Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Sci Rep ; 8(1): 74, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29311617

ABSTRACT

Peptide-YY (PYY) and Glucagon-Like Peptide-1 (GLP-1) play important roles in the regulation of food intake and insulin secretion, and are of translational interest in the field of obesity and diabetes. PYY production is highest in enteroendocrine cells located in the distal intestine, mirroring the sites where high concentrations of short chain fatty acids (SCFAs) are produced by gut microbiota. We show here that propionate and butyrate strongly increased expression of PYY but not GCG in human cell line and intestinal primary culture models. The effect was predominantly attributable to the histone deacetylase inhibitory activity of SCFA and minor, but significant contributions of FFA2 (GPR43). Consistent with the SCFA-dependent elevation of PYY gene expression, we also observed increased basal and stimulated PYY hormone secretion. Interestingly, the transcriptional stimulation of PYY was specific to human-derived cell models and not reproduced in murine primary cultures. This is likely due to substantial differences in PYY gene structure between mouse and human. In summary, this study revealed a strong regulation of PYY production by SCFA that was evident in humans but not mice, and suggests that high fibre diets elevate plasma concentrations of the anorexigenic hormone PYY, both by targeting gene expression and hormone secretion.


Subject(s)
Enteroendocrine Cells/metabolism , Fatty Acids, Volatile/metabolism , Peptide YY/metabolism , Cell Line , Cells, Cultured , Enteroendocrine Cells/drug effects , Fatty Acids, Volatile/pharmacology , Gene Expression , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Humans , Peptide YY/genetics
3.
Int J Obes (Lond) ; 38(11): 1440-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24480860

ABSTRACT

BACKGROUND AND OBJECTIVES: Gut hormones secreted by enteroendocrine cells (EECs) play a major role in energy regulation. Differentiation of EEC is controlled by the expression of basic helix-loop-helix (bHLH) transcription factors. High-fat (HF) feeding alters gut hormone levels; however, the impact of HF feeding on bHLH transcription factors in mediating EEC differentiation and subsequent gut hormone secretion and expression is not known. METHODS: Outbred Sprague-Dawley rats were maintained on chow or HF diet for 12 weeks. Gene and protein expression of intestinal bHLH transcription factors, combined with immunofluorescence studies, were analyzed for both groups in the small intestine and colon. Gut permeability, intestinal lipid and carbohydrate transporters as well as circulating levels and intestinal protein expression of gut peptides were determined. RESULTS: We showed that HF feeding resulted in hyperphagia and increased adiposity. HF-fed animals exhibited decreased expression of bHLH transcription factors controlling EEC differentiation (MATH1, NGN3, NEUROD1) and increased expression of bHLH factors modulating enterocyte expression. Furthermore, HF-fed animals had decreased number of total EECs and L-cells. This was accompanied by increased gut permeability and expression of lipid and carbohydrate transporters, and a decrease in circulating and intestinal gut hormone levels. CONCLUSIONS: Taken together, our results demonstrate that HF feeding caused decreased secretory lineage (that is, EECs) differentiation through downregulation of bHLH transcription factors, resulting in reduced EEC number and gut hormone levels. Thus, impaired EEC differentiation pathways by HF feeding may promote hyperphagia and subsequent obesity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Diet, High-Fat , Dietary Fats/adverse effects , Enteroendocrine Cells/metabolism , Gastrointestinal Hormones/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Obesity/metabolism , Animals , Blotting, Western , Cell Differentiation/drug effects , Disease Models, Animal , Energy Intake , Energy Metabolism , Hyperphagia , Intestinal Mucosa/cytology , Male , Obesity/pathology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
4.
Am J Physiol Gastrointest Liver Physiol ; 287(6): G1168-74, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15308471

ABSTRACT

The mucus layer covering the gastrointestinal mucosa is considered the first line of defense against aggressions arising from the luminal content. It is mainly composed of high molecular weight glycoproteins called mucins. Butyrate, a short-chain fatty acid produced during carbohydrate fermentation, has been shown to increase mucin secretion. The aim of this study was to test 1) whether butyrate regulates the expression of various MUC genes, which are coding for protein backbones of mucins, and 2) whether this effect depends on butyrate status as the major energy source of colonocytes. Butyrate was provided at the apical side of human polarized colonic goblet cell line HT29-Cl.16E in glucose-rich or glucose-deprived medium. In glucose-rich medium, butyrate significantly increased MUC3 and MUC5B expression (1.6-fold basal level for both genes), tended to decrease MUC5AC expression, and had no effect on MUC2 expression. In glucose-deprived medium, i.e., when butyrate was the only energy source available, MUC3 and MUC5B increase persisted, whereas MUC5AC expression was significantly enhanced (3.7-fold basal level) and MUC2 expression was strikingly increased (23-fold basal level). Together, our findings show that butyrate is able to upregulate colonic mucins at the transcriptional level and even better when it is the major energy source of the cells. Thus the metabolism of butyrate in colonocytes is closely linked to some of its gene-regulating effects. The distinct effects of butyrate according to the different MUC genes could influence the composition and properties of the mucus gel and thus its protective function.


