Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
Plus de filtres










Gamme d'année
1.
Proc Natl Acad Sci U S A ; 113(40): E5934-E5943, 2016 10 04.
Article de Anglais | MEDLINE | ID: mdl-27638207

RÉSUMÉ

Diet is among the most important factors contributing to intestinal homeostasis, and basic functions performed by the small intestine need to be tightly preserved to maintain health. Little is known about the direct impact of high-fat (HF) diet on small-intestinal mucosal defenses and spatial distribution of the microbiota during the early phase of its administration. We observed that only 30 d after HF diet initiation, the intervillous zone of the ileum-which is usually described as free of bacteria-became occupied by a dense microbiota. In addition to affecting its spatial distribution, HF diet also drastically affected microbiota composition with a profile characterized by the expansion of Firmicutes (appearance of Erysipelotrichi), Proteobacteria (Desulfovibrionales) and Verrucomicrobia, and decrease of Bacteroidetes (family S24-7) and Candidatus arthromitus A decrease in antimicrobial peptide expression was predominantly observed in the ileum where bacterial density appeared highest. In addition, HF diet increased intestinal permeability and decreased cystic fibrosis transmembrane conductance regulator (Cftr) and the Na-K-2Cl cotransporter 1 (Nkcc1) gene and protein expressions, leading to a decrease in ileal secretion of chloride, likely responsible for massive alteration in mucus phenotype. This complex phenotype triggered by HF diet at the interface between the microbiota and the mucosal surface was reversed when the diet was switched back to standard composition or when mice were treated for 1 wk with rosiglitazone, a specific agonist of peroxisome proliferator-activated receptor-γ (PPAR-γ). Moreover, weaker expression of antimicrobial peptide-encoding genes and intervillous bacterial colonization were observed in Ppar-γ-deficient mice, highlighting the major role of lipids in modulation of mucosal immune defenses.


Sujet(s)
Alimentation riche en graisse , Microbiome gastro-intestinal , Intestin grêle/microbiologie , Intestin grêle/physiologie , Récepteur PPAR gamma/métabolisme , Transduction du signal , Animaux , Peptides antimicrobiens cationiques/génétique , Peptides antimicrobiens cationiques/métabolisme , Caecum/microbiologie , Chlorures/métabolisme , Protéine CFTR/métabolisme , Fèces/microbiologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Intestin grêle/effets des médicaments et des substances chimiques , Microdissection au laser , Mâle , Souris de lignée C57BL , Mucus/métabolisme , Récepteur PPAR gamma/génétique , Phénotype , Rosiglitazone , Transduction du signal/effets des médicaments et des substances chimiques , Thiazolidinediones/pharmacologie
2.
Gastroenterology ; 150(2): 454-64.e9, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26481855

RÉSUMÉ

BACKGROUND & AIMS: Bariatric procedures, such as Roux-en-Y gastric bypass (RYGB) or vertical sleeve gastrectomy (VSG), are the most effective approaches to resolve type 2 diabetes in obese individuals. Alimentary glucose absorption and intestinal disposal of blood glucose have not been directly compared between individuals or animals that underwent RYGB vs VSG. We evaluated in rats and humans how the gut epithelium adapts after surgery and the consequences on alimentary glucose absorption and intestinal disposal of blood glucose. METHODS: Obese male rats underwent RYGB, VSG, or sham (control) operations. We collected intestine segments from all rats; we performed histologic analyses and measured levels of messenger RNAs encoding the sugar transporters SGLT1, GLUT1, GLUT2, GLUT3, GLUT4, and GLUT5. Glucose transport and consumption were assayed using ex vivo jejunal loops. Histologic analyses were also performed on Roux limb sections from patients who underwent RYGB 1-5 years after surgery. Roux limb glucose consumption was assayed after surgery by positron emission and computed tomography imaging. RESULTS: In rats and humans that underwent RYGB, the Roux limb became hyperplasic, with an increased number of incretin-producing cells compared with the corresponding jejunal segment of controls. Furthermore, expression of sugar transporters and hypoxia-related genes increased and the nonintestinal glucose transporter GLUT1 appeared at the basolateral membrane of enterocytes. Ingested and circulating glucose was trapped within the intestinal epithelial cells of rats and humans that underwent RYGB. By contrast, there was no hyperplasia of the intestine after VSG, but the intestinal absorption of alimentary glucose was reduced and density of endocrine cells secreting glucagon-like peptide-1 increased. CONCLUSIONS: The intestine adapts differently to RYGB vs VSG. RYGB increases intestinal glucose disposal and VSG delays glucose absorption; both contribute to observed improvements in glycemia.


