Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Nutr Sci Vitaminol (Tokyo) ; 70(2): 139-149, 2024.
Article in English | MEDLINE | ID: mdl-38684384

ABSTRACT

Since propionate exerts several physiological effects, maintenance of its normal colonic fermentation is essential. To investigate whether vitamin B12 (VB12) is essential for normal propionate fermentation by colonic bacteria, via the succinate pathway, we examined if high-amylose cornstarch (HACS) feeding activated such a pathway, if high HACS feeding impaired propionate fermentation, and if oral VB12 supplementation normalized propionate fermentation. Male rats were given control, 20% HACS or 3% fucose diets (Expt. 1); a VB12-free control diet or one supplemented with 5-30% HACS (Expt. 2); and the 20% HACS diet supplemented with 0.025-25 mg/kg of VB12 (Expt. 3), for 14 d. HACS feeding significantly increased cecal succinate concentration, activating the succinate pathway (Expt. 1). Cecal cobalamin concentration in 20% and 30% HACS groups was about 75% of that in the control group (Expt. 2). Cecal succinate and propionate concentrations significantly increased and decreased in 30% HACS groups, respectively, compared with the control group. Although HACS group supplemented with 0.025 mg/kg of VB12 had a low concentration of cecal propionate, adding high amounts of VB12 to HACS diets provided sufficient amounts of VB12 to rat ceca and increased cecal propionate concentration (Expt. 3). Compared with the non-HACS group, the relative abundance of Akkermansia muciniphila, but not Bacteroides/Phocaeicola, was lower in the HACS counterpart and showed improvement with increased VB12 doses. To summarize, feeding high HACS decreased and increased cecal VB12 and succinate concentrations, respectively. Furthermore, colonic delivery of sufficient amounts of VB12 to rats likely reduced accumulation of succinate and normalized propionate fermentation.


Subject(s)
Amylose , Cecum , Colon , Dietary Supplements , Fermentation , Propionates , Starch , Vitamin B 12 , Animals , Male , Propionates/metabolism , Cecum/microbiology , Cecum/metabolism , Vitamin B 12/administration & dosage , Vitamin B 12/pharmacology , Colon/metabolism , Colon/microbiology , Starch/metabolism , Starch/administration & dosage , Amylose/administration & dosage , Amylose/metabolism , Rats , Succinic Acid/metabolism , Diet , Rats, Wistar , Rats, Sprague-Dawley
2.
J Nutr Sci Vitaminol (Tokyo) ; 68(2): 104-111, 2022.
Article in English | MEDLINE | ID: mdl-35491199

ABSTRACT

To investigate whether the oral intake of slowly digestible α-glucan (SDG) could have a trophic (i.e., thickening) effect on their ileal mucosae, for 10 d, rats were given control (non-SDG), 10% isomaltodextrin (IMD) or 10% resistant maltodextrin (RMD) diets. In addition, experimental rat groups were further divided into two groups each and their diets either had or had not 1% sodium carboxymethylcellulose (CMC) added as a thickening agent. In the jejuna and the ilea, compared with control rats, the villus length and the mucosal thickness, but not the crypt depth, were significantly greater in the RMD-fed rats, with the trophic effect being weaker in the IMD-fed rats than in the RMD-fed rats. The colonic crypt depth was significantly greater in SDG groups than in the control group. The concentration of plasma glucagon-like peptide (GLP)-2 in the portal veins of the RMD group but not the IMD group was significantly higher than in the control group, with no effect of CMC supplementation on its concentration. The concentrations of cecal short-chain fatty acids did not significantly increase with SDG supplementation except for propionate concentration of the IMD-supplemented rats, compared with those in the control rats. We concluded that SDGs, especially RMD, thickened the mucosae of the rat distal small intestines. In particular, this effect of RMD but not IMD could have resulted from increased glucose available as a secretagogue of the trophic hormone GLP-2, in the ileum.


Subject(s)
Glucagon-Like Peptide 2 , Glucans , Animals , Dextrins , Ileum , Maltose/analogs & derivatives , Polysaccharides , Rats
3.
Biosci Microbiota Food Health ; 41(2): 54-65, 2022.
Article in English | MEDLINE | ID: mdl-35433160