Subject(s)
Butyrates/pharmacology , Gene Expression Regulation/drug effects , Glucose/deficiency , Goblet Cells/metabolism , Intestinal Mucosa/metabolism , Mucins/biosynthesis , Mucins/genetics , Cell Death , Culture Media , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fatty Acids/metabolism , Goblet Cells/drug effects , HT29 Cells , Humans , Hydrogen-Ion Concentration , Hydroxamic Acids/pharmacology , L-Lactate Dehydrogenase/metabolism , Mucin-2 , Mucin-3 , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
5.
Dig Liver Dis ; 35(8): 557-65, 2003 Aug.
Article in English | MEDLINE | ID: mdl-14567460

ABSTRACT

BACKGROUND: Cyclooxygenase (COX)-2 is up-regulated in most colorectal cancers. Chronic use of non-steroidal anti-inflammatory drugs, which target cyclooxygenases, have been shown to reduce the risk of these cancers. However, the mechanisms underlying this protective effect remain unclear. AIMS: The aim of our study was to characterize the effects of two COX-2 selective inhibitors, NS-398 and nimesulide, on colorectal cancer cell proliferation, and to describe the molecular mechanisms involved. MATERIALS AND METHODS: HT-29 and SW-1116 cell lines were cultured with either NS-398 or nimesulide. Cell proliferation was assessed by staining DNA with crystal violet. Cell cycle repartition and apoptosis were analysed by flow cytometry. The expression of COX-1 and COX-2. and of two cyclin dependent kinase inhibitors, p21Cip1 and p27Kip1, was analysed by Western blotting and RT-PCR. RESULTS: Both drugs dose-dependently inhibited cell proliferation and induced G1 cell cycle blockade. HT-29 cells were more sensitive to both drugs than SW-1116 cells. p21Cip1 and p27Kip1 were induced on both cell lines. Concomitant induction of p21Cip1 mRNA indicates transcriptional modulation, whereas induction of p27Kip1 only at the protein level suggests post-translational modulation. CONCLUSION: NS-398 and nimesulide inhibit colorectal cell proliferation through induction of p21Cip1 and p27Kip1.


Subject(s)
Adenocarcinoma/metabolism , Cell Division/drug effects , Colorectal Neoplasms/metabolism , Cyclooxygenase Inhibitors/pharmacology , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , Apoptosis , Blotting, Western , Cell Cycle Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/biosynthesis , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Enzyme Inhibitors/metabolism , Flow Cytometry , G1 Phase/drug effects , Humans , Isoenzymes/metabolism , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Proteins/biosynthesis
6.
Aliment Pharmacol Ther ; 18(7): 731-40, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14510747

ABSTRACT

BACKGROUND: Cyclo-oxygenase-2 over-expression has been reported in most advanced human colorectal cancers. AIMS: To assess the prevalence of cyclo-oxygenase-2 over-expression in non-advanced colorectal cancers, to investigate the correlation between cyclo-oxygenase-2 status and tumour clinicopathological features and molecular phenotype, and to determine the impact of cyclo-oxygenase-2 status on long-term clinical outcome. METHODS: Sixty-one patients who had undergone surgery for colorectal cancer without lymph node involvement were evaluated retrospectively. Cyclo-oxygenase-2 expression was determined by immunohistochemistry. The tumour replication error phenotype was assessed by amplification of the two microsatellites, BAT-25 and BAT-26. RESULTS: Thirty-six tumours were classified as cyclo-oxygenase-2 positive and 25 as cyclo-oxygenase-2 negative. No correlation was found between cyclo-oxygenase-2 over-expression and clinicopathological features or molecular phenotype. Cyclo-oxygenase-2 over-expression was an independent predictor of a poor prognosis. Indeed, the relative risk of tumour recurrence or death for patients with cyclo-oxygenase-2-positive tumours was 2.13 times that of patients with cyclo-oxygenase-2-negative tumours (P=0.008; 95% confidence interval, 1.22-3.73). This difference remained significant when post-operative deaths were censored in the multivariate analysis (P=0.014). CONCLUSION: Cyclo-oxygenase-2 over-expression is not associated with tumour phenotype, but is indicative of a poorer clinical outcome in patients with non-advanced colorectal carcinoma.


