fructooligosaccharide and Disease-Models--Animal

fructooligosaccharide has been researched along with Disease-Models--Animal* in 27 studies

Trials

1 trial(s) available for fructooligosaccharide and Disease-Models--Animal

ArticleYear
Short-chain fructooligosaccharides influence insulin sensitivity and gene expression of fat tissue in obese dogs.
    The Journal of nutrition, 2008, Volume: 138, Issue:9

    Dietary fibers may modulate insulin resistance and glucose homeostasis in dogs. Their efficacy is, however, dependent on their origin, physical properties, and fermentability in the large bowel. Eight healthy Beagle dogs were fed a commercial diet at twice their maintenance requirements until they became obese. They were then maintained in the obese state and used in a cross-over design study to evaluate the effects of short-chain fructooligosaccharide (scFOS) supplementation (1% wt:wt dry matter in the diet). The euglycemic hyperinsulinemic clamp technique was performed before and after fattening and at the end of each 6-wk cross-over period. Fat tissue biopsies were taken in food-deprived and postprandial phases to measure mRNA abundance of genes involved with fatty acid, glucose metabolism, or inflammation. Insulin resistance appeared progressively with fattening and the rate of glucose infusion during euglycemic clamp was lower (P < 0.05) at the end of the fattening period (7.39 mg.kg(-1).min(-1)) than at baseline (21.21 mg.kg(-1).min(-1)). In stable obese dogs, scFOS increased (P < 0.05) the rate of glucose infusion compared with control (7.77 vs. 4.72 mg.kg(-1).min(-1)). Plasma insulin and triglyceride concentrations were greater in obese than in lean dogs but were not altered by scFOS. Whereas mRNA was not affected in food-deprived dogs, scFOS increased uncoupling protein 2 (P = 0.05) and tended to increase carnitine palmitoyl transferase 1 adipose mRNA levels during the postprandial period (P = 0.09). Adding 1% scFOS to the diet of obese dogs decreases insulin resistance and appears to modulate the transcription of genes involved in fatty acid or glucose metabolism.

    Topics: Adipose Tissue; Animals; Blood Glucose; Body Weight; Cholesterol; Cross-Over Studies; Disease Models, Animal; Dogs; Female; Gene Expression Regulation; Glucose Clamp Technique; Insulin; Insulin Resistance; Lipid Metabolism; Male; Obesity; Oligosaccharides; RNA, Messenger; Triglycerides

2008

Other Studies

26 other study(ies) available for fructooligosaccharide and Disease-Models--Animal

ArticleYear
Butyl-fructooligosaccharides modulate gut microbiota in healthy mice and ameliorate ulcerative colitis in a DSS-induced model.
    Food & function, 2022, Feb-21, Volume: 13, Issue:4

    Topics: Animals; Colitis, Ulcerative; Dextran Sulfate; Disease Models, Animal; Gastrointestinal Microbiome; Mice; Oligosaccharides

2022
Fructooligosaccharide supplementation alleviated the pathological immune response and prevented the impairment of intestinal barrier in DSS-induced acute colitis mice.
    Food & function, 2021, Oct-19, Volume: 12, Issue:20

    The dysbiosis of gut microbiota is closely related to the occurrence and development of inflammatory bowel disease (IBD). The manipulation of intestinal flora through prebiotics or probiotics is expected to induce and maintain the remission of IBD symptoms. 6-week-old C57BL/J mice were daily gavaged with fructooligosaccharides (FOS) or the synbiotic two weeks before the administration of dextran sulfate sodium (DSS). The supplementation of FOS or synbiotic could significantly ameliorate the body weight loss and colon histological damage in DSS-induced acute colitis mice. The altered composition of gut microbiota in acute colitis mice was reversed by FOS or Synbiotic supplementation, with a characteristic of decreased abundance of

    Topics: Animals; Colitis; Colon; Cytokines; Dextran Sulfate; Disease Models, Animal; Dysbiosis; Gastrointestinal Microbiome; Immunity; Inflammatory Bowel Diseases; Interleukin-10; Intestinal Mucosa; Lactobacillus; Male; Mice; Mice, Inbred C57BL; Oligosaccharides; Prebiotics; Probiotics; Synbiotics; Tight Junction Proteins

2021
Prophylactic and therapeutic supplementation using fructo-oligosaccharide improves the intestinal homeostasis after mucositis induced by 5- fluorouracil.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2021, Volume: 133

    The beneficial effects of prebiotic, such as fructo-oligosaccharides (FOS), in intestinal inflammation have been demonstrated in several studies. Herein, we evaluate whether joint treatment with FOS, both before and during mucositis, had additional beneficial effects and investigated the mechanisms underlying in the action of FOS on the intestinal barrier. BALB/c mice were randomly divided into five groups: CTR (without mucositis + saline solution), FOS (without mucositis + 6 % FOS), MUC (mucositis + saline solution), PT (mucositis + 6 % FOS supplementation before disease induction), and TT (mucositis + 6 % FOS supplementation before and during disease induction). Mucositis was induced by intraperitoneal injection (300 mg/kg) of 5-fluorouracil (5-FU). After 72 h, the animals were euthanized and intestinal permeability (IP), tight junction, bacterial translocation (BT), histology and morphometry, and immunoglobulin A secretory (sIgA), inflammatory infiltrate, and production of short-chain fatty acids (acetate, butyrate and propionate) were evaluated. The MUC group showed an increase in the IP, BT, and inflammatory infiltrate but a decrease in the tight junction expression and butyrate and propionate levels (P < 0.05). In the PT and TT groups, FOS supplementation maintained the IP, tight junction expression, and propionate concentration within physiologic levels, increased butyrate levels, and reduced BT and inflammatory infiltrate (P < 0.05). Total treatment with FOS (TT group) was more effective in maintaining histological score, morphometric parameters, and sIgA production. Thus, total treatment (prophylactic and therapeutic supplementation) with FOS was more effective than pretreatment alone, in reducing 5-FU-induced damage to the intestinal barrier.