ABSTRACT

Although lipopolysaccharide (LPS)-binding protein (LBP) is an acute-phase protein mainly produced by hepatocytes, it has also been proposed to be a pro-inflammatory adipokine. Obesity and the consumption of a high-fat diet (HFD) are reportedly associated with elevated levels of LPS in plasma and free fatty acids (FFAs) in white adipose tissue (WAT). We examined whether circulating LPS or local FFAs are responsible for the HFD-induced increase of LBP in WAT. Male C57BL/6J mice were fed either a normal-fat diet (NFD) or an HFD. The mRNA levels in the liver and mesenteric WAT (mWAT), total FFA content in mWAT, and LBP and LPS concentrations in plasma were determined. The Lbp mRNA level in mWAT was higher in mice fed the HFD than in those fed the NFD for 3, 7, or 28 days or 14 weeks, whereas the hepatic Lbp mRNA level did not differ between the groups. The Lbp mRNA level in mWAT was also increased by the HFD in germ-free mice, which do not have gut microbiota, the source of LPS. The plasma LPS level did not show a significant correlation with the mWAT Lbp mRNA level. The total FFA content in mWAT was higher in mice fed the HFD than in those fed the NFD and positively correlated with the Lbp mRNA level. Supplementation with palmitic acid increased the Lbp mRNA level in 3T3-L1 adipocytes. We propose that local FFAs, but not circulating LPS, are the trigger for increased Lbp expression in mWAT of mice fed the HFD.

4.
Biosci Biotechnol Biochem ; 86(3): 397-406, 2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35026010

ABSTRACT

Dietary factors, affect Akkermansia muciniphila (AM) abundance in the colon, have attracted attention, driven by the inverse correlation between AM abundance and metabolic disorders. We prepared skate-skin mucin (SM), porcine stomach mucin (PM), and rat gastrointestinal mucin (RM). SM contained more sulfated sugars and threonine than PM or RM. Rats were fed a control diet or diets including SM, PM, or RM (15 g/kg), or SM (12 g/kg) from 5 different threonine contents for 14 d. Cecal total bacteria and AM were less and more numerous, respectively, in SM-fed rats than the others, but SM did not affect microbial species richness. Low-threonine SM did not induce AM proliferation. The in vitro fermentation with human feces showed that the rate of AM increase was greater with SM than PM. Collectively, heavy SM sulfation facilitates a priority supply of SM-derived amino sugars and threonine that promotes AM proliferation in rats and human feces.


Subject(s)
Mucins
5.
Br J Nutr ; 127(4): 513-525, 2022 02 28.
Article in English | MEDLINE | ID: mdl-33849681

ABSTRACT

SCFA increase serotonin (5-hydroxytryptamine, 5-HT) synthesis and content in the colon in vitro and ex vivo, but little is known in vivo. We tested whether dietary indigestible saccharides, utilised as a substrate to produce SCFA by gut microbiota, would increase colonic 5-HT content in mice. Male C57BL/6J mice were fed a purified diet and water supplemented with 4 % (w/v) 1-kestose (KES) for 2 weeks. Colonic 5-HT content and enterochromaffin (EC) cell numbers were lower in mice supplemented with KES than those without supplementation, while monoamine oxidase A activity and mRNA levels of tryptophan hydroxylase 1 (Tph1), chromogranin A (Chga), Slc6a4 and monoamine oxidase A (Maoa) genes in the colonic mucosa, serum 5-HT concentration and total 5-HT content in the colonic contents did not differ between groups. Caecal acetate concentration and Bifidobacterium pseudolongum population were higher in KES-supplemented mice. Similar trends were observed in mice supplemented with other indigestible saccharides, that is, fructo-oligosaccharides, inulin and raffinose. Intragastric administration of live B. pseudolongum (108 colony-forming units/d) for 2 weeks reduced colonic 5-HT content and EC cell numbers. These results suggest that changes in synthesis, reuptake, catabolism and overflow of 5-HT in the colonic mucosa are not involved in the reduction of colonic 5-HT content by dietary indigestible saccharides in mice. We propose that gut microbes including B. pseudolongum could contribute to the reduction of 5-HT content in the colonic mucosa via diminishing EC cells.


Subject(s)
Colon , Serotonin , Animals , Bifidobacterium , Colon/metabolism , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Monoamine Oxidase/metabolism , Serotonin/metabolism
6.
J Nutr Sci Vitaminol (Tokyo) ; 66(4): 331-338, 2020.
Article in English | MEDLINE | ID: mdl-32863306

ABSTRACT

Pectin enhances mucin secretion in the rat small intestine. However, what structural features of pectin to stimulate mucin secretion remain unclear. The study aimed to clarify active constituents of pectin using a human goblet cell line, HT29-MTX. Various pectins at 100 mg/L commonly stimulated MUC5AC secretion, irrespective of their differences in molecular size, plant origin and degree of methoxylation, whereas other dietary fiber materials at 100 mg/L did not show any effects, except fucoidan. Hairy region concentrate (HRC) and its further fractions (F1-F3) were prepared by polygalacturonase treatment of citrus pectin and successive anion exchange chromatography. Neutral sugars, such as galactose and arabinose were enriched in these fractions. HRC and F1-F3 at 30 mg/L significantly increased MUC5AC secretion, which were 3 times more potent compared with a starting material (citrus pectin). Further, a dose-dependent study showed that F1 significantly increased MUC5AC secretion from at 0.3 mg/L, much stronger than that of mucin-secretagogue lipopolysaccharides. Rats consumed 5% apple pectin diet showed significant increases of luminal mucin contents and Muc2 expression in the small intestine, while the luminal mucin contents in rats consumed 1.5% HRC diet were increased by 24% compared to those in rats consumed control diet, but the difference did not reach significant. Thus, HRC is supposed to be active constituents of mucin-secretory effect of pectin in vitro. At present, however, the effect of HRC has not been verified in vivo.