Subject(s)
Colorectal Neoplasms/metabolism , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Aged , Cyclooxygenase 2 , DNA, Neoplasm/genetics , Disease-Free Survival , Female , Humans , Immunohistochemistry/methods , Male , Membrane Proteins , Microsatellite Repeats , Neoplasm Recurrence, Local/metabolism , Phenotype , Prognosis , Regression Analysis , Retrospective Studies , Survival Analysis
7.
J Appl Microbiol ; 94(2): 312-20, 2003.
Article in English | MEDLINE | ID: mdl-12534824

ABSTRACT

AIMS: In vitro studies have suggested that fructo-oligosaccharides (FOS) and resistant starch (two fermentable non-digestible carbohydrates) display different fermentation kinetics. This study investigated whether these substrates affect the metabolic activity and bacterial composition of the intestinal microflora differently depending on the caecocolonic segment involved. METHODS AND RESULTS: Eighteen rats were fed a low-fibre diet (Basal) or the same diet containing raw potato starch (RPS) (9%) or short-chain FOS (9%) for 14 days. Changes in wet-content weights, bacterial populations and metabolites were investigated in the caecum, proximal and distal colon and faeces. Both substrates exerted a prebiotic effect compared with the Basal diet. However, FOS increased lactic acid-producing bacteria (LAPB) throughout the caecocolon and in faeces, whereas the effect of RPS was limited to the caecum and proximal colon. As compared with RPS, FOS doubled the pool of caecal fermentation products, while the situation was just the opposite distally. This difference was mainly because of the anatomical distribution of lactate, which accumulated in the caecum with FOS and in the distal colon with RPS. Faeces reflected these impacts only partly, showing the prebiotic effect of FOS and the metabolite increase induced by RPS. CONCLUSIONS: This study demonstrates that FOS and RPS exert complementary caecocolonic effects. SIGNIFICANCE AND IMPACT OF THE STUDY: The RPS and FOS combined ingestion could be beneficial by providing health-promoting effects throughout the caecocolon.


Subject(s)
Bacteria, Anaerobic/isolation & purification , Intestine, Large/microbiology , Oligosaccharides/pharmacology , Starch/pharmacology , Animals , Bacteria, Anaerobic/drug effects , Bacteria, Anaerobic/metabolism , Cecum/microbiology , Colon/microbiology , Colony Count, Microbial , Fermentation/drug effects , Lactic Acid/biosynthesis , Male , Rats , Rats, Wistar , Solanum tuberosum
8.
Am J Physiol Gastrointest Liver Physiol ; 281(6): G1340-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11705738

ABSTRACT

To assess the effect of glutamine availability on rates of protein synthesis in human enterocytes, Caco-2 cells were grown until differentiation and then submitted to glutamine deprivation produced by exposure to glutamine-free medium or methionine sulfoximine [L-S-[3-amino-3-carboxypropyl]-S-methylsulfoximine (MSO)], a glutamine synthetase inhibitor. Cells were then incubated with (2)H(3)-labeled leucine with or without glutamine, and the fractional synthesis rate (FSR) of total cell protein was determined from (2)H(3)-labeled enrichments in protein-bound and intracellular free leucine measured by gas chromatography-mass spectrometry. Both protein FSR (28 +/- 1.5%/day) and intracellular glutamine concentration (6.1 +/- 0.6 micromol/g protein) remained unaltered when cells were grown in glutamine-free medium. In contrast, MSO treatment resulted in a dramatic reduction in protein synthesis (4.6 +/- 0.6 vs. 20.2 +/- 0.8%/day, P < 0.01). Supplementation with 0.5-2 mM glutamine for 4 h after MSO incubation, but not with glycine nor glutamate, restored protein FSR to control values (24 +/- 1%/day). These results demonstrate that in Caco-2 cells, 1) de novo glutamine synthesis is highly active, since it can maintain intracellular glutamine pool during glutamine deprivation, 2) inhibition of glutamine synthesis is associated with reduced protein synthesis, and 3) when glutamine synthesis is depressed, exogenous glutamine restores normal intestinal FSR. Due to the limitations intrinsic to the use of a cell line as an experimental model, the physiological relevance of these findings for the human intestine in vivo remains to be determined.