    Topics: Acetates; Animals; Bacteria; Bacterial Translocation; Butyrates; Disease Models, Animal; Fatty Acids, Volatile; Fluorouracil; Gastrointestinal Microbiome; Ileum; Immunoglobulin A, Secretory; Inflammation Mediators; Intestinal Mucosa; Male; Mice, Inbred BALB C; Mucositis; Oligosaccharides; Permeability; Prebiotics; Propionates; Tight Junctions

2021
Oligosaccharides from Polygonatum Cyrtonema Hua: Structural characterization and treatment of LPS-induced peritonitis in mice.
    Carbohydrate polymers, 2021, Mar-01, Volume: 255

    Fructooligosaccharide was isolated from Polygonatum Cyrtonema Hua (PFOS) for the first time. Structure characterized using FT-IR, MALDI-TOF-MS, NMR, AFM, and TEM, indicated that PFOS was graminan-type fructan with a degree of polymerization ranging from 5 to 10. A murine model of lipopolysaccharide (LPS)-induced peritonitis was used to evaluate the in vivo anti-inflammatory and lung protective efficacy of PFOS. The result shown that pretreatment with PFOS (1.0 mg/mL) in peritonitis-induced mice could significantly inhibit the level of pro-inflammatory cytokines (TNF-α, IL-1β) in serum (P < 0.001), increase mice survival rate from 12.5 % to 54 % (P < 0.05), and alleviated lung injury through ameliorating the damage of the pulmonary cellular architecture and reducing inflammatory monocyte accumulation in lung tissue. This effect of oligosaccharides could explain the traditional usage of P. cyrtonema as a tonic medicine for respiratory problems and it could be used as a potential natural ingredient with anti-inflammatory activity.

    Topics: Acute Lung Injury; Animals; Anti-Inflammatory Agents; Cell Movement; Disease Models, Animal; Drugs, Chinese Herbal; Gene Expression; Humans; Interleukin-1beta; Lipopolysaccharides; Lung; Male; Mice; Mice, Inbred BALB C; Monocytes; Oligosaccharides; Peritonitis; Polygonatum; Survival Analysis; Tumor Necrosis Factor-alpha

2021
Effects of High Intakes of Fructose and Galactose, with or without Added Fructooligosaccharides, on Metabolic Factors, Inflammation, and Gut Integrity in a Rat Model.
    Molecular nutrition & food research, 2021, Volume: 65, Issue:6

    A high fructose and galactose intake show adverse metabolic effects in animal models and in humans, but it is yet unknown if addition of fermentable dietary fiber can mitigate such effects. This study investigate the effects of high intakes of fructose and galactose, with/without added fructooligosaccharides (FOS), on metabolic factors, inflammation, and gut integrity markers in rats.. Rats (n = 6/group) receive different carbohydrates at isocaloric conditions for 12 weeks as follows: 1) starch (control), 2) fructose, 3) galactose, 4) starch + FOS (FOS control), 5) fructose + FOS, and 6) galactose + FOS, together with a high amount of n-6 polyunsaturated fatty acids (n-6 PUFA) in all diets except for in 7) starch + olive oil (negative control). The rats fed the galactose and galactose + FOS diets exhibit lower body weight than other groups. High-galactose diets has more pronounced effects on metabolic factors and gut permeability than high-fructose diets. High-fructose diets show less pronounced effect on these selected markers. No differences in inflammatory markers are detected for any of the diets.. The results suggest potential adverse effects of high galactose and fructose on metabolic factors and gut integrity markers, but not on inflammation. However, several mechanisms are at play, and general net effects are difficult to determine conclusively for the conditions tested.

    Topics: Animals; Biomarkers; Blood Glucose; Body Weight; Disease Models, Animal; Energy Intake; Fructose; Galactose; Glycation End Products, Advanced; Inflammation; Insulin; Intestines; Lipids; Male; Oligosaccharides; Permeability; Rats, Sprague-Dawley

2021
Fructooligosaccharides protect against OVA-induced food allergy in mice by regulating the Th17/Treg cell balance using tryptophan metabolites.
    Food & function, 2021, Apr-07, Volume: 12, Issue:7

    Fructooligosaccharides (FOS) can change gut microbiota composition and play a protective role in food allergy (FA). Furthermore, the protective mechanism of FOS against FA is unclear. In this study, intestinal flora and tryptophan (Trp) metabolites were investigated in a mouse model with FA supplemented with FOS. Meanwhile, we injected aryl hydrocarbon receptor antagonists (AhR-A) into a mouse model of FA supplemented with FOS to investigate whether T helper cell (Th) 17/regulatory T (Treg) cell balance was affected. Our research studies showed that dietary intake of FOS provided moderate protection from the intestinal inflammation induced by ovalbumin (OVA). This protective effect disappeared in AhR-A mice. The OVA mice manifestations had significantly lower bacterial richness, when compared to the normal control (NC) mice. Among fecal bacteria, the abundance of Akkermansiaceae (family level) and Verrucomicrobia (phylum level) increased and Ruminococcacere (phylum level) decreased in the feces of allergic mice. These changes were reversed by FOS treatment. FOS modulated the gut microbiome profiles that were altered in OVA mice, which showed an increase in the abundance of Ruminococcacere (phylum level) and a decrease in the abundance of Akkermansiaceae (family level) and Verrucomicrobia (phylum level). Liquid chromatography/tandem mass spectrometry (LC-MS/MS) analysis of Trp metabolites showed significant reductions in the level of kynurenine (kyn) in the serum of OVA mice, as compared to NC and FOS mice. Conversely, the levels of Trp and 5-hydroxytryptamine (5-HT) were significantly increased in OVA mice. Correlation analysis revealed a negative relationship between the relative abundance of Verrucomicrobiae (class level) and Akkermansiaceae (family level) with kyn, and a positive relationship with 5-HT. FOS significantly reduced interleukin-17A (IL-17A) and retinoic acid-associated nuclear orphan receptor-γt (RORγt) in FOS mice but not in AhR-A mice. FOS increased the level of interleukin-10 (IL-10) and Forkhead box P3 (Foxp3) in FOS mice but not in AhR-A mice. These findings suggest that FOS ameliorates allergic symptoms and impacts Th17/Treg balance in mice by modulating the gut microbiota composition and Trp metabolites. FOS may serve as an effective tool for the treatment of FA by regulating immune and gut microbiota.