Subject(s)
Intestine, Small/metabolism , Mucin 5AC/metabolism , Mucin-2/metabolism , Pectins/chemistry , Pectins/pharmacology , Animals , Diet , Dietary Fiber/administration & dosage , Dietary Fiber/pharmacology , HT29 Cells , Humans , Intestinal Mucosa/metabolism , Male , Pectins/administration & dosage , Rats , Rats, Sprague-Dawley
7.
J Nutr ; 150(10): 2656-2665, 2020 10 12.
Article in English | MEDLINE | ID: mdl-32286621

ABSTRACT

BACKGROUND: Intestinal mucins escape digestion and enter the large bowel where they are degraded by the microbiota. To what extent and how mucins impact large-bowel physiology remain unclear. OBJECTIVE: This study examined the large-bowel fermentation characteristics of mucins and mucin-derived O-glycan sugars and whether they affect gut immunity. METHODS: Mucin secretion from the terminal ileum was determined from feces of ileorectostomized male Wistar rats (age 6 wk) fed an AIN76-based control diet (CD) for 15 d (experiment 1). Normal male Wistar rats (age 6 wk; 4 wk for experiment 4) were fed CD ± porcine stomach mucin (PM) at 6 or 12 g/kg diet, equivalent to 1.5 and 3 times the daily mucin secretion, for 14 d (experiment 2); CD ± N-acetylglucosamine (GlcNAc), fucose, or N-acetylneuraminic acid at 10 g/kg diet for 14 d (experiment 3); or CD ± PM (15 g/kg diet) or GlcNAc (10 g/kg diet) for 29 d (experiment 4). SCFAs, microbial composition, and cecal O-glycan content were assessed. IgA+ plasma cells and regulatory T cells and inflammatory cytokine expression in the cecum were evaluated (experiment 4). RESULTS: Daily mucin secretion corresponded to 43.2 µmol of O-glycans. Cecal O-glycan contents were comparable between CD- and PM-fed rats. PM-fed rats harbored more mucin-degrading bacteria. Cecal concentrations of acetate (+37%) and n-butyrate (+73%) were higher in 12-g/kg PM diet-fed rats versus CD (P < 0.05). Among O-glycan sugars, only GlcNAc produced higher n-butyrate concentrations (+68%) versus CD (P < 0.05), with increased numbers of butyrate-producing bacteria. GlcNAc increased the abundance of IgA+ plasma cells (+29%) and regulatory T cells (+33%) versus CD, whereas PM increased IgA+ plasma cells (+25%) (all P < 0.05). GlcNAc and PM decreased expression of Tnfa (-30%, -40%) and Ifng (-30%, -70%) versus CD (all P < 0.05). CONCLUSIONS: Mucin-derived O-glycans act as endogenous fiber and maintain mucosal immune homeostasis via large-bowel SCFA production in rats.


Subject(s)
Dietary Fiber/administration & dosage , Dietary Fiber/pharmacology , Fatty Acids, Volatile/metabolism , Immunity, Mucosal/drug effects , Mucins/metabolism , Polysaccharides/metabolism , Animals , Cecum/drug effects , Dietary Fiber/analysis , Feces , Fermentation , Mucins/chemistry , Polysaccharides/chemistry , Rats , Rats, Wistar
8.
Br J Nutr ; 123(6): 619-626, 2020 03 28.
Article in English | MEDLINE | ID: mdl-31813401