Sujet(s)
Glycémie/métabolisme , Gastrectomie/méthodes , Dérivation gastrique , Absorption intestinale , Muqueuse intestinale/métabolisme , Jéjunum/métabolisme , Obésité/chirurgie , Adaptation physiologique , Adulte , Animaux , Modèles animaux de maladie humaine , Glucagon-like peptide 1/métabolisme , Transporteurs de glucose par diffusion facilitée/génétique , Transporteurs de glucose par diffusion facilitée/métabolisme , Humains , Hyperplasie , Muqueuse intestinale/anatomopathologie , Jéjunum/anatomopathologie , Mâle , Adulte d'âge moyen , Tomographie par émission de positons , ARN messager/métabolisme , Rats , Études rétrospectives , Facteurs temps , Tomodensitométrie
3.
J Nutr ; 145(8): 1754-62, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26108543

RÉSUMÉ

BACKGROUND: High-fat diets induce intestinal barrier alterations and promote intestinal diseases. Little is known about the effects of long-chain fatty acids (LCFAs) on mucin 2 (MUC2) production by goblet cells, which are crucial for intestinal protection. OBJECTIVE: We investigated the effects of LCFAs on the differentiation of colonic goblet cells, MUC2 expression, and colonic barrier function. METHODS: Upon reaching confluence, human colonic mucus-secreting HT29-MTX cells were stimulated (21 d) with a saturated LCFA (palmitic or stearic acid), a monounsaturated LCFA (oleic acid), or a polyunsaturated LCFA (linoleic, γ-linolenic, α-linolenic, or eicosapentaenoic acid). In addition, rat pups underwent oral administration of oil (palm, rapeseed, or sunflower oil) or water (10 µL/g body weight, postnatal days 10-15). Subsequently, colon goblet cells were studied by Western blotting, reverse transcriptase-quantitative polymerase chain reaction, and immunohistochemistry and colonic transmucosal electrical resistance was measured by using Ussing chambers. RESULTS: In vitro, palmitic acid enhanced MUC2 production (140% of control) and hepatocyte nuclear factor 4α expression, whereas oleic, linoleic, γ-linolenic, α-linolenic, and eicosapentaenoic acids reduced MUC2 expression (at least -50% of control). All unsaturated LCFAs decreased the expression of human atonal homolog 1, a transcription factor controlling goblet cell differentiation (at least -31% vs. control). In vivo, rats fed palm oil had higher palmitic acid concentrations (3-fold) in their colonic contents and increased mucus granule surfaces in their goblet cells (>2-fold) than did all other groups. Palm oil also increased colonic transmucosal electrical resistance (245% of control), yet had no effect on occludin and zonula occludens-1 expression. In contrast, sunflower and rapeseed oils decreased goblet cell number when compared with control (at least -10%) and palm oil (at least -14%) groups. CONCLUSIONS: Palm oil in rat pups and palmitic acid in HT29-MTX cells increase the production of MUC2 and strengthen the intestinal barrier. In contrast, unsaturated LCFAs decrease MUC2 expression. These data should be taken into account in the context of preventive or therapeutic nutritional programs.


Sujet(s)
Côlon/cytologie , Matières grasses alimentaires/pharmacologie , Acides gras insaturés/pharmacologie , Acides gras/pharmacologie , Cellules caliciformes/effets des médicaments et des substances chimiques , Aliment pour animaux/analyse , Animaux , Régime alimentaire , Matières grasses alimentaires/administration et posologie , Acides gras/administration et posologie , Acides gras insaturés/administration et posologie , Cellules caliciformes/métabolisme , Cellules HT29 , Humains , Mucine-5AC/génétique , Mucine-5AC/métabolisme , Mucine-2/génétique , Mucine-2/métabolisme , Huiles végétales/administration et posologie , Huiles végétales/composition chimique , Rats , Rat Wistar
4.
J. physiol. biochem ; 71(2): 311-317, jun. 2015.
Article de Anglais | IBECS | ID: ibc-140538

RÉSUMÉ

Leptin is secreted into the digestive tract and contributes to the absorption of dietary molecules by regulating transporters activity. Here, we studied the effect of luminal leptin on the intestinal transport of L-glutamate, an important component of human diet. We examined the effect of leptin on L-glutamate uptake in rat intestine in vitro measuring glutamate-induced short-circuit current (Isc) in Ussing chambers and L-[3H (U)]-glutamate uptake in jejunal everted rings. Glutamate-induced Isc was only observed in Na+-free conditions. This Isc was concentration (1–60 mmol L−1) and pH dependent. Luminal leptin increased glutamate Isc (∼100 %). Dose-response curve showed a biphasic pattern, with maximal stimulations observed at 10−13 and 10−10 mmol L−1, that were sensitive to leptin receptor antagonist. In everted rings, two glutamate transport mechanisms were distinguished: a Na+-dependent, H+-independent, that was inhibited by leptin (∼20 %), and a Na+-independent but H+-dependent, that was enhanced by leptin (∼20 %), in line with data obtained in Ussing chambers. Altogether, these data reveal original non-monotonic effect of luminal leptin in the intestine and demonstrate a new role for this hormone in the modulation of L-glutamate transport, showing that luminal active gut peptides can influence absorption of amino acids