Subject(s)
Enterocytes/metabolism , Glutamine/administration & dosage , Protein Biosynthesis , Apolipoproteins A/metabolism , Caco-2 Cells , Culture Media , Deuterium , Enterocytes/chemistry , Enzyme Inhibitors/pharmacology , Gas Chromatography-Mass Spectrometry , Glutamate-Ammonia Ligase/antagonists & inhibitors , Glutamine/analysis , Glutamine/metabolism , Humans , Kinetics , Methionine Sulfoximine/pharmacology
10.
Eur J Biochem ; 267(21): 6435-42, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11029587

ABSTRACT

Butyrate is a short chain fatty acid (SCFA) produced by bacterial fermentation of dietary fibers in the colon lumen which severely affects the proliferation of colon cancer cells in in vitro experiments. Although butyrate is able to interfere with numerous cellular targets including cell cycle regulator expression, little is known about butyrate metabolism and its possible involvement in its effect upon colon carcinoma cell growth. In this study, we found that HT-29 Glc-/+ cells strongly accumulated and oxidized sodium butyrate without producing ketone bodies, nor modifying oxygen consumption nor mitochondrial ATP synthesis. HT-29 cells accumulated and oxidized sodium acetate at a higher level than butyrate. However, sodium butyrate, but not sodium acetate, reduced cell growth and increased the expression of the cell cycle effector cyclin D3 and the inhibitor of the G1/S cdk-cyclin complexes p21/WAF1/Cip1, demonstrating that butyrate metabolism downstream of acetyl-CoA synthesis is not required for the growth-restraining effect of this SCFA. Furthermore, HT-29 cells modestly incorporated the 14C-labelled carbon from sodium butyrate into cellular triacylglycerols and phospholipids. This incorporation was greatly increased when D-glucose was present in the incubation medium, corresponding to the capacity of hexose to circulate in the pentose phosphate pathway allowing NADPH synthesis required for lipogenesis. Interestingly, when HT-29 cells were cultured in the presence of sodium butyrate, their capacity to incorporate 14C-labelled sodium butyrate into triacylglycerols and phospholipids was increased more than twofold. In such experimental conditions, HT-29 cells when observed under an electronic microscope, were found to be characterized by an accumulation of lipid droplets in the cytosol. Our data strongly suggest that butyrate acts upon colon carcinoma cells upstream of acetyl-CoA synthesis. In contrast, the metabolism downstream of acetyl-CoA [i.e. oxidation in the tricarboxylic acid (TCA) cycle and lipid synthesis] likely acts as a regulator of butyrate intracellular concentration.


Subject(s)
Acetyl Coenzyme A/biosynthesis , Butyrates/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Blotting, Western , Butyrates/pharmacology , Cell Division/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Respiration/drug effects , Cell Size/drug effects , Colonic Neoplasms/enzymology , Cyclin D3 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Dipeptidyl Peptidase 4/metabolism , Gene Expression Regulation/drug effects , Humans , Ketone Bodies/metabolism , Ornithine Decarboxylase/metabolism , Phospholipids/metabolism , Sodium Acetate/metabolism , Sodium Acetate/pharmacology , Triglycerides/metabolism , Tumor Cells, Cultured
11.
Dig Dis Sci ; 45(8): 1623-30, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11007115

ABSTRACT

Short-chain fatty acids are the main end products of bacterial fermentation of carbohydrates. Their role on the metabolism and biology of colonocytes is now well characterized. However, the functional consequences of their presence on intestinal smooth muscle cells remain poorly studied. We aimed to assess the effect of different short-chain fatty acids on ileal and colonic smooth muscle cells in primary culture and on A7R5 line. Butyrate (above 0.1 mM) inhibited A7R5 cell proliferation, while at low concentration (0.05 to 0.5 mM) butyrate significantly stimulated the proliferation of ileal and colonic myocytes in primary culture. An inhibition was observed at higher concentrations. Collagenous and noncollagenous protein synthesis was stimulated by butyrate. Moreover, butyrate stimulated actin and myosin expression. Thus, butyrate, which is produced by dietary fiber fermentation, may affect intestinal muscles by directly acting at the molecular level on myocytes.


Subject(s)
Cytoskeletal Proteins/biosynthesis , Extracellular Matrix Proteins/biosynthesis , Fatty Acids, Volatile/pharmacology , Intestines/cytology , Muscle, Smooth/cytology , Animals , Butyrates/pharmacology , Cell Division/drug effects , Cells, Cultured , Colon/cytology , Ileum/cytology , Male , Rats , Rats, Wistar
12.
Gut ; 47(3): 397-403, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10940278