ABSTRACT

To investigate whether oral intake of highly branched α-glucan isomaltodextrin (IMD) could stimulate ileal glucagon-like peptide-1 (GLP-1) secretion, we examined (1) the digestibility of IMD, (2) the digestion and absorption rates of IMD, in rat small intestine and (3) portal GLP-1 concentration in rats given IMD. In Expt 1, ileorectostomised rats were given a 3 % IMD diet for 10 d. Separately, a 16-h in vitro digestion of IMD, using porcine pancreatic α-amylase and brush-border membrane vesicles from rat small intestine, was conducted. In Expt 2, upon 24-h fasting, rats were given any of glucose, IMD and high-amylose maize starch (HAMS) (1 g/kg of body weight). In Expt 3, caecectomised rats were given 0·2 % neomycin sulphate and a 5 % IMD diet for 10 d. The in vivo and in vitro digestibility of IMD was 70-80 %. The fraction of IMD digested in vitro for the first 120 min was 67 % of that in maize starch. The AUC for 0-120 min of plasma glucose concentration was significantly lower in HAMS group and tended to be lower in IMD group than in the glucose group. Finally, we also observed that, when compared with control rats, glucose of IMD significantly stimulated and improved the concentration of portal active GLP-1 in antibiotic-administered, caecectomised rats. We concluded that IMD was slowly digested and the resulting glucose stimulated GLP-1 secretion in rat small intestine. Oral delivery of slowly released IMD glucose to the small intestine probably exerts important, yet unknown, physiological effects on the recipient.


Subject(s)
Dextrins/administration & dosage , Dextrins/pharmacology , Glucagon-Like Peptide 1/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Maltose/analogs & derivatives , Administration, Oral , Animal Feed/analysis , Animals , Area Under Curve , Blood Glucose/drug effects , Diet/veterinary , Digestion , Gene Expression Regulation/drug effects , Half-Life , Male , Maltose/administration & dosage , Maltose/pharmacology , Rats , Rats, Sprague-Dawley
9.
Br J Nutr ; 123(5): 537-544, 2020 03 14.
Article in English | MEDLINE | ID: mdl-31791424

ABSTRACT

We investigated whether non-digestible saccharide fermentation-derived hydrogen molecules (H2) in rat colon could improve the in vivo reduction-oxidation (redox) balance via regeneration of α-tocopherol, by assessing their effect on hydroxyl radicals, the α-tocopherol concentration and the redox balance. In Expt 1, a Fenton reaction with phenylalanine (0 or 1·37 mmol/l of H2) was conducted. In Expt 2, rats received intraperitoneally maize oil containing phorone (400 mg/kg) 7 d after drinking ad libitum water containing 0 or 4 % fructo-oligosaccharides (FOS) (groups CP and FP, respectively). In Expt 3, rats unable to synthesise ascorbic acid drank ad libitum for 14 d water with 240 mg ascorbic acid/l (group AC), 20 mg of ascorbic acid/l (group DC) or 20 mg of ascorbic acid/l and 4 % FOS (group DCF). In the Fenton reaction, H2 reduced tyrosine produced from phenylalanine to 72 % when platinum was added and to 92 % when platinum was excluded. In Expt 2, liver glutathione was depleted by administration of phorone to rats. However, compared with CP, no change in the m-tyrosine concentration in the liver of FP was detected. In Expt 3, net H2 excretion was higher in DCF than in the other rats after 3 d of the experiment. Furthermore, the concentrations of H2 and α-tocopherol and the redox glutathione ratio in perirenal adipose tissue of rats were significantly higher in DCF than in DC. To summarise, in rat colon, fermentation-derived H2 further shifted the redox balance towards a more reducing status in perirenal adipose tissue through increased regeneration of α-tocopherol.


Subject(s)
Hydrogen/metabolism , Oxidation-Reduction/drug effects , Regeneration/drug effects , alpha-Tocopherol/metabolism , Adipose Tissue/metabolism , Animals , Colon/metabolism , Dietary Carbohydrates/administration & dosage , Fermentation , Kidney/metabolism , Oligosaccharides/administration & dosage , Rats
10.
EBioMedicine ; 48: 513-525, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31521614

ABSTRACT

BACKGROUND: The dysbiosis of gut microbiota has been implicated in the pathogenesis of inflammatory bowel diseases; however, the underlying mechanisms have not yet been elucidated. Heavily glycosylated mucin establishes a first-line barrier against pathogens and serves as a niche for microbial growth. METHODS: To elucidate relationships among dysbiosis, abnormal mucin utilisation, and microbial metabolic dysfunction, we analysed short-chain fatty acids (SCFAs) and mucin components in stool samples of 40 healthy subjects, 49 ulcerative colitis (UC) patients, and 44 Crohn's disease (CD) patients from Japan. FINDINGS: Levels of n-butyrate were significantly lower in stools of both CD and UC patients than in stools of healthy subjects. Correlation analysis identified seven bacterial species positively correlated with n-butyrate levels; the major n-butyrate producer, Faecalibacterium prausnitzii, was particularly underrepresented in CD patients, but not in UC patients. In UC patients, there were inverse correlations between mucin O-glycan levels and the production of SCFAs, such as n-butyrate, suggesting that mucin O-glycans serve as an endogenous fermentation substrate for n-butyrate production. Indeed, mucin-fed rodents exhibited enhanced n-butyrate production, leading to the expansion of RORgt+Treg cells and IgA-producing cells in colonic lamina propria. Microbial utilisation of mucin-associated O-glycans was significantly reduced in n-butyrate-deficient UC patients. INTERPRETATION: Mucin O-glycans facilitate symbiosynthesis of n-butyrate by gut microbiota. Abnormal mucin utilisation may lead to reduced n-butyrate production in UC patients. FUND: Japan Society for the Promotion of Science, Health Labour Sciences Research Grant, AMED-Crest, AMED, Yakult Foundation, Keio Gijuku Academic Development Funds, The Aashi Grass Foundation, and The Canon Foundation.