Sujet(s)
Animaux , Rats , Transporteurs vésiculaires du glutamate , Leptine/pharmacocinétique , Intestins/physiopathologie
5.
J Physiol Biochem ; 71(2): 311-7, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25935421

RÉSUMÉ

Leptin is secreted into the digestive tract and contributes to the absorption of dietary molecules by regulating transporters activity. Here, we studied the effect of luminal leptin on the intestinal transport of L-glutamate, an important component of human diet. We examined the effect of leptin on L-glutamate uptake in rat intestine in vitro measuring glutamate-induced short-circuit current (Isc) in Ussing chambers and L-[(3)H (U)]-glutamate uptake in jejunal everted rings. Glutamate-induced Isc was only observed in Na(+)-free conditions. This Isc was concentration (1-60 mmol L(-1)) and pH dependent. Luminal leptin increased glutamate Isc (∼100 %). Dose-response curve showed a biphasic pattern, with maximal stimulations observed at 10(-13) and 10(-10) mmol L(-1), that were sensitive to leptin receptor antagonist. In everted rings, two glutamate transport mechanisms were distinguished: a Na(+)-dependent, H(+)-independent, that was inhibited by leptin (∼20 %), and a Na(+)-independent but H(+)-dependent, that was enhanced by leptin (∼20 %), in line with data obtained in Ussing chambers. Altogether, these data reveal original non-monotonic effect of luminal leptin in the intestine and demonstrate a new role for this hormone in the modulation of L-glutamate transport, showing that luminal active gut peptides can influence absorption of amino acids.


Sujet(s)
Acide glutamique/métabolisme , Intestin grêle/métabolisme , Leptine/métabolisme , Animaux , Transport biologique/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Acide glutamique/pharmacocinétique , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Intestin grêle/effets des médicaments et des substances chimiques , Leptine/antagonistes et inhibiteurs , Leptine/pharmacologie , Mâle , Techniques de culture d'organes/instrumentation , Techniques de culture d'organes/méthodes , Rat Wistar , Sodium/métabolisme
6.
PLoS One ; 10(3): e0121414, 2015.
Article de Anglais | MEDLINE | ID: mdl-25822172

RÉSUMÉ

Whereas the remodeling of intestinal mucosa after bariatric surgeries has been the matter of numerous studies to our knowledge, very few reported on the remodeling of the residual gastric mucosa. In this study, we analyzed remodeling of gastric mucosa after Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) in rats. Diet-induced obese rats were subjected to RYGB, VSG or sham surgical procedures. All animals were assessed for food intake, body-weight, fasting blood, metabolites and hormones profiling, as well as insulin and glucose tolerance tests before and up to 5 weeks post-surgery. Remodeling of gastric tissues was analyzed by routine histology and immunohistochemistry studies, and qRT-PCR analyses of ghrelin and gastrin mRNA levels. In obese rats with impaired glucose tolerance, VSG and RYGB caused substantial weight loss and rats greatly improved their oral glucose tolerance. The remaining gastric mucosa after VSG and gastric pouch (GP) after RYGB revealed a hyperplasia of the mucous neck cells that displayed a strong immunoreactivity for parietal cell H+/K+-ATPase. Ghrelin mRNA levels were reduced by 2-fold in remaining fundic mucosa after VSG and 10-fold in GP after RYGB. In the antrum, gastrin mRNA levels were reduced after VSG in line with the reduced number of gastrin positive cells. This study reports novel and important observations dealing with the remaining gastric mucosa after RYGB and VSG. The data demonstrate, for the first time, a hyperplasia of the mucous neck cells, a transit cell population of the stomach bearing differentiating capacities into zymogenic and peptic cells.