    Topics: Animals; Disease Models, Animal; Food Hypersensitivity; Male; Mice; Mice, Inbred BALB C; Oligosaccharides; Ovalbumin; T-Lymphocytes, Regulatory; Th17 Cells; Tryptophan

2021
The Administration of the Synbiotic
    Toxins, 2021, 03-07, Volume: 13, Issue:3

    Indoxyl sulfate (IS) is involved in the progression of chronic kidney disease (CKD) and in its cardiovascular complications. One of the approaches proposed to decrease IS is the administration of synbiotics. This work aimed to search for a probiotic strain capable to decrease serum IS levels and mix it with two prebiotics (inulin and fructooligosaccharide (FOS)) to produce a putative synbiotic and test it in a rat CKD model. Two groups of Sprague-Dawley rats were nephrectomized. One group (Lac) received the mixture for 16 weeks in drinking water and the other no (Nef). A control group (C) included sham-nephrectomized rats. Serum creatinine and IS concentrations were measured using high-performance liquid chromatography with diode array detector (HPLC-DAD). Optical microscopy and two-photon excitation microscopy was used to study kidney and heart samples. The Lac group, which received the synbiotic, reduced IS by 0.8% while the Nef group increased it by 38.8%. Histological analysis of kidneys showed that the Lac group increased fibrotic areas by 12% and the Nef group did it by 25%. The synbiotic did not reduce cardiac fibrosis. Therefore, the putative synbiotic showed that function reducing IS and the progression of CKD in a rat model, but no heart protection was observed.

    Topics: Animals; Creatinine; Disease Models, Animal; Disease Progression; Female; Fibrosis; Heart Diseases; Indican; Inulin; Kidney; Lactobacillus delbrueckii; Myocardium; Oligosaccharides; Rats, Sprague-Dawley; Renal Insufficiency, Chronic; Synbiotics; Toxins, Biological

2021
Yacón (Smallanthus sonchifolius) prevented inflammation, oxidative stress, and intestinal alterations in an animal model of colorectal carcinogenesis.
    Journal of the science of food and agriculture, 2020, Volume: 100, Issue:15

    Yacón (Smallanthus sonchifolius) roots store carbohydrate in the form of prebiotic fructooligosaccharides (FOS), which improve intestinal health. Yacon has the potential to prevent the intestinal barrier alterations associated with colorectal cancer (CRC). This study aimed to investigate the preventive effects of yacón flour (YF) on alterations promoted by CRC induced by 1,2-dimethylhydrazine in rats.. YF as a source of fructooligosaccharides may help to maintain the integrity of intestinal health, which is altered in induced CRC in rats. © 2020 Society of Chemical Industry.

    Topics: Animals; Asteraceae; Carcinogenesis; Colorectal Neoplasms; Disease Models, Animal; Fatty Acids, Volatile; Humans; Interleukin-10; Intestines; Male; Oligosaccharides; Oxidative Stress; Plant Extracts; Plant Roots; Prebiotics; Rats; Rats, Wistar; Tumor Necrosis Factor-alpha

2020
RETRACTED: Dietary oligosaccharides attenuate DSS-induced colitis in mice, induce PGlyRP3 expression, and inhibit NF-κB and MEK/ERK signaling.
    Cellular immunology, 2020, Volume: 354

    This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal).\ \ This article has been retracted at the request of the Editors.\ \ The Editors of Cellular Immunology have been informed by Elsevier that the article had been submitted to another journal while under consideration at "Cellular Immunology", which is a case of double submission.\ \ Based on the above infringement and its deleterious impact on the mutual trust necessary for the evaluation of scientific work - the corresponding authors had stated that the article was not submitted to another journal - it was decided to retract this article.

    Topics: Animals; Anti-Inflammatory Agents; Caco-2 Cells; Carrier Proteins; Colitis; Colitis, Ulcerative; Dextran Sulfate; Diet; Disease Models, Animal; Humans; Male; MAP Kinase Signaling System; Mice; Mice, Inbred C57BL; NF-kappa B; Oligosaccharides; PPAR gamma; Up-Regulation

2020
A prebiotic, short-chain fructo-oligosaccharides promotes peak bone mass and maintains bone mass in ovariectomized rats by an osteogenic mechanism.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2020, Volume: 129

    In preclinical studies, fructooligosaccharide (FOS) showed beneficial skeletal effects but its effect on peak bone mass (PBM) and bone loss caused by estrogen (E2) deficiency has not been studied, and we set out to study these effects in rats. Short-chain (sc)-FOS had no effect on body weight, body composition, and energy metabolism of ovary intact (sham) and ovariectomized (OVX) rats. scFOS did not affect serum and urinary calcium and phosphorus levels, and on calcium absorption, although an increasing trend was noted in the sham group. Sham and OVX rats given scFOS had better skeletal parameters than their respective controls. scFOS treatment resulted in a higher bone anabolic response but had no effect on the catabolic parameters. scFOS increased serum levels of a short-chain fatty acid, butyrate which is known to have osteogenic effect. Our study for the first time demonstrates that in rats scFOS at the human equivalent dose enhances PBM and protects against E2 deficiency-induced bone loss by selective enhancement of new bone formation, and implicates butyrate in this process.

    Topics: Animals; Biomarkers; Bone and Bones; Bone Remodeling; Butyrates; Disease Models, Animal; Female; Gastrointestinal Microbiome; Humans; Oligosaccharides; Osteogenesis; Osteoporosis, Postmenopausal; Ovariectomy; Prebiotics; Rats, Sprague-Dawley

2020
Dietary Fructooligosaccharide and Glucomannan Alter Gut Microbiota and Improve Bone Metabolism in Senescence-Accelerated Mouse.
    Journal of agricultural and food chemistry, 2019, Jan-23, Volume: 67, Issue:3

    Gut microbiota improved using prebiotics may delay the onset of senescence-related health problems. We hypothesized that prolonged intake of prebiotics delays senile osteoporosis. Forty-five male senescence-accelerated mouse prone 6 (SAMP6) aged four weeks were raised on 5% fructooligosaccharide (FOS), 5% glucomannan (GM), or a control diet for 31 weeks. Gut microbiota were identified using culture-dependent analytical methods. Mineral content in femoral bone was analyzed using atomic absorption spectrophotometry. Bone metabolism and inflammatory markers were measured using enzyme-linked immunosorbent assay. The numbers of Lactobacillus and Bacteroides in cecal contents were significantly higher in the FOS than in the control group ( p < 0.05); the number of Clostridium was significantly higher in the GM than in the control group ( p < 0.05). Calcium content was significantly higher in the femoral bones of the FOS group (30.5 ± 0.8 mg) than in the control group (27.5 ± 1.5 mg) ( p < 0.05). There was no difference between the GM (29.1 ± 2.0 mg) and control groups. During senescence, urinary deoxypyridinoline and serum high-sensitivity C-reactive protein levels significantly decreased in the FOS (1.2 ± 0.2 nmol/3 d and 80 ± 6.1 ng/100 mL) and GM groups (1.2 ± 0.2 nmol/3 d and 80 ± 6.1 ng/100 mL) compared with the control group (1.8 ± 0.5 nmol/3 d and 93 ± 7.4 ng/100 mL) ( p < 0.05). Thus, dietary FOS and GM modified gut microbiota and reduced bone resorption by reducing systemic inflammation in SAMP6.