Subject(s)
Homeostasis , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mucins/metabolism , Polysaccharides/metabolism , Adult , Animals , Biomarkers , Butyrates/metabolism , Case-Control Studies , Female , Gastrointestinal Microbiome , Humans , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Male , Metagenome , Metagenomics , Mice , Middle Aged , Symbiosis
11.
J Nutr Sci Vitaminol (Tokyo) ; 64(5): 357-366, 2018.
Article in English | MEDLINE | ID: mdl-30381626

ABSTRACT

The effects of fructo-oligosaccharides (FOS) on gut-barrier function are still controversial in human and animal studies. Diet conditions would be a major factor for the controversy in animal studies. We fed rats a semi-purified (SP) or a non-purified diet (NP) with or without FOS (60 g/kg diet) for 9 (experiment 1) or 10 d (experiment 2). We assessed microbial fermentation, gut permeability, and inflammatory responses in the cecum (experiment 1), and mucus layer in the cecum, intestinal transit time and microbiota composition (experiment 2). FOS supplementation induced a very acidic fermentation due to the accumulation of lactate and succinate in SP, while short-chain fatty acids were major products in NP. Gut permeability estimated by urinary chromium-EDTA excretion, bacterial translocation into mesenteric lymph nodes, myeloperoxidase activity, and expressions of the inflammatory cytokine genes in the cecal mucosa were greater in SP+FOS than in SP, but these alterations were not observed between NP and NP+FOS (experiment 1). FOS supplementation destroyed the mucus layer on the epithelial surface in SP, but not in NP. Intestinal transit time was 3-fold longer in SP+FOS than in SP, but this was not the case between NP and NP+FOS. Lower species richness of cecal microbiota was manifest solely in SP+FOS (experiment 2). These factors suggest that impact of FOS on gut permeability and inflammatory responses in the cecal mucosa quite differs between SP and NP. Increased gut permeability in SP+FOS could be evoked by the disruption of the mucus layer due to stasis of the very acidic luminal contents.


Subject(s)
Animal Feed , Cecum/drug effects , Diet , Gastrointestinal Microbiome/drug effects , Inflammation , Intestinal Mucosa/drug effects , Oligosaccharides/pharmacology , Animals , Bacterial Translocation/drug effects , Cecum/metabolism , Cecum/microbiology , Cecum/pathology , Chromium/urine , Cytokines/metabolism , Digestion , Edetic Acid/urine , Fatty Acids, Volatile/metabolism , Fermentation , Fructose/pharmacology , Gastrointestinal Transit/drug effects , Inflammation/etiology , Inflammation/metabolism , Inflammation/prevention & control , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lactic Acid/metabolism , Male , Permeability , Peroxidase/metabolism , Prebiotics , Rats, Wistar , Succinic Acid/metabolism
12.
J Agric Food Chem ; 66(45): 11909-11916, 2018 Nov 14.
Article in English | MEDLINE | ID: mdl-30354117

ABSTRACT

We hypothesized that water-soluble cellulose acetate (WSCA) could be useful tool for the delivery of short-chain fatty acids to the large intestine. Rats were fed a control diet or a diet containing graded levels of WSCA for up to 21 days. Consuming WSCA dose-dependently increased large-bowel acetate and propionate concentrations through the bacterial fermentation. When WSCA was used as substrate, acetyl esterase activity in the cecal bacteria was detected solely in rats fed WSCA, in which the activity increased over time accompanied by an increased number of Bacteroides xylanisolvens. Consuming WSCA at a 4% level increased the goblet cell numbers and mucin contents in the cecum and lowered plasma cholesterol concentrations, which tended to correlate with the portal plasma concentrations of propionate. The results suggest that bacterial fermentation of WSCA is characterized by the greater production of acetate and propionate, which may contribute to the physiologic alterations.