Sujet(s)
Muqueuse gastrique/physiopathologie , Obésité/physiopathologie , Animaux , Glycémie/physiologie , Poids/physiologie , Régime alimentaire/méthodes , Consommation alimentaire/physiologie , Gastrectomie/méthodes , Dérivation gastrique/méthodes , Muqueuse gastrique/métabolisme , Ghréline/métabolisme , Hyperglycémie provoquée/méthodes , H(+)-K(+)-Exchanging ATPase/métabolisme , Insuline/métabolisme , Mâle , Obésité/sang , Obésité/métabolisme , Obésité/chirurgie , ARN messager/métabolisme , Rats , Rat Wistar , Perte de poids/physiologie
7.
ISME J ; 9(1): 46-58, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25012905

RÉSUMÉ

We investigated the effects of early colonizing bacteria on the colonic epithelium. We isolated dominant bacteria, Escherichia coli, Enterococcus faecalis, Lactobacillus intestinalis, Clostridium innocuum and a novel Fusobacterium spp., from the intestinal contents of conventional suckling rats and transferred them in different combinations into germfree (GF) adult rats. Animals were investigated after various times up to 21 days. Proliferative cell markers (Ki67, proliferating cell nuclear antigen, phospho-histone H3, cyclin A) were higher in rats monocolonized with E. coli than in GF at all time points, but not in rats monocolonized with E. faecalis. The mucin content of goblet cells declined shortly after E. coli administration whereas the mucus layer doubled in thickness. Fluorescence in situ hybridization analyses revealed that E. coli resides in this mucus layer. The epithelial mucin content progressively returned to baseline, following an increase in KLF4 and in the cell cycle arrest-related proteins p21(CIP1) and p27(KIP1). Markers of colonic differentiated cells involved in electrolyte (carbonic anhydrase II and slc26A3) and water (aquaglyceroporin3 (aqp3)) transport, and secretory responses to carbachol were modulated after E. coli inoculation suggesting that ion transport dynamics were also affected. The colonic responses to simplified microbiotas differed substantially according to whether or not E. coli was combined with the other four bacteria. Thus, proliferation markers increased substantially when E. coli was in the mix, but very much less when it was absent. This work demonstrates that a pioneer strain of E. coli elicits sequential epithelial remodeling affecting the structure, mucus layer and ionic movements and suggests this can result in a microbiota-compliant state.


Sujet(s)
Côlon/microbiologie , Escherichia coli/physiologie , Muqueuse intestinale/microbiologie , Animaux , Protéines du cycle cellulaire/métabolisme , Différenciation cellulaire , Prolifération cellulaire , Côlon/cytologie , Côlon/métabolisme , Homéostasie , Muqueuse intestinale/cytologie , Muqueuse intestinale/métabolisme , Facteur-4 de type Kruppel , Mâle , Mucines/métabolisme , Rats , Rats de lignée F344
8.
PLoS One ; 9(7): e101669, 2014.
Article de Anglais | MEDLINE | ID: mdl-25014110

RÉSUMÉ

Ulcerative colitis (UC) is a chronic inflammatory bowel disease affecting the rectum which progressively extents. Its etiology remains unknown and the number of treatments available is limited. Studies of UC patients have identified an unbalanced endoplasmic reticulum (ER) stress in the non-inflamed colonic mucosa. Animal models with impaired ER stress are sensitive to intestinal inflammation, suggesting that an unbalanced ER stress could cause inflammation. However, there are no ER stress-regulating strategies proposed in the management of UC partly because of the lack of relevant preclinical model mimicking the disease. Here we generated the IL10/Nox1dKO mouse model which combines immune dysfunction (IL-10 deficiency) and abnormal epithelium (NADPH oxidase 1 (Nox1) deficiency) and spontaneously develops a UC-like phenotype with similar complications (colorectal cancer) than UC. Our data identified an unanticipated combined role of IL10 and Nox1 in the fine-tuning of ER stress responses in goblet cells. As in humans, the ER stress was unbalanced in mice with decreased eIF2α phosphorylation preceding inflammation. In IL10/Nox1dKO mice, salubrinal preserved eIF2α phosphorylation through inhibition of the regulatory subunit of the protein phosphatase 1 PP1R15A/GADD34 and prevented colitis. Thus, this new experimental model highlighted the central role of epithelial ER stress abnormalities in the development of colitis and defined the defective eIF2α pathway as a key pathophysiological target for UC. Therefore, specific regulators able to restore the defective eIF2α pathway could lead to the molecular remission needed to treat UC.


Sujet(s)
Rectocolite hémorragique/étiologie , Modèles animaux de maladie humaine , Stress du réticulum endoplasmique , Inflammation/étiologie , Interleukine-10/physiologie , NADH, NADPH oxidoreductases/physiologie , Animaux , Technique de Western , Études cas-témoins , Prolifération cellulaire , Cellules cultivées , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/anatomopathologie , Côlon/immunologie , Côlon/métabolisme , Côlon/anatomopathologie , Réticulum endoplasmique/métabolisme , Réticulum endoplasmique/anatomopathologie , Femelle , Cytométrie en flux , Technique d'immunofluorescence , Humains , Techniques immunoenzymatiques , Inflammation/métabolisme , Inflammation/anatomopathologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , NADPH Oxidase 1 , Phosphorylation , Protein Phosphatