oligosaccharides (TOS) and α-gluco-oligosaccharides (GOS)) on intestinal bacterial metabolism were compared. The animals were fed on either a control diet or diets containing 4Og/kg of GOS, FOS or TOS. FOS and TOS were the preferred growth substrates for Bifidobacteria which increased in number by 2 log values in faeces of rats when compared with rats fed on GOS or control diets. Ingestion of TOS specifically induced hydrolysis of the substrate, and did not modify the activity of any other enzymes measured in the caecum. GOS led to a non-specific enzymic induction of β-galactosidase (EC 3.2.1.23), β-glucosidase (EC 3.2.1.21) and α-glucosidase (EC 3.2.1.20) activities whereas β-glucuronidase (EC 3.2.1.31) was lowered. Compared with the control group, FOS and TOS diets led to a significant increase in H2and CH4excretion; the GOS diet increased only CH4. Analysis of caecal contents revealed a decrease in pH for all diets compared with controls. Total short-chain fatty acid (SCFA) concentration increased significantly in all groups, but the SCFA profile differed between treatment groups. It was concluded that the three oligosaccharides studied had different effects which may be linked to their chemical structure. Some of these effects may be beneficial to human health
Styles APA, Harvard, Vancouver, ISO, etc.
3

Chen, Tso-Hsiao, Chung-Te Liu, Chung-Yi Cheng, Yuh-Mou Sue, Nai-Jen Huang et Cheng-Hsien Chen. « Oligosaccharides Ameliorate Acute Kidney Injury by Alleviating Cluster of Differentiation 44-Mediated Immune Responses in Renal Tubular Cells ». Nutrients 14, no 4 (11 février 2022) : 760. http://dx.doi.org/10.3390/nu14040760.

Texte intégral
Résumé :
Acute kidney injury (AKI) is a sudden episode of kidney damage that commonly occurs in patients admitted to hospitals. To date, no ideal treatment has been developed to reduce AKI severity. Oligo-fucoidan (FC) interferes with renal tubular cell surface protein cluster of differentiation 44 (CD44) to prevent renal interstitial fibrosis; however, the influence of oligosaccharides on AKI remains unknown. In this study, FC, galacto-oligosaccharide (GOS), and fructo-oligosaccharide (FOS) were selected to investigate the influence of oligosaccharides on AKI. All three oligosaccharides have been proven to be partially absorbed by the intestine. We found that the oligosaccharides dose-dependently reduced CD44 antigenicity and suppressed the hypoxia-induced expression of CD44, phospho-JNK, MCP-1, IL-1β, and TNF-α in NRK-52E renal tubular cells. Meanwhile, CD44 siRNA transfection and JNK inhibitor SP600125 reduced the hypoxia-induced expression of phospho-JNK and cytokines. The ligand of CD44, hyaluronan, counteracted the influence of oligosaccharides on CD44 and phospho-JNK. At 2 days post-surgery for ischemia–reperfusion injury, oligosaccharides reduced kidney inflammation, serum creatine, MCP-1, IL-1β, and TNF-α in AKI mice. At 7 days post-surgery, kidney recovery was promoted. These results indicate that FC, GOS, and FOS inhibit the hypoxia-induced CD44/JNK cascade and cytokines in renal tubular cells, thereby ameliorating AKI and kidney inflammation in AKI mice. Therefore, oligosaccharide supplementation is a potential healthcare strategy for patients with AKI.
Styles APA, Harvard, Vancouver, ISO, etc.
4

Matsuki, T., S. Tajima, T. Hara, K. Yahagi, E. Ogawa et H. Kodama. « Infant formula with galacto-oligosaccharides (OM55N) stimulates the growth of indigenous bifidobacteria in healthy term infants ». Beneficial Microbes 7, no 4 (1 septembre 2016) : 453–61. http://dx.doi.org/10.3920/bm2015.0168.

Texte intégral
Résumé :
The objective of the study was to investigate whether an infant formula supplemented with galacto-oligosaccharides (GOS; OM55N) was able to stimulate the growth of indigenous bifidobacteria and to establish microbiota similar to that of breastfed infants. A randomised, double-blind, placebo-controlled trial was performed using 35 healthy term infants (31-54 days of age; 42±6 days) to determine whether infant formula with 0.3 g/dl GOS (OM55N) stimulated the growth of bifidobacteria in the infants’ guts. At the trial onset and 2 weeks after, the infants’ faecal samples were examined for microbiota composition (bacterial abundance and α-diversity) and faecal characteristics. Among the 35 infants, 5 were withdrawn and 8 were excluded from the final evaluation before breaking the blinding since the indigenous bifidobacteria were not detected at the trial onset. After 2 weeks, the abundance of Bifidobacteriaceae was significantly increased in the GOS feeding group compared to the control (+11.6±24.1% vs -3.9±13.0%; P=0.043). The Shannon index, which accounts for both abundance and evenness of the present species, was significantly decreased with GOS supplementation (-0.1±0.4 vs +0.4±0.4; P=0.014). Faecal characteristics such as pH and organic acids were similar in both groups, with no statistical differences. No adverse side effects related to the formula consumption were reported. Although the concentration of GOS was relatively low, the infant formula with GOS increased the abundance of bifidobacteria and resulted in a reduced α-diversity of the microbiota.
Styles APA, Harvard, Vancouver, ISO, etc.
5

Godínez-Méndez, Lucila A., Carmen M. Gurrola-Díaz, José Sergio Zepeda-Nuño, Natali Vega-Magaña, Rocio Ivette Lopez-Roa, Liliana Íñiguez-Gutiérrez, Pedro M. García-López, Mary Fafutis-Morris et Vidal Delgado-Rizo. « In Vivo Healthy Benefits of Galacto-Oligosaccharides from Lupinus albus (LA-GOS) in Butyrate Production through Intestinal Microbiota ». Biomolecules 11, no 11 (9 novembre 2021) : 1658. http://dx.doi.org/10.3390/biom11111658.

Texte intégral
Résumé :
Animal digestive systems host microorganism ecosystems, including integrated bacteria, viruses, fungi, and others, that produce a variety of compounds from different substrates with healthy properties. Among these substrates, α-galacto-oligosaccharides (GOS) are considered prebiotics that promote the grow of gut microbiota with a metabolic output of Short Chain Fatty Acids (SCFAs). In this regard, we evaluated Lupinus albus GOS (LA-GOS) as a natural prebiotic using different animal models. Therefore, the aim of this work was to evaluate the effect of LA-GOS on the gut microbiota, SCFA production, and intestinal health in healthy and induced dysbiosis conditions (an ulcerative colitis (UC) model). Twenty C57BL/6 mice were randomly allocated in four groups (n = 5/group): untreated and treated non-induced animals, and two groups induced with 2% dextran sulfate sodium to UC with and without LA-GOS administration (2.5 g/kg bw). We found that the UC treated group showed a higher goblet cell number, lower disease activity index, and reduced histopathological damage in comparison to the UC untreated group. In addition, the abundance of positive bacteria to butyryl-CoA transferase in gut microbiota was significantly increased by LA-GOS treatment, in healthy conditions. We measured the SCFA production with significant differences in the butyrate concentration between treated and untreated healthy groups. Finally, the pH level in cecum feces was reduced after LA-GOS treatment. Overall, we point out the in vivo health benefits of LA-GOS administration on the preservation of the intestinal ecosystem and the promotion of SCFA production.
Styles APA, Harvard, Vancouver, ISO, etc.
6

Cervera-Tison, M., L. E. Tailford, C. Fuell, L. Bruel, G. Sulzenbacher, B. Henrissat, J. G. Berrin, M. Fons, T. Giardina et N. Juge. « Functional Analysis of Family GH36 α-Galactosidases from Ruminococcus gnavus E1 : Insights into the Metabolism of a Plant Oligosaccharide by a Human Gut Symbiont ». Applied and Environmental Microbiology 78, no 21 (24 août 2012) : 7720–32. http://dx.doi.org/10.1128/aem.01350-12.

Texte intégral
Résumé :
ABSTRACTRuminococcus gnavusbelongs to the 57 most common species present in 90% of individuals. Previously, we identified an α-galactosidase (Aga1) belonging to glycoside hydrolase (GH) family 36 fromR. gnavusE1 (M. Aguilera, H. Rakotoarivonina, A. Brutus, T. Giardina, G. Simon, and M. Fons, Res. Microbiol. 163:14–21, 2012). Here, we identified a novel GH36-encoding gene from the same strain and termed itaga2. Althoughaga1showed a very simple genetic organization,aga2is part of an operon of unique structure, including genes putatively encoding a regulator, a GH13, two phosphotransferase system (PTS) sequences, and a GH32, probably involved in extracellular and intracellular sucrose assimilation. The 727-amino-acid (aa) deduced Aga2 protein shares approximately 45% identity with Aga1. Both Aga1 and Aga2 expressed inEscherichia colishowed strict specificity for α-linked galactose. Both enzymes were active on natural substrates such as melibiose, raffinose, and stachyose. Aga1 and Aga2 occurred as homotetramers in solution, as shown by analytical ultracentrifugation. Modeling of Aga1 and Aga2 identified key amino acids which may be involved in substrate specificity and stabilization of the α-linked galactoside substrates within the active site. Furthermore, Aga1 and Aga2 were both able to perform transglycosylation reactions with α-(1,6) regioselectivity, leading to the formation of product structures up to [Hex]12and [Hex]8, respectively. We suggest that Aga1 and Aga2 play essential roles in the metabolism of dietary oligosaccharides and could be used for the design of galacto-oligosaccharide (GOS) prebiotics, known to selectively modulate the beneficial gut microbiota.
Styles APA, Harvard, Vancouver, ISO, etc.
7

Lian, Puqiao, Paul A. J. Henricks, Harry J. Wichers, Gert Folkerts et Saskia Braber. « Differential Effects of Oligosaccharides, Antioxidants, Amino Acids and PUFAs on Heat/Hypoxia-Induced Epithelial Injury in a Caco-2/HT-29 Co-Culture Model ». International Journal of Molecular Sciences 24, no 2 (6 janvier 2023) : 1111. http://dx.doi.org/10.3390/ijms24021111.

Texte intégral
Résumé :
(1) Exposure of intestinal epithelial cells to heat and hypoxia causes a (heat) stress response, resulting in the breakdown of epithelial integrity. There are indications that several categories of nutritional components have beneficial effects on maintaining the intestinal epithelial integrity under stress conditions. This study evaluated the effect of nine nutritional components, including non-digestible oligosaccharides (galacto-oligosaccharides (GOS), fructo-oligosaccharides (FOS), chitosan oligosaccharides (COS)), antioxidants (α-lipoic acid (ALA), resveratrol (RES)), amino acids (l-glutamine (Glu), l-arginine (Arg)) and polyunsaturated fatty acids (PUFAs) (docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA)), on heat/hypoxia-induced epithelial injury. (2) Two human colonic cell lines, Caco-2 and HT-29, were co-cultured and pre-treated with the nutritional components for 48 h. After pre-treatment, the cells were exposed to heat/hypoxia (42 °C, 5% O2) for 2 h. Epithelial integrity was evaluated by measuring trans-epithelial electrical resistance (TEER), paracellular Lucifer Yellow (LY) permeability, and tight junction (TJ) protein expression. Heat stress and oxidative stress levels were evaluated by determining heat-shock protein-70 (HSP-70) expression and the concentration of the lipid peroxidation product malondialdehyde (MDA). (3) GOS, FOS, COS, ALA, RES, Arg, and EPA presented protective effects on epithelial damage in heat/hypoxia-exposed Caco-2/HT-29 cells by preventing the decrease in TEER, the increase in LY permeability, and/or decrease in TJ proteins zonula occludens-1 (ZO-1) and claudin-3 expression. COS, RES, and EPA demonstrated anti-oxidative stress effects by suppressing the heat/hypoxia-induced MDA production, while Arg further elevated the heat/hypoxia-induced increase in HSP-70 expression. (4) This study indicates that various nutritional components have the potential to counteract heat/hypoxia-induced intestinal injury and might be interesting candidates for future in vivo studies and clinical trials in gastrointestinal disorders related to heat stress and hypoxia.
Styles APA, Harvard, Vancouver, ISO, etc.
8

Pico, Joana, Natalia P. Vidal, Listiya Widjaja, Louis Falardeau, Lionel Albino et Mario M. Martinez. « Development and assessment of GC/MS and HPAEC/PAD methodologies for the quantification of α-galacto-oligosaccharides (GOS) in dry beans (Phaseolus vulgaris) ». Food Chemistry 349 (juillet 2021) : 129151. http://dx.doi.org/10.1016/j.foodchem.2021.129151.

Texte intégral
Styles APA, Harvard, Vancouver, ISO, etc.
9

Yao, Qianqian, Huiying Li, Yanan Gao, Nan Zheng, Véronique Delcenserie et Jiaqi Wang. « The Milk Active Ingredient, 2′-Fucosyllactose, Inhibits Inflammation and Promotes MUC2 Secretion in LS174T Goblet Cells In Vitro ». Foods 12, no 1 (1 janvier 2023) : 186. http://dx.doi.org/10.3390/foods12010186.

Texte intégral
Résumé :
In several mice inflammatory models, human milk oligosaccharides (HMOs) were shown to protect the intestinal barrier by promoting mucin secretion and suppressing inflammation. However, the functions of the individual HMOs in enhancing mucin expression in vivo have not been compared, and the related mechanisms are not yet to be clarified. In this study, we investigated the modulatory effects of 2′-fucosyllactose (2′-FL), 3′-sialyllactose (3′-SL), galacto-oligosaccharide (GOS) and lactose (Lac) on goblet cells’ functions in vitro. The appropriate dosage of the four chemicals was assessed in LS174T cells using the CCK-8 method. Then they were supplemented into a homeostasis and inflammatory environment to further investigate their effects under different conditions. Mucin secretion-related genes, including mucin 2 (MUC2), trefoil factor family 3 (TFF3), resistin-like β (RETNLB), carbohydrate sulfotransferase 5 (CHST5) and galactose-3-O-sulfotransferase 2 (GAL3ST2), in LS174T cells were detected using quantitative RT-qPCR. The results showed that 2′-FL (2.5 mg/mL, 72 h) was unable to increase MUC2 secretion in a steady-state condition. Comparatively, it exhibited a greater ability to improve mucin secretion under an inflammatory condition compared with GOS, demonstrated by a significant increase in TFF3 and CHST5 mRNA expression levels (p > 0.05). However, 3′-SL and Lac exhibited no effects on mucin secretion. To further investigate the underlying mechanism via which 2′-FL enhanced goblet cells’ secretion function, the NOD-like receptor family pyrin domain containing 6 (NLRP6) gene, which is closely related to MUC2 secretion, was silenced using the siRNA method. After silencing the NLRP6 gene, the mRNA expression levels of MUC2, TFF3 and CHST5 in the (2′-FL + tumor necrosis factor α (TNF-α) + NLRP6 siRNA) group were significantly decreased compared with the (2′-FL + TNF-α) group (p > 0.05), indicating that NLRP6 was essential for MUC2 expression in goblet cells. We further found that 2′-FL could significantly decrease toll-like receptor 4 (TLR4, p < 0.05), myeloid differential protein-88 (MyD88, p < 0.05) and nuclear factor kappa-B (NF-κB, p < 0.05) levels in LS174T inflammatory cells, even when the NLRP6 was silenced. Altogether, these results indicated that in goblet cells, 2′-FL exerts its function via multiple processes, i.e., by promoting mucin secretion through NLRP6 and suppressing inflammation by inhibiting the TLR4/MyD88/NF-κB pathway.
Styles APA, Harvard, Vancouver, ISO, etc.
10

Lalitsuradej, Ekasit, Sasithorn Sirilun, Phakkharawat Sittiprapaporn, Bhagavathi Sundaram Sivamaruthi, Komsak Pintha, Payungsak Tantipaiboonwong, Suchanat Khongtan, Pranom Fukngoen, Sartjin Peerajan et Chaiyavat Chaiyasut. « The Effects of Synbiotics Administration on Stress-Related Parameters in Thai Subjects—A Preliminary Study ». Foods 11, no 5 (6 mars 2022) : 759. http://dx.doi.org/10.3390/foods11050759.

Texte intégral
Résumé :
Urbanization influences our lifestyle, especially in fast-paced environments where we are more prone to stress. Stress management is considered advantageous in terms of longevity. The use of probiotics for psychological treatment has a small amount of diverse proven evidence to support this. However, studies on stress management in stressed subjects using synbiotics are still limited. The present study aimed to investigate the effects of synbiotics on stress in the Thai population. A total of 32 volunteers were enrolled and screened using a Thai Stress Test (TST) to determine their stress status. Participants were divided into the stressed and the non-stressed groups. Synbiotics preparation comprised a mixture of probiotics strains in a total concentration of 1 × 1010 CFU/day (5.0 × 109 CFU of Lactobacillus paracasei HII01 and 5.0 × 109 CFU of Bifidobacterium animalis subsp. lactis) and 10 g prebiotics (5 g galacto-oligosaccharides (GOS), and 5 g oligofructose (FOS)). All parameters were measured at baseline and after the 12th week of the study. In the stressed group, the administration of synbiotics significantly (p < 0.05) reduced the negative scale scores of TST, and tryptophan. In the non-stressed group, the synbiotics administration decreased tryptophan significantly (p < 0.05), whereas dehydroepiandrosterone sulfate (DHEA-S), tumor necrosis factor-α (TNF-α), 5-hydroxyindoleacetic acid (5-HIAA), and short-chain fatty acids (SCFAs), acetate and propionate were increased significantly (p < 0.05). In both groups, cortisol, and lipopolysaccharide (LPS) were reduced, whereas anti-inflammatory mediator interleukin-10 (IL-10) and immunoglobulin A (IgA) levels were increased. In conclusion, synbiotics administration attenuated the negative feelings via the negative scale scores of TST in stressed participants by modulating the HPA-axis, IL-10, IgA, and LPS. In comparison, synbiotics administration for participants without stress did not benefit stress status but showed remodeling SCFAs components, HPA-axis, and tryptophan catabolism.
Styles APA, Harvard, Vancouver, ISO, etc.
Nous offrons des réductions sur tous les plans premium pour les auteurs dont les œuvres sont incluses dans des sélections littéraires thématiques. Contactez-nous pour obtenir un code promo unique!

Vers la bibliographie