Subject(s)
Acetates/metabolism , Bacteria/metabolism , Cellulose/analogs & derivatives , Cholesterol/blood , Intestine, Large/microbiology , Propionates/metabolism , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Cellulose/metabolism , Dietary Fiber/analysis , Dietary Fiber/metabolism , Fermentation , Gastrointestinal Microbiome , Intestine, Large/metabolism , Male , Rats , Rats, Wistar
13.
J Nutr ; 147(10): 1900-1908, 2017 10.
Article in English | MEDLINE | ID: mdl-28835391

ABSTRACT

Background: The mechanism underlying transient increases in immunoglobulin (Ig) A concentrations in the cecal contents of rats fed fructo-oligosaccharide (FOS) is unclear.Objective: This study was designed to test whether increased IgA concentrations represent one aspect of the inflammatory response to increased permeability induced by FOS in the cecum.Methods: Seven-week-old male Wistar rats were fed a fiber-free semipurified diet (FFP) with or without supplemental FOS (60 g/kg diet) for 9 or 58 d [experiment (expt.) 1], 7 d (expt. 2), or 7 or 56 d (expt. 3). In addition to measuring IgA concentrations in cecal content, we assessed gut permeability, inflammatory responses (expt. 1), the number of IgA plasma cells in the cecal lamina propria, polymeric Ig receptor (pIgR) expression in the cecal mucosa (expt. 2), and the condition of the cecal mucus layer (expt. 3).Results: The cecal IgA concentration in the FOS-fed rats was 15-fold higher than that of the rats fed FFP for 9 d (P < 0.05). Gut permeability estimated by urinary chromium-EDTA excretion, bacterial translocation to mesenteric lymph nodes, myeloperoxidase activity, and expression of inflammatory cytokine genes in the cecal mucosa was greater in the FOS-fed rats than in the rats fed FFP for 9 d. These effects were not observed in the rats fed FOS for 58 d (expt. 1). Accompanying the higher cecal IgA concentration, pIgR protein and the number of IgA plasma cells in the cecal mucosa were higher in the FOS-fed rats than in the rats fed FFP for 7 d (expt. 2). Destruction of the mucus layer on the epithelial surface, as evidenced by Alcian blue staining in the cecal sections, was evident in the rats fed FOS for 7 d, but the mucus layer appeared normal in the rats fed FOS for 56 d (expt. 3).Conclusions: These findings suggest that transient increases in cecal IgA concentrations induced by FOS in rats are associated with mucosal inflammation in response to increased gut permeability; these are presumably evoked by disruption of the cecal mucus barrier. The observed responses could contribute to the maturation of the gut immune system.


Subject(s)
Cecum/metabolism , Fructose/pharmacology , Immunoglobulin A/metabolism , Intestinal Mucosa/drug effects , Mucositis/metabolism , Oligosaccharides/pharmacology , Prebiotics , Animals , Bacterial Translocation , Cecum/drug effects , Cecum/pathology , Cytokines/metabolism , Fructose/immunology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lymph Nodes , Male , Mesentery , Mucositis/etiology , Mucositis/pathology , Oligosaccharides/immunology , Permeability , Peroxidase/metabolism , Rats, Wistar , Receptors, Polymeric Immunoglobulin/metabolism
14.
J Nutr ; 147(3): 330-336, 2017 03.
Article in English | MEDLINE | ID: mdl-28100603

ABSTRACT

Background: Digestion-resistant dextrin derivatives (DRDDs), including resistant maltodextrin (RM), polydextrose, and resistant glucan (RG), have been developed as low-energy foods. However, data on the resistance of DRDDs to small-intestinal digestion are scarce.Objective: We sought to determine the site and extent of DRDD breakdown in the rat intestine and to predict its energy contributions.Methods: In vitro small-intestinal resistance of DRDDs was evaluated by the AOAC method for dietary fiber measurement and by artificial digestion with the use of pancreatic α-amylase and brush-boarder membrane vesicles. In vivo small-intestinal resistance of DRDDs was determined from the feces of male ileorectostomized Sprague-Dawley rats fed a control diet or a diet containing one of the DRDDs at 50 g/kg for 9 d (period 1) and then for 10 d (period 2), during which they received 1 g neomycin/L in their drinking water. Separately, male Sprague-Dawley rats were fed the same diets for 4 wk, and the whole-gut recoveries of DRDDs were determined from feces at days 8-10.Results: Small-intestinal resistances determined in vitro by artificial digestion (RM: 70%; polydextrose: 67%; RG: 69%) were lower than those measured by the AOAC method (RM: 92%; polydextrose: 80%; RG: 82%). In the ileorectostomized rats, fecal dry-matter excretions were consistently greater in the DRDDs than in the control. The small-intestinal resistances of the DRDDs were 68%, 58%, and 62% in period 1 and 66%, 61%, and 67% during period 2 for RM, polydextrose, and RG, respectively. The resistances did not differ among the DRDDs at either time. In the normal rats, food intakes and body weight gains did not differ among the groups. The whole-gut recovery of RM (13%) was lower than that of polydextrose (33%) and RG (29%), which did not differ.Conclusions: DRDDs were more digestible in the rat small intestine than the AOAC method. The energy contribution from small-intestine digestibility, not just large-bowel fermentability, must be considered in determining the energy contribution of DRDDs. Whether humans respond similarly needs to be tested.


Subject(s)
Dextrins/chemistry , Dietary Fiber/metabolism , Digestion/physiology , Intestine, Large/physiology , Intestine, Small/physiology , Animal Feed/analysis , Animals , Dextrins/metabolism , Diet/veterinary , Energy Metabolism , Feces/chemistry , Fermentation , Male , Rats
15.
Int J Food Sci Nutr ; 67(6): 678-85, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27282074

ABSTRACT

Using barley cultivars differing widely in ß-glucan content, we aimed to determine their effects on small intestinal macronutrient digestion in 24 ileorectostomised rats. The rats were fed 1 of 4 experimental diets, each containing a different barley variety, for 11 d. The diets had a content of 0, 2.1, 2.6 and 4.3 g of ß-glucan/100 g. Feed intake and faecal excretion of fat, protein, starch, and non-starch polysaccharides were determined in the final 5 d of the study and apparent macronutrient digestibility calculated. Higher dietary levels of ß-glucan (2.6% and 4.3%) lowered feed intake (by 15 and 19%, respectively) but final body weight was only lowered by the 4.3% ß-glucan diet relative to rats fed the 0% ß-glucan diet (all ps < 0.05). Protein, lipid and starch digestibility was unrelated to the dietary ß-glucan content. Higher dietary levels of barley ß-glucan lower feed intake of ileorectostomised rats, which is independent of intestinal fermentation and unrelated to macronutrient digestibility.


Subject(s)
Digestion , Hordeum/chemistry , Ileostomy , Intestine, Small/metabolism , Whole Grains/chemistry , beta-Glucans/analysis , Animals , Diet , Dietary Fats/administration & dosage , Dietary Fats/analysis , Dietary Proteins/administration & dosage , Dietary Proteins/analysis , Feces/chemistry , Fermentation , Male , Polysaccharides/administration & dosage , Polysaccharides/analysis , Rats , Rats, Sprague-Dawley , Starch/administration & dosage , Starch/analysis , beta-Glucans/administration & dosage
16.
Biosci Biotechnol Biochem ; 80(3): 564-73, 2016.
Article in English | MEDLINE | ID: mdl-26745604

ABSTRACT

Among 131 rice endosperm proteins previously identified by MS-based proteomics, most of the proteins showed low or almost no sequence similarity to known allergens in databases, whereas nine proteins did it significantly. The sequence of two proteins showed high overall identity with Hsp70-like hazel tree pollen allergen (Cor a 10) and barley α-amylase (Hor v 16), respectively, whereas the others showed low identity (28-58%) with lemon germin-like protein (Cit l 1), corn zein (Zea m 50 K), wheat chitinase-like xylanase inhibitor (Tri a XI), and kinase-like pollen allergen of Russian thistle (Sal k 1). Immuno-dot blot analysis showed that recombinant proteins for these rice seed homologs were positive in the IgE-binding, but not necessarily similarity dependent, from some allergic patients. These results suggest that utilization of proteome and sequence databases in combination with IgE-binding analysis was effective to screen and evaluate allergenic potential of rice seed protein components.


Subject(s)
Allergens/immunology , Databases, Protein , Immunoglobulin E/immunology , Oryza/immunology , Plant Proteins/immunology , Seeds/immunology , Oryza/embryology , Proteome , Recombinant Proteins/immunology
17.
Biosci Biotechnol Biochem ; 79(6): 992-6, 2015.
Article in English | MEDLINE | ID: mdl-25727739

ABSTRACT

We histologically examined the effects of elemental diet (ED) on the goblet cell profile in the rat small intestine. The sulfomucin goblet cells were predominant throughout the small intestine in the control group, while sialomucin goblet cells were manifest in the duodenum and jejunum in the ED group. Next, we investigated the possible relevance of luminal osmolality to the goblet cell profile. Gastric osmolality in the ED group was within the physiological range. Meanwhile, ingestion of high glucose diet elevated gastric osmolality and increased the number of sialomucin goblet cells in the duodenum and jejunum. Further, it turned out that the lower sulfur contents in ED was not related to the unique goblet cell profile by ED ingestion. It is inductively suggested that the influx of high concentrations of low molecular nutrients into the small intestine could be associated with the goblet cell alteration, but the alteration was not necessarily due to the changes in the gastric osmolality by ED ingestion.