ABSTRACT

BACKGROUND/AIM: Proinflammatory cytokines are key factors in the pathogenesis of Crohn's disease (CD). Activation of nuclear factor kappa B (NFkappaB), which is involved in their gene transcription, is increased in the intestinal mucosa of CD patients. As butyrate enemas may be beneficial in treating colonic inflammation, we investigated if butyrate promotes this effect by acting on proinflammatory cytokine expression. METHODS: Intestinal biopsy specimens, isolated lamina propria cells (LPMC), and peripheral blood mononuclear cells (PBMC) were cultured with or without butyrate for assessment of secretion of tumour necrosis factor (TNF) and mRNA levels. NFkappaB p65 activation was determined by immunofluorescence and gene reporter experiments. Levels of NFkappaB inhibitory protein (IkappaBalpha) were analysed by western blotting. The in vivo efficacy of butyrate was assessed in rats with trinitrobenzene sulphonic acid (TNBS) induced colitis. RESULTS: Butyrate decreased TNF production and proinflammatory cytokine mRNA expression by intestinal biopsies and LPMC from CD patients. Butyrate abolished lipopolysaccharide (LPS) induced expression of cytokines by PBMC and transmigration of NFkappaB from the cytoplasm to the nucleus. LPS induced NFkappaB transcriptional activity was decreased by butyrate while IkappaBalpha levels were stable. Butyrate treatment also improved TNBS induced colitis. CONCLUSIONS: Butyrate decreases proinflammatory cytokine expression via inhibition of NFkappaB activation and IkappaBalpha degradation. These anti-inflammatory properties provide a rationale for assessing butyrate in the treatment of CD.


Subject(s)
Butyrates/therapeutic use , Crohn Disease/drug therapy , Gastrointestinal Agents/therapeutic use , NF-kappa B/drug effects , Adolescent , Adult , Aged , Animals , Blotting, Western , Cells, Cultured , Crohn Disease/metabolism , Dose-Response Relationship, Drug , Drug Evaluation , Female , Humans , Interleukin-1/metabolism , Interleukin-6/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , NF-kappa B/metabolism , Prospective Studies , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
13.
Gut ; 46(4): 507-14, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10716680

ABSTRACT

BACKGROUND: Sodium butyrate, a product of colonic bacterial fermentation, is able to inhibit cell proliferation and to stimulate cell differentiation of colonic epithelial cell lines. It has been proposed that these cellular effects could be linked to its ability to cause hyperacetylation of histone through the inhibition of histone deacetylase. AIM: To analyse the molecular mechanisms of butyrate action on cell proliferation/differentiation and to compare them with those of trichostatin A, a well known inhibitor of histone deacetylase. METHODS: HT-29 cells were grown in the absence or presence of butyrate or trichostatin A. Cell proliferation and cell cycle distribution were studied after DNA staining by crystal violet and propidium iodide respectively. Cell cycle regulatory proteins were studied by western blot and reverse transcription-polymerase chain reaction. Cell differentiation was followed by measuring brush border enzyme activities. Histone acetylation was studied by acid/urea/Triton acrylamide gel electrophoresis. RESULTS: Butyrate blocked cells mainly in the G(1) phase of the cell cycle, whereas trichostatin A was inhibitory in both G(1) and G(2) phases. Butyrate inhibited the mRNA expression of cyclin D1 without affecting its protein expression and stimulated the protein expression of cyclin D3 without affecting its mRNA expression. Trichostatin A showed similar effects on cyclin D1 and D3. Butyrate and trichostatin A stimulated p21 expression both at the mRNA and protein levels, whereas their effects on the expression of cyclin dependent kinases were slightly different. Moreover, butyrate strongly stimulated the activity of alkaline phosphatase and dipeptidyl peptidase IV, whereas trichostatin A had no effect. Finally, a six hour exposure to butyrate or trichostatin A induced histone H4 hyperacetylation. At 15 and 24 hours, histone H4 remained hyperacetylated in the presence of butyrate, whereas it returned to control levels in the presence of trichostatin A. CONCLUSIONS: The data may explain how butyrate acts on cell proliferation/differentiation, and they show that trichostatin A does not reproduce every effect of butyrate, mainly because of its shorter half life.


Subject(s)
Butyrates/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Intestinal Mucosa/drug effects , Acetylation , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Cyclin D1/genetics , Cyclin D3 , Cyclins/genetics , Epithelial Cells/drug effects , Gene Expression/drug effects , HT29 Cells , Histones/metabolism , Humans , Intestinal Mucosa/cytology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
14.
Gut ; 46(3): 370-5, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10673299