    Topics: Aging; Animals; Bacteroides; Bone and Bones; C-Reactive Protein; Disease Models, Animal; Gastrointestinal Microbiome; Humans; Lactobacillus; Male; Mannans; Mice; Oligosaccharides; Osteoporosis; Prebiotics

2019
Fructooligosaccharides Ameliorating Cognitive Deficits and Neurodegeneration in APP/PS1 Transgenic Mice through Modulating Gut Microbiota.
    Journal of agricultural and food chemistry, 2019, Mar-13, Volume: 67, Issue:10

    Alzheimer's disease (AD) is closely related to gut microbial alteration. Prebiotic fructooligosaccharides (FOS) play major roles by regulating gut microbiota. The present study aimed to explore the effect and mechanism of FOS protection against AD via regulating gut microbiota. Male Apse/PSEN 1dE9 (APP/PS1) transgenic (Tg) mice were administrated with FOS for 6 weeks. Cognitive deficits and amyloid deposition were evaluated. The levels of synaptic plasticity markers including postsynaptic density protein 95 (PSD-95) and synapsin I, as well as phosphorylation of c-Jun N-terminal kinase (JNK), were determined. The intestinal microbial constituent was detected by 16S rRNA sequencing. Moreover, the levels of glucagon-like peptide-1 (GLP-1) in the gut and GLP-1 receptor (GLP-1R) in the brain were measured. The results indicated that FOS treatment ameliorated cognitive deficits and pathological changes in the Tg mice. FOS significantly upregulated the expression levels of synapsin I and PSD-95, as well as decreased phosphorylated level of JNK. The sequencing results showed that FOS reversed the altered microbial composition. Furthermore, FOS increased the level of GLP-1 and decreased the level of GLP-1R in the Tg mice. These findings indicated that FOS exerted beneficial effects against AD via regulating the gut microbiota-GLP-1/GLP-1R pathway.

    Topics: Alzheimer Disease; Amyloid beta-Peptides; Animals; Brain; Cognition; Disease Models, Animal; Disks Large Homolog 4 Protein; Gastrointestinal Microbiome; Glucagon-Like Peptide 1; Humans; Male; Mice; Mice, Transgenic; Neuronal Plasticity; Oligosaccharides; Prebiotics; Presenilin-1; Synapsins

2019
A synbiotic composed of Lactobacillus fermentum CECT5716 and FOS prevents the development of fatty acid liver and glycemic alterations in rats fed a high fructose diet associated with changes in the microbiota.
    Molecular nutrition & food research, 2017, Volume: 61, Issue:8

    We investigated the effect of a high fructose diet (HFD) on Sprague Dawley rats and the impact of a synbiotic composed of Lactobacillus fermentum CECT5716 and fructooligosaccharides. Feeding the HFD for 5 weeks resulted in liver steatosis and insulin resistance but not obesity. These changes were associated with increased production of short-chain fatty acids and increased Bacteroidetes in feces, with an augmented Bacteroidetes/Firmicutes ratio, among other changes in the microbiota. In addition, barrier function was weakened, with increased LPS plasma levels. These data are consistent with increased fructose availability in the distal gut due to saturation of absorptive mechanisms, leading to dysbiosis, endotoxemia, hepatic steatosis, and insulin resistance. Treatment with the synbiotic prevented some of the pathological effects, so that treated rats did not develop steatosis or systemic inflammation, while dysbiosis and barrier function were greatly ameliorated. In addition, the synbiotic had hypolipidemic effects. The synbiotic composed by L. fermentum CECT5716 and fructooligosaccharides has beneficial effects in a model of metabolic syndrome induced by a HFD, suggesting it might be clinically useful in this type of condition, particularly considering that high fructose intake has been related to metabolic syndrome in humans.

    Topics: Animals; Diet; Disease Models, Animal; Fatty Acid-Binding Proteins; Fatty Acids, Volatile; Fructose; Gastrointestinal Microbiome; Insulin Resistance; Limosilactobacillus fermentum; Male; Metabolic Syndrome; Non-alcoholic Fatty Liver Disease; Oligosaccharides; Rats, Sprague-Dawley; Receptors, Adiponectin; Synbiotics

2017
Restoration of cefixime-induced gut microbiota changes by Lactobacillus cocktails and fructooligosaccharides in a mouse model.
    Microbiological research, 2017, Volume: 200

    Probiotics have been used to rebuild the antibiotic-induced dysfunction in gut microbiota, but whether the different strains of probiotics result in similar or reverse effects remains unclear. In this study, the different recovery effects of two cocktails (each contains four strains) of Lactobacillus and fructooligosaccharide against cefixime-induced change of gut microbiota were evaluated in C57BL/6J mice. The results show that the use of cefixime caused a reduction in the diversities of the microbial community and led to significantly decreasing to one preponderant Firmicutes phylum, which was difficult to restore naturally in the short term. The gut microbiota compositions of the groups treated with the probiotic cocktails were much more diverse than those of the natural recovery group. The effects of Lactobacillus cocktails against the cefixime-induced gut microbiota change may mainly be due to the beneficial SCFAs production in vivo and also be related to the good cell adhesion properties performed in vitro. Meanwhile, the restoration of the cefixime-induced gut microbiota was significantly different between two Lactobacillus groups since the Lactobacillus strains with high levels of fructooligosaccharide use and better cell adhesion properties performed considerably better than the Lactobacillus strains with high survival rates in the gastrointestinal tract. The contents of short-chain fatty acids in ceca were increased to 26.483±1.925 and 25.609±2.782μmol/g in the two probiotic cocktail groups respectively compared to 15.791±0.833μmol/g (P<0.05) in control group. Moreover, intestinal inflammation was alleviated by administration of the Lactobacillus cocktails. However, fructooligasaccharide administration showed certain effects on gut microbiota restoration (such as an increase of Akkermansia), although its effect on the entire microbiome structure is not so obvious.

    Topics: Animals; Blood Chemical Analysis; Cecum; Cefixime; Cell Adhesion; Colon; Disease Models, Animal; DNA, Bacterial; Fatty Acids, Volatile; Feces; Gastrointestinal Microbiome; Gastrointestinal Tract; HT29 Cells; Humans; Ileum; Intestines; Lactobacillus; Male; Metagenomics; Mice; Mice, Inbred C57BL; Microbial Consortia; Oligosaccharides; Probiotics; RNA, Ribosomal, 16S

2017
Fructo-oligosaccharides and intestinal barrier function in a methionine-choline-deficient mouse model of nonalcoholic steatohepatitis.
    PloS one, 2017, Volume: 12, Issue:6

    Impairments in intestinal barrier function, epithelial mucins, and tight junction proteins have been reported to be associated with nonalcoholic steatohepatitis. Prebiotic fructo-oligosaccharides restore balance in the gastrointestinal microbiome. This study was conducted to determine the effects of dietary fructo-oligosaccharides on intestinal barrier function and steatohepatitis in methionine-choline-deficient mice. Three groups of 12-week-old male C57BL/6J mice were studied for 3 weeks; specifically, mice were fed a methionine-choline-deficient diet, a methionine-choline-deficient diet plus 5% fructo-oligosaccharides in water, or a normal control diet. Fecal bacteria, short-chain fatty acids, and immunoglobulin A (IgA) levels were investigated. Histological and immunohistochemical examinations were performed using mice livers for CD14 and Toll-like receptor-4 (TLR4) expression and intestinal tissue samples for IgA and zonula occludens-1 expression in epithelial tight junctions. The methionine-choline-deficient mice administered 5% fructo-oligosaccharides maintained a normal gastrointestinal microbiome, whereas methionine-choline-deficient mice without prebiotic supplementation displayed increases in Clostridium cluster XI and subcluster XIVa populations and a reduction in Lactobacillales spp. counts. Methionine-choline-deficient mice given 5% fructo-oligosaccharides exhibited significantly decreased hepatic steatosis (p = 0.003), decreased liver inflammation (p = 0.005), a decreased proportion of CD14-positive Kupffer cells (p = 0.01), decreased expression of TLR4 (p = 0.04), and increases in fecal short-chain fatty acid and IgA concentrations (p < 0.04) compared with the findings in methionine-choline-deficient mice that were not administered this prebiotic. This study illustrated that in the methionine-choline-deficient mouse model, dietary fructo-oligosaccharides can restore normal gastrointestinal microflora and normal intestinal epithelial barrier function, and decrease steatohepatitis. The findings support the role of prebiotics, such as fructo-oligosaccharides, in maintaining a normal gastrointestinal microbiome; they also support the need for further studies on preventing or treating nonalcoholic steatohepatitis using dietary fructo-oligosaccharides.

    Topics: Animals; Choline Deficiency; Dietary Supplements; Disease Models, Animal; Intestines; Male; Methionine; Mice; Mice, Inbred C57BL; Non-alcoholic Fatty Liver Disease; Oligosaccharides; Prebiotics

2017
Fructo-oligosaccharide improved brain β-amyloid, β-secretase, cognitive function, and plasma antioxidant levels in D-galactose-treated Balb/cJ mice.
    Nutritional neuroscience, 2017, Volume: 20, Issue:4

    Long-term d-galactose injection induces accelerated aging in experimental rodent models. The aim of this study was to determine the effects of dietary fructo-oligosaccharide (FO) on the brain β-amyloid (Aβ), amyloid-associated enzymes, cognitive function, and plasma antioxidant levels in d-galactose-treated Balb/c mice.. The subcutaneous (s.c.) injection and the dietary treatment were conducted simultaneously for 49 days. Mice (12 weeks of age) were divided into five groups (n = 14/group): control (s.c. saline, control diet) serving as a young control, DG (s.c. 1.2 g d-galactose/kg body weight, control diet), DG + LFO (2.5% w/w FO, low-dose FO diet), DG + HFO (5% w/w FO, high-dose FO diet), and DG + E (α-tocopherol 0.2% w/w, vitamin E diet) as an antioxidant reference group. Another group of older mice (64 weeks of age) without any injection served as a natural aging (NA) group.. The DG and NA groups had greater Aβ levels in the cortex, hippocampus, and the whole brain. High-dose FO, similar to α-tocopherol, attenuated the d-galactose-induced Aβ density in the cortex and hippocampus. In addition, FO attenuated the d-galactose-induced protein expression of Aβ and beta-site amyloid precursor cleaving enzyme of the whole brain in a dose-response manner. Either dose of FO supplementation, similar to α-tocopherol, attenuated the d-galactose-induced cognitive dysfunction. In addition, FO improved the plasma ascorbic acid level in a dose-response manner.. Dietary FO (2.5-5% w/w diet) could attenuate the development of Alzheimer's disease, which was likely to be associated with its systematic antioxidant effects.

    Topics: alpha-Tocopherol; Amyloid beta-Peptides; Amyloid Precursor Protein Secretases; Animals; Antioxidants; Ascorbic Acid; Brain; Cognition; Cognition Disorders; Disease Models, Animal; Dose-Response Relationship, Drug; Galactose; Male; Mice; Mice, Inbred BALB C; Oligosaccharides

2017
Fructooligosaccharides exert intestinal anti-inflammatory activity in the CD4+ CD62L+ T cell transfer model of colitis in C57BL/6J mice.
    European journal of nutrition, 2016, Volume: 55, Issue:4

    Fructooligosaccharides (FOS) are used as functional foods due to their prebiotic effects. Intestinal anti-inflammatory activity has been established in most, but not all, studies in animal models of colitis, using mainly chemically induced inflammation. Our goal was to test the effect of FOS (degree of polymerization 2-8) in the chronic, lymphocyte-driven CD4+ CD62L+ T cell transfer model of colitis.. Colitis was induced by transfer of CD4+ CD62L+ T cells to C57BL/6J Rag1(-/-) mice. FOS (75 mg day(-1)) was administered by gavage as a post-treatment. Three groups were established: non-colitic (NC), colitic control (C, CD4+ CD62L+ transferred mice treated with vehicle) and colitic+FOS (C+FOS, similar but treated with FOS). Mice were killed after 13 days.. Treatment of mice with FOS ameliorated colitis, as evidenced by an increase in body weight, a lesser myeloperoxidase and alkaline phosphatase activities, a lower secretion of proinflammatory cytokines by mesenteric lymph node cells ex vivo (IFN-γ, IL-17, and TNF-α), and a higher colonic expression of occludin (C+FOS vs. C, p < 0.05). Increased relative abundance of lactic acid bacteria was observed in FOS-treated mice (p < 0.05).. FOS exert intestinal anti-inflammatory activity in T lymphocyte-dependent colitis, suggesting it may be useful in the management of inflammatory bowel disease in appropriate conditions.

    Topics: Alkaline Phosphatase; Animals; Anti-Inflammatory Agents; Calgranulin A; CD4-Positive T-Lymphocytes; Claudin-4; Claudin-5; Colitis; Disease Models, Animal; Dose-Response Relationship, Drug; Female; Gastrointestinal Microbiome; Gene Expression Regulation; Interferon-gamma; Interleukin-10; Interleukin-17; Interleukin-1beta; Intestinal Mucosa; Intestines; L-Selectin; Lactobacillus; Mice; Mice, Inbred C57BL; Occludin; Oligosaccharides; Peroxidase; Tumor Necrosis Factor-alpha

2016
Effect of fructooligosaccharides fraction from Psacalium decompositum on inflammation and dyslipidemia in rats with fructose-induced obesity.
    Nutrients, 2014, Jan-29, Volume: 6, Issue:2

    Psacalium decompositum, commonly known as "Matarique," is a medicinal plant used in Mexico for diabetes mellitus empirical therapy. Previous studies have shown that the fructooligosaccharides (FOS) present in the roots of this plant exhibit a notable hypoglycemic effect in animal models; this effect might be associated with the attenuation of the inflammatory process and other metabolic disorders. In this study, we examined the effects of FOS fraction administration in a fructose-fed rat model for obesity. Phytochemical chromatographic studies (high performance thin layer chromatography and nuclear magnetic resonance) were performed to verify isolation of FOS. 24 male Wistar rats were maintained for 12 weeks on a diet of 20% HFCS in drinking water and chow. Glucose, cholesterol, triglycerides and liver transaminases levels were measured monthly, after administering FOS fraction intragastrically (150 mg/kg/day for 12 weeks), while the levels of inflammatory cytokines were only quantified at the end of the treatments. Rats treated with FOS fraction decreased body weight, cholesterol, triglycerides, and significantly reduced IL-6, IFN-γ, MCP-1, IL-1β and VEGF levels (p < 0.05). These results suggest that P. decompositum has anti-inflammatory and hypolipidemic properties that might be used as an alternative treatment for the control of obesity.

    Topics: Animals; Anti-Inflammatory Agents; Asteraceae; Body Weight; Chemokine CCL2; Cholesterol; Disease Models, Animal; Dyslipidemias; Fructose; Hypoglycemic Agents; Inflammation; Interferon-gamma; Interleukin-1beta; Interleukin-6; Male; Obesity; Oligosaccharides; Plant Extracts; Plant Roots; Plants, Medicinal; Rats; Rats, Wistar; Triglycerides; Vascular Endothelial Growth Factor A

2014
Di-D-fructose dianhydride-enriched caramels: effect on colon microbiota, inflammation, and tissue damage in trinitrobenzenesulfonic acid-induced colitic rats.
    Journal of agricultural and food chemistry, 2010, May-26, Volume: 58, Issue:10

    In the present study we describe the preparation and chemical characterization of a caramel with a high (70%) content of difructose dianhydrides (DFAs) and glycosylated derivatives (DFAs). This product was obtained by thermal activation (90 degrees C) of highly concentrated (90% w/v) aqueous D-fructose solutions using the sulfonic acid ion-exchange resin Lewatit S2328 as caramelization catalyst. DFAs represent a unique family of cyclic fructans with prebiotic properties already present in low proportions (<15%) in commercial caramel. We report the antiinflammatory activity of the new DFA-enriched caramel in the trinitrobenzenesulfonic acid (TNBS) model of rat colitis, an experimental model that resembles human inflammatory bowel disease (IBD), and compare its effects with those obtained with a commercial sucrose caramel and with linear fructooligosaccharides (FOS). For this purpose, the effects on colon tissue damage, gut microbiota, short-chain fatty acid (SCFAs) production, and different inflammatory markers were evaluated. The administration of DFA-enriched caramel to colitic rats showed intestinal antiinflammatory effect, as evidenced macroscopically by a significant reduction in the extent of the colonic damage induced by TNBS. This effect was similar to that obtained with FOS in the same experimental settings, whereas commercial caramel was devoid of any significant antiinflammatory effect. The beneficial effect was associated with the inhibition of the colonic levels of the proinflammatory cytokines, tumor necrosis factor alpha (TNF alpha) and interleukin 1beta (IL-1beta), and the reduction in colonic myeloperoxidase (MPO) activity and inducible nitric oxide synthase (iNOS) expression. The DFA-enriched caramel also promoted a more favorable intestinal microbiota, increasing lactobacilli and bifidobacteria counts as well as inducing higher concentrations of SCFAs in the luminal colonic contents. These results reinforce the concept of DFAs and glycosyl-DFAs as dietary beneficial compounds with prebiotic properties and suggest that the novel DFA-enriched caramel here reported may be an interesting candidate to be explored for the dietary treatment of human IBD.

    Topics: Animals; Anti-Inflammatory Agents; Candy; Colitis; Colon; Disaccharides; Disease Models, Animal; Female; Food Handling; Food, Fortified; Glycosylation; Hot Temperature; Oligosaccharides; Prebiotics; Rats; Rats, Wistar; Stereoisomerism; Sucrose; Trinitrobenzenesulfonic Acid

2010
The combination of fructooligosaccharides and resistant starch shows prebiotic additive effects in rats.
    Clinical nutrition (Edinburgh, Scotland), 2010, Volume: 29, Issue:6

    Different types of dietary fiber can be distinguished considering their rate of fermentability, thus determining the location of the large intestine where they exert their beneficial effect. Their combination could be interesting to obtain health-promoting effects throughout the entire colon. The aim of the present study was to evaluate the synergistic effect of two dietary fibers with different fermentation patterns, fructooligosaccharides (FOS) (Beneo(®)-95) and resistant starch (Fibersol(®)-2), after their administration to healthy rats or in trinitrobenzenesulphonic acid-(TNBS) colitic rats, with an altered colonic immune response. In healthy rats, the administration of the combination of FOS and resistant starch induced changes in the intestinal microbiota, by increasing lactobacilli and bifidobacteria in caecum and colonic contents. Furthermore, its administration up-regulated the expression of the trefoil factor-3 and MUC-2 in comparison with untreated rats, thus improving the intestinal barrier function. The beneficial effects observed with this combination were confirmed in the TBNS model of rat colitis, since it was able to exert intestinal anti-inflammatory effect, associated with an increase of protective bacteria and up-regulation of epithelial defense mechanisms. In conclusion, the combination of two different dietary fibers may result in a synergistic prebiotic effect, and may confer greater health benefits to the host.

    Topics: Animals; Anti-Inflammatory Agents; Colitis; Colon; Dietary Fiber; Disease Models, Animal; Female; Fermentation; Lactobacillus; Oligosaccharides; Prebiotics; Rats; Rats, Wistar; Starch; Up-Regulation

2010
Immunomodulatory impact of a synbiotic in T(h)1 and T(h)2 models of infection.
    Therapeutic advances in respiratory disease, 2010, Volume: 4, Issue:5

    The immunomodulatory activity of a synbiotic combination containing three bacterial strains (Lactobacillus helveticus R0052, Bifidobacterium longum subsp. infantis R0033 and Bifidobacterium bifidum R0071) and short-chain fructooligosaccharide was examined in two distinct infectious rat models. In the T(h)1 model, Wistar rats were administered the synbiotic combination for 2 weeks prior to challenge with a single oral dose of enterotoxigenic Escherichia coli or vehicle. In the T(h)2 model, pretreated rats were challenged with a single subcutaneous dose of hook worm, Nippostrongylus brasiliensis. Blood samples were collected 3 hours or 4 days postchallenge and serum levels of pro- and anti-inflammatory cytokines were measured.. Significant reductions in pro-inflammatory cytokines interleukin (IL)-1α, IL-1β, IL-6, and tumour necrosis factor (TNF)-α were observed in both models suggesting a single, unifying mode of action on an upstream regulator. The N. brasiliensis study also compared the effect of the individual strains to synbiotic. For most of cytokines the combination appeared to average the effect of the individual strains with the exception of IL-4 and IL-10 where there was apparent synergy for the combination. Furthermore, the cytokine response varied by strain.. It was concluded that this synbiotic combination of these three microbes could be beneficial in both T(h)1 and T(h)2 diseases.

    Topics: Animals; Bifidobacterium; Disease Models, Animal; Escherichia coli Infections; Interleukins; Lactobacillus helveticus; Male; Nippostrongylus; Oligosaccharides; Rats; Rats, Wistar; Strongylida Infections; Synbiotics; Th1 Cells; Th2 Cells; Tumor Necrosis Factor-alpha

2010
Fructo-oligosaccharide reduces inflammation in a dextran sodium sulphate mouse model of colitis.
    Digestive diseases and sciences, 2007, Volume: 52, Issue:1

    Fructo-oligosaccharide (FOS) is a prebiotic that stimulates the colonic growth of bifidobacteria to promote intestinal health. This study assessed whether FOS can reduce intestinal damage associated with ulcerative colitis and accelerate recovery in a mouse model. C57BL/6 mice received 2% dextran sulphate sodium for 7 days (days 8-14). FOS (1.5 g/mL) treatment was administered twice daily (n=10/group): before and during colitis (days 1-14); during colitis (days 10-14); and during colitis and the recovery period (days 10-19). Disease activity was scored daily and colonic damage was assessed by histological analysis. FOS treatment significantly reduced disease activity and damage in the distal colon (P < .05). Treatment with FOS (days 10-14) had increased crypt depth (116+/-6 microm) compared to water treatment (90+/-4 microm, P < .05). FOS treatment (days 10-19) produced a faster recovery from damage with increased crypt depth and crypt area. These results demonstrate the protective effect of FOS treatment.

    Topics: Animals; Body Weight; Breath Tests; Colitis; Dextran Sulfate; Disease Models, Animal; Epithelium; Female; Mice; Mice, Inbred C57BL; Oligosaccharides

2007
Increased number of CCR4-positive cells in the duodenum of ovalbumin-induced food allergy model Nc/jic mice and antiallergic activity of fructooligosaccharides.
    Allergology international : official journal of the Japanese Society of Allergology, 2007, Volume: 56, Issue:2

    Fructooligosaccharides (FOS) in prebiotic foods can alter intestinal immune responses. The combination of probiotics with oligosaccharides has been reported to alter intestinal flora and suggested to be beneficial against food allergy in humans.. All male Nc/jic mice used in this 8-week study were 6 weeks of age and were allotted to the following three groups: (1) the nonsensitization group; (2) the ovalbumin (OVA) sensitization +5% fructose-containing control food administration group; and (3) the OVA sensitization +5% FOS-containing food administration group. Duodenal tissues were collected and then immunohistochemically stained with monoclonal antibodies to CCR4 and CCR5. The number of mast cells and the villus edema formation rate in the duodenum were determined by image analysis.. The number of CCR4-positive cells increased significantly in Group 2 as compared with Group 1 and tended to decrease in Group 3 as compared with Group 2. Relatively few CCR5-positive cells were observed in the duodenum. FOS tended to reduce the number of CCR4-positive cells but significantly reduced the number of mast cells and the edema formation rate in the duodenum.. This study demonstrated a correlation between the number of CCR4-positive cells and villus edema formation rate. Therefore, FOS, which we inferred to show antiallergic activity for food allergy in this study and which has already been established to be safe for use as food in humans, can be considered to be potentially useful for the prevention of food allergy in pediatric patients with allergy.

    Topics: Animals; Anti-Allergic Agents; Cell Count; Disease Models, Animal; Duodenal Diseases; Duodenum; Edema; Food Hypersensitivity; Immunoglobulin E; Male; Mast Cells; Mice; Microvilli; Oligosaccharides; Ovalbumin; Probiotics; Receptors, CCR4; Receptors, CCR5; Receptors, Chemokine; T-Lymphocytes, Helper-Inducer

2007
Prebiotic and synbiotic fructooligosaccharide administration fails to reduce the severity of experimental colitis in rats.
    Diseases of the colon and rectum, 2007, Volume: 50, Issue:7

    Opposing effects of the prebiotic, fructooligosaccharide, have been reported in experimental colitis. We compared the effects of the prebiotic, fructooligosaccharide, alone and in synbiotic combination with Lactobacillus fermentum BR11, on the development of dextran sulfate sodium-induced colitis in rats. Rats consumed an 18 percent casein-based diet or diet supplemented with 6 percent fructooligosaccharide or maltodextrin for 14 days. The synbiotic group was gavaged 1 ml of L. fermentum BR11 (1x10(9) cfu/ml) twice daily. From Days 7 to 14, colitis was induced via 3 percent dextran sulfate sodium in drinking water. Disease activity was assessed daily, and at killing, gastrointestinal organs were measured, weighed, and examined by quantitative histology, proliferating cell nuclear antigen immunohistochemistry, and colonic myeloperoxidase activity. Administration of dextran sulfate sodium resulted in an increased colitic disease activity, and an increased colon and cecum weight compared with normal controls. Colon and cecum weights were further increased in dextran sulfate sodium+fructooligosaccharide (colon: 19 percent; cecum: 48 percent) and dextran sulfate sodium+fructooligosaccharide/L. fermentum BR11-treated rats (16 and 62 percent) compared with dextran sulfate sodium+vehicle-treatment. Dextran sulfate sodium+fructooligosaccharide-treated rats displayed an 81 percent increase in colonic myeloperoxidase activity compared with dextran sulfate sodium-treated controls. Histologic damage severity scores increased in dextran sulfate sodium+vehicle, dextran sulfate sodium+fructooligosaccharide, and dextran sulfate sodium+fructooligosaccharide/L. fermentum BR11-treated rats compared with normal controls (P<0.05). Crypt depth increased in all treatments compared with normal controls (P<0.01). No protection from dextran sulfate sodium-colitis was accorded by fructooligosaccharide alone or in synbiotic combination with L. fermentum BR11, whereas fructooligosaccharide actually increased some indicators of colonic injury.

    Topics: Administration, Oral; Animals; Cell Proliferation; Colitis; Colon; Dextran Sulfate; Disease Models, Animal; Limosilactobacillus fermentum; Male; Oligosaccharides; Peroxidase; Probiotics; Rats; Rats, Sprague-Dawley; Severity of Illness Index; Treatment Failure

2007
Fructooligosaccharides maximize bone-sparing effects of soy isoflavone-enriched diet in the ovariectomized rat.
    Calcified tissue international, 2004, Volume: 75, Issue:2

    Isoflavones (IF) have been increasingly implicated for use in the prevention of osteoporosis. As their bioavailability could be improved by modulating intestinal microflora, the present study was undertaken to investigate whether IF and fructooligosaccharides (FOS), which are known to modify large-bowel flora and metabolism, may exhibit a cooperative bone-sparing effect. This work was carried out on 3-month-old Wistar rats assigned to 12 groups: 2 SH (sham-operated) and 10 OVX (ovariectomized). Animals received a diet for 90 days containing total IF (Prevastei HC, Central Soya) at 0 (OVX and SH), 10 (IF10), 20 (IF20), 40 (IF40), or 80 (IF80) microg/g body weight per day. FOS (Actilight, Beghin-Meiji) were orally given to half of the groups, (OVX FOS), (IF10 FOS), (IF20 FOS), (IF40 FOS), (IF80 FOS), and (SH FOS). Isoflavones exhibited a bone-sparing effect as soon as consumption reached 20 microg/g/day, whereas only the highest dose induced a weak uterotrophic activity. Indeed, total femoral bone mineral density (BMD) was significantly enhanced (compared with that of OVX rats), as was the metaphyseal compartment. Bone strength was improved as well. As far as the FOS diet is concerned, addition of prebiotics significantly raised the efficiency of the IF protective effect on both femoral BMD and mechanical properties. The trend toward higher BMD levels with the lowest IF dose (IF10) even reached a significant level when FOS were added. This effect could be explained by a reduced bone resorption. In conclusion, daily IF consumption prevented castration-induced osteopenia by decreasing bone resorption when given at 20, 40, or 80 microg (total isoflavones)/g/day. Simultaneous FOS consumption improved IF protective effect on the skeleton, with the lowest IF dose becoming efficient. Enhancement of IF bioavailability, following FOS fermentation, is probably involved.

    Topics: Animals; Bone and Bones; Bone Density; Bone Development; Bone Resorption; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Synergism; Female; Femur; Food, Formulated; Humans; Isoflavones; Oligosaccharides; Osteoporosis, Postmenopausal; Ovariectomy; Rats; Rats, Wistar; Soy Foods; Treatment Outcome

2004
Fermentable carbohydrate and digestive nitrogen excretion.
    Journal of renal nutrition : the official journal of the Council on Renal Nutrition of the National Kidney Foundation, 2001, Volume: 11, Issue:3

    Interventions that restrict protein intake lower plasma urea concentration and may slow the progression of renal failure. The question arises whether the effect of a dietary protein restriction could be reinforced by enrichment of the diet with fermentable carbohydrate because these carbohydrates may stimulate the extra-renal route of nitrogen (N) excretion through the digestive route.. The influence of fermentable carbohydrate and moderate protein restriction on N metabolism was investigated in a rat model of renal failure with ablation of 70% of renal mass compared with control rats with intact kidneys. Animals were adapted to diets varying with respect to nondigestible fermentable carbohydrate (0% or 10% fructooligosaccharide [FOS]) and with respect to protein content (10% or 18% casein).. Feeding FOS led to a considerable enlargement of the cecum (increase in contents, wall thickness, and blood flow). These changes resulted in a concomitant enhancement of urea N uptake into the cecum and a decrease in plasma urea concentration (-30%). The extent of urea uptake by the cecum was influenced by plasma urea level that was determined by the dietary protein level and by the renal function. Thus, compared with control rats, the rate of urea uptake by the cecum and the total N excreted by the uremic rats was greater under all nutritional conditions. It is noteworthy that, when expressed as a percentage of total N excretion, fecal N excretion nearly doubled in rats adapted to the low-protein diets containing FOS. These effects occurred in both control rats and in uremic rats, in which a 22% decrease in urinary N was recorded as a result of FOS in addition to the low-protein diet. Globally, decreasing the amount of protein in the diet and adding a fermentable carbohydrate led to a decrease in urinary N excretion of more than 65% in uremic rats.. These results suggest a possible usefulness for combining fermentable carbohydrate, such as FOS, with a low-protein diet to increase N excretion through the digestive route in detriment of the renal route. This may represent an efficient preventive measure to relieve the renal function in case of renal failure.

    Topics: Ammonia; Animals; Cecum; Dietary Carbohydrates; Dietary Proteins; Disease Models, Animal; Disease Progression; Feces; Fermentation; Male; Nitrogen; Oligosaccharides; Rats; Rats, Wistar; Renal Insufficiency; Urea; Weight Gain

2001