Subject(s)
Duodenum/cytology , Food, Formulated/adverse effects , Goblet Cells/cytology , Goblet Cells/drug effects , Jejunum/cytology , Sialomucins/metabolism , Animals , Body Weight/drug effects , Cell Proliferation/drug effects , Duodenum/drug effects , Eating/drug effects , Goblet Cells/metabolism , Hydrogen-Ion Concentration , Jejunum/drug effects , Osmolar Concentration , Rats , Sulfur/adverse effects , Sulfur/analysis
18.
Biosci Biotechnol Biochem ; 78(9): 1592-602, 2014.
Article in English | MEDLINE | ID: mdl-25209509

ABSTRACT

We examined the effects of fructo-oligosaccharides (FOS) on IgA and mucin secretion in the rat cecum after different ingestion periods. Rats were fed a control diet or a diet containing FOS for 1, 2, 4, and 8 wk. FOS ingestion greatly increased IgA and mucin concentrations at 1 and 2 wk, but the effects were disappeared or attenuated at 4 and 8 wk. After 1 wk, FOS induced higher lactobacilli and lactate concentrations and lower cecal pH in the cecum, but the alterations were moderated with the prolonged ingestion accompanying with increasing short-chain fatty acid concentrations. At 1 and 2 wk, FOS increased IgA plasma cells and polymeric immunoglobulin receptor expression in the cecal mucosa and strongly depressed fecal mucinase activities related to the lower cecal pH. These findings may explain the FOS-induced early elevation of IgA and mucin. Clearly, FOS effects on IgA and mucin secretion considerably differ depending on the ingestion period.


Subject(s)
Cecum/metabolism , Immunoglobulin A/biosynthesis , Mucins/biosynthesis , Oligosaccharides/administration & dosage , Animals , Cecum/enzymology , Diet , Eating , Fatty Acids, Volatile , Feces , Humans , Immunoglobulin A/metabolism , Lactic Acid/metabolism , Lactobacillus , Mucins/metabolism , Polysaccharide-Lyases/metabolism , Rats
19.
Nature ; 504(7480): 446-50, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24226770

ABSTRACT

Gut commensal microbes shape the mucosal immune system by regulating the differentiation and expansion of several types of T cell. Clostridia, a dominant class of commensal microbe, can induce colonic regulatory T (Treg) cells, which have a central role in the suppression of inflammatory and allergic responses. However, the molecular mechanisms by which commensal microbes induce colonic Treg cells have been unclear. Here we show that a large bowel microbial fermentation product, butyrate, induces the differentiation of colonic Treg cells in mice. A comparative NMR-based metabolome analysis suggests that the luminal concentrations of short-chain fatty acids positively correlates with the number of Treg cells in the colon. Among short-chain fatty acids, butyrate induced the differentiation of Treg cells in vitro and in vivo, and ameliorated the development of colitis induced by adoptive transfer of CD4(+) CD45RB(hi) T cells in Rag1(-/-) mice. Treatment of naive T cells under the Treg-cell-polarizing conditions with butyrate enhanced histone H3 acetylation in the promoter and conserved non-coding sequence regions of the Foxp3 locus, suggesting a possible mechanism for how microbial-derived butyrate regulates the differentiation of Treg cells. Our findings provide new insight into the mechanisms by which host-microbe interactions establish immunological homeostasis in the gut.


Subject(s)
Butyrates/metabolism , Cell Differentiation , Colon/immunology , Colon/microbiology , Fermentation , Symbiosis , T-Lymphocytes, Regulatory/cytology , Acetylation/drug effects , Adoptive Transfer , Animals , Butyrates/analysis , Butyrates/pharmacology , Cell Differentiation/drug effects , Colitis/drug therapy , Colitis/pathology , Colon/cytology , Colon/metabolism , Conserved Sequence , Female , Forkhead Transcription Factors/genetics , Germ-Free Life , Histones/metabolism , Homeostasis/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Lymphocyte Count , Magnetic Resonance Spectroscopy , Male , Metabolome , Mice , Promoter Regions, Genetic/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
20.
Biosci Biotechnol Biochem ; 77(8): 1782-4, 2013.
Article in English | MEDLINE | ID: mdl-23924719

ABSTRACT

We examined the effect or not of an antibiotic treatment on dietary fiber-induced goblet cell proliferation in the rat ileum. The number of goblet cells in the ileum increased when rats consumed dietary fiber. However, this effect was maintained with a concurrent treatment of antibiotics, suggesting that the fiber effect on goblet cell response would remain irrespective of a bacterial component such as endotoxin.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Cell Proliferation/drug effects , Goblet Cells/drug effects , Animals , Dietary Fiber/administration & dosage , Endotoxins/metabolism , Goblet Cells/metabolism , Ileum/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...