ABSTRACT

BACKGROUND: Peptide YY (PYY) is involved in the regulation of several gut functions, including secretion and motility. It exerts its effects through a family of six receptors, commonly named the Y receptor family. AIMS: To characterise the effects of PYY on strips of rat proximal colon in vitro, and to determine the pathways and receptors involved. METHODS: Contractions of strips removed from the muscle layer of rat proximal colon were recorded under isometric conditions, using PYY, Y receptor agonists and antagonists, and nerve blockers. Reverse transcription-polymerase chain reaction was also performed to detect the presence of mRNA coding for Y receptors. Finally, smooth muscle cells were isolated to estimate the cell length and intracellular Ca(2+) concentration in the presence and absence of PYY. RESULTS: PYY, neuropeptide Y (NPY), pancreatic polypeptide (PP) and [Leu31,Pro34]NPY induced a dose dependent contraction of strips from proximal colon. Tetrodotoxin partially inhibited the PYY and NPY induced contractions, and strongly inhibited the PP induced contraction. Specific antagonists showed the involvement of cholinergic nicotinic receptors and NK1 receptor. BIBP 3226, a specific Y1 antagonist, did not modify the colonic smooth muscle response to PYY, whereas blocking L-type Ca(2+) channels with D-600 abolished its effects. Moreover, PYY induced an increase in intracellular Ca(2+) concentration, associated with a reduction in cell length. mRNA encoding Y1 and Y4 receptors were detected in the muscle strips. CONCLUSIONS: These findings suggest that PYY stimulates colonic contractile activity in vitro through (a) a nervous Y4 dependent pathway and (b) a pathway involving a potential new receptor on myocytes.


Subject(s)
Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Peptide YY/pharmacology , Animals , Colon/drug effects , Colon/physiology , Dose-Response Relationship, Drug , Male , Muscle Contraction/physiology , Muscle, Smooth/physiology , Peptide YY/antagonists & inhibitors , Rats , Rats, Wistar , Receptors, Cholinergic/drug effects , Receptors, Cholinergic/physiology , Reverse Transcriptase Polymerase Chain Reaction
15.
J Nutr ; 129(12): 2231-5, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10573555

ABSTRACT

While the prebiotic effects of fructo-oligosaccharides (FOS), short-chain polymers of fructose, have been thoroughly described after 2-3 wk of ingestion, effects after intake for several months are unknown. We tested the hypothesis that these effects would differ after ingestion for short and long periods in rats. Rats were fed a basal low-fiber diet (Basal) or the same diet containing 9 g/100 g of FOS for 2, 8 or 27 wk, and cecal contents were collected at the end of each time period. Cecal short-chain fatty acid concentration was higher in rats fed FOS than in those fed Basal, and this effect persisted over time: 83.8 +/- 4.1 vs. 62.4 +/- 6.5 micromol/g at 2 wk and 103.5 +/- 5.8 vs. 73.2 +/- 7.4 micromol/g at 27 wk (P < 0.05). The molar butyrate ratio was higher in rats fed FOS regardless of the time period (14.8 +/- 0.6% vs. 6.7 +/- 1.1% at 27 wk, P < 0.05). Lactate concentration in rats fed FOS was elevated after 2 wk and then decreased: 63.5 +/- 21.6 micromol/g at 2 wk vs. 8.8 +/- 3.3 micromol/g at 8 wk (P < 0.05). After 2 wk, FOS increased the concentrations of total lactic acid-producing bacteria, and Lactobacillus sp. (P < 0.05), without modifying total anaerobes. However, most of these effects were abolished after 8 and 27 wk of FOS consumption. In the long term, the FOS-induced increase in intestinal lactic acid-producing bacteria was lost, but the butyrogenic properties of FOS were maintained.


Subject(s)
Bacteria/growth & development , Bacteria/metabolism , Butyrates/metabolism , Cecum/metabolism , Lactic Acid/biosynthesis , Oligosaccharides/pharmacology , Animals , Colony Count, Microbial , Fatty Acids, Volatile/metabolism , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Male , Oligosaccharides/administration & dosage , Rats , Rats, Wistar , Time Factors
16.
Planta Med ; 65(6): 527-31, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10483372

ABSTRACT

Ulvans (from Ulva lactuca) constitute a dietary fiber structurally similar to the mammalian glycosaminoglycans but with unexplored biological or cytotoxic activities. From native low-viscosity preparations containing 33.5 molar % and 18.4 molar % of sulfate residues and uronic acid residues, respectively, we derived desulfated, reduced and desulfated-reduced polysaccharides with respectively 5.2, 2.9, and 4.5-4.9 molar % of sulfate residues and uronic acid residues. The effects of these preparations were examined on the adhesion, proliferation and differentiation of normal or tumoral colonic epithelial cells cultured in conventional (0.3-0.8 x 10(6) cells/ml) or rotating bioreactor (3-8 x 10(6) cells/ml) culture conditions. In conventional culture conditions, ulvan modified the adhesion phase and the proliferation of normal colonic cells and undifferentiated HT-29 cells according to their molecular weights and to the relative molar proportion of sulfate residues. From the native polysaccharides, we have screened sulfated ulvans (MW < 5,000) which inhibited the Caco-2 cell proliferation/differentiation program by inducing a low cell reactivity to Ulex europeaus-1 lectins in defined (p < 0.001) or serum-supplemented media (p < 0.01) but were inactive on normal colonocytes. In conclusion, this dietary fiber could be a source of oligosaccharides with a bioactivity, a cytotoxicity or a cytostaticity targeted to normal or cancerous epithelial cells.


Subject(s)
Dietary Fiber/pharmacology , Epithelial Cells/drug effects , Intestinal Mucosa/cytology , Plant Extracts/chemistry , Polysaccharides/pharmacology , Seaweed , Animals , Cell Adhesion/drug effects , Cell Division/drug effects , Colonic Neoplasms , Colony-Forming Units Assay , Epithelial Cells/cytology , Humans , Intestinal Mucosa/drug effects , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Rats , Sulfuric Acids , Tumor Cells, Cultured
17.
Eur J Clin Invest ; 29(1): 48-55, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10092989

ABSTRACT

BACKGROUND: Colonic epithelial cells may behave as antigen-presenting cells. Interleukin 10 (IL-10) is known to play a major role in the intestinal immune system; however, it remains to be determined whether human intestinal epithelial cells express IL-10 receptor, and whether this cytokine modulates their expression of antigen presentation-associated molecules. METHODS: The binding of biotinylated IL-10 was studied in SW 1116, HT-29 and T84 human colonic epithelial cell lines and freshly isolated normal colonic epithelial cells. Reverse transcription-polymerase chain reaction was also performed to detect IL-10 receptor mRNA. The effect of IL-10 on antigen presentation associated molecules was assessed by flow cytometry. RESULTS: Biotinylated IL-10 bound to SW 1116, HT-29, T84, and normal colonic epithelial cells. IL-10 receptor mRNA was detected in SW 1116 and normal epithelial cells. SW 1116 and HT-29 cells expressed MHC class I and ICAM-1, but not CD80, and SW 1116 constitutively expressed HLA-DR. Interferon-gamma up-regulated HLA-DR and ICAM-1 expression on both cells, whereas lipopolysaccharide increased ICAM-1 expression only on SW 1116. IL-10 failed to modulate these antigens, even after stimulation by lipopolysaccharide or interferon-gamma. Moreover, these molecules decreased IL-10 binding in both lines. CONCLUSION: The presence of IL-10 receptor on intestinal epithelial cells suggest that IL-10 may play a role in mucosal physiology, however its effect on the immune response remains to be determined.


Subject(s)
Antigen Presentation , Colon/immunology , Interleukin-10/immunology , Intestinal Mucosa/immunology , Receptors, Interleukin/isolation & purification , Adenocarcinoma/immunology , B7-1 Antigen/immunology , Cell Separation , Colon/cytology , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/immunology , HLA-DR Antigens/immunology , Histocompatibility Antigens Class I/immunology , Humans , Intercellular Adhesion Molecule-1/immunology , Interferon-gamma/immunology , Intestinal Mucosa/cytology , Lipopolysaccharides/immunology , Major Histocompatibility Complex , Receptors, Interleukin-10 , Tumor Cells, Cultured
18.
Br J Nutr ; 82(5): 419-26, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10673915

ABSTRACT

Some data suggest that the colonic microflora may adapt to produce more butyrate if given time and the proper substrate. To test this hypothesis, we investigated the effect of prolonged feeding of resistant potato starch on butyrate production. Rats were fed on either a low-fibre diet (basal) or the same diet supplemented with 90 g resistant potato starch/kg (PoS) for 0.5, 2 and 6 months. Short-chain fatty acid (SCFA) concentrations were determined in caecal and colonic contents at the end of each ingestion period. Total SCFA concentration increased over time throughout the caecocolonic tract with PoS, but was not modified with the basal diet. While propionate concentration was unchanged, butyrate concentration was highly increased by PoS at each time period in both the caecum and colon. Moreover, the butyrogenic effect of PoS increased over time, and the amount of butyrate was increased 6-fold in the caecum and proximal colon and 3-fold in the distal colon after 6 months compared with 0.5 months. Accordingly, the ratio butyrate:- total SCFA increased over time throughout the caecocolonic tract (12.6 (SE 2.8) v. 28 (SE 1.8)% in the caecum, 10.5 (SE 1.4) v. 26.8 (SE 0.9)% in the proximal colon, and 7.3 (SE 2.4) v. 23.9 (SE 2.7)% in the distal colon at 0.5 v. 6 months respectively), while the proportion of acetate decreased. Neither the proportion nor the concentration of butyrate was modified over time with the basal diet. Butyrate production was thus promoted by long-term ingestion of PoS, from the caecum towards the distal colon, which suggests that a slow adaptive process occurs within the digestive tract in response to a chronic load of indigestible carbohydrates.


Subject(s)
Butyrates/metabolism , Dietary Fiber/administration & dosage , Intestine, Large/metabolism , Solanum tuberosum , Starch/administration & dosage , Animals , Cecum/metabolism , Cecum/microbiology , Colon/metabolism , Colon/microbiology , Colony Count, Microbial , Fatty Acids, Volatile/analysis , Feces/chemistry , Fermentation , Intestine, Large/microbiology , Lactic Acid/analysis , Male , Rats , Rats, Wistar , Time Factors
19.
Am J Physiol ; 275(6): G1415-22, 1998 12.
Article in English | MEDLINE | ID: mdl-9843779

ABSTRACT

Short-chain fatty acids (SCFAs) are recognized as the major anions of the large intestinal content in humans, but their effect on colonic motility is controversial. This study explores the colonic motor effect of SCFAs and their mechanisms in the rat. Colonic motility (electromyography) and transit time (plastic markers) were measured in conscious rats while SCFAs were infused into the colon, either alone or after administration of neural antagonists or immunoneutralization of circulating polypeptide YY (PYY). SCFA-induced PYY release was measured by RIA and then simulated by infusing exogenous PYY. Intracolonic infusion of 0.4 mmol/h SCFAs had no effect, whereas 2 mmol/h SCFAs reduced colonic motility (36 +/- 3 vs. 57 +/- 4 spike bursts/h with saline, P < 0.05) by decreasing the ratio of nonpropulsive to propulsive activity. This resulted in an increased transit rate (P < 0.01). Neither alpha-adrenoceptor blockade nor nitric oxide synthase inhibition prevented SCFA-induced motility reduction. Intraluminal procaine infusion suppressed the SCFA effect, indicating that a local neural mechanism was involved. SCFA colonic infusion stimulated PYY release in blood. Immunoneutralization of circulating PYY abolished the effect of SCFAs on colonic motility, whereas exogenous PYY infusion partly reproduced this effect. SCFAs modify colonic motor patterns in the rat and increase transit rate; local nerve fibers and PYY are involved in this effect.


Subject(s)
Colon/drug effects , Colon/innervation , Fatty Acids/pharmacology , Gastrointestinal Motility/drug effects , Peptide YY/metabolism , Animals , Colon/chemistry , Dose-Response Relationship, Drug , Electromyography , Fatty Acids/analysis , Gastrointestinal Transit/drug effects , Male , Myoelectric Complex, Migrating/drug effects , Myoelectric Complex, Migrating/physiology , Nervous System Physiological Phenomena , Osmolar Concentration , Peptide YY/antagonists & inhibitors , Peptide YY/pharmacology , Rats , Rats, Wistar
20.
Gut ; 43(6): 783-90, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9824605

ABSTRACT

BACKGROUND: Although leptin, an adipocyte derived hormone which regulates food intake and energy balance, is released after injections of tumour necrosis factor (TNF) and interleukin 1, plasma concentrations have not been characterised in chronic inflammation. Leptin may contribute to the anorexia and body weight loss associated particularly with the acute stages of inflammatory bowel disease. AIMS: To investigate plasma leptin concentrations during the time course of intestinal inflammation in different animal models. METHODS: Plasma leptin was measured at different time points in rats with trinitrobenzene sulphonic acid (TNBS) induced colitis, indomethacin induced ileitis, or endotoxic shock caused by lipopolysaccharide (LPS). Systemic TNF-alpha was also measured during acute inflammation. RESULTS: Plasma leptin concentrations increased fourfold eight hours after induction of TNBS colitis (p<0.0001) and twofold after administration of ethanol alone (p<0.02). Plasma leptin responses throughout the first post-treatment day were correlated with myeloperoxidase activity and gross damage scores. Similar leptin overexpression was observed in indomethacin induced ileitis and in rats with endotoxic shock. Plasma concentrations were lower in TNBS treated rats than in controls on day 5 before reaching a similar concentration on day 14. Anorexia and body weight loss were observed during the first four days post-TNBS. A significant increase in systemic TNF-alpha was only detected in LPS treated rats. CONCLUSION: Elevated plasma leptin concentrations, correlated with the degree of inflammation and associated with anorexia, were induced in rats during the early stages of experimental intestinal inflammation but proved transient; this might account for discrepancies in recent results concerning concentrations in patients with inflammatory bowel diseases.


Subject(s)
Colitis/metabolism , Ileitis/blood , Proteins/metabolism , Animals , Body Weight , Eating , Leptin , Male , Rats , Rats, Wistar , Trinitrobenzenesulfonic Acid/adverse effects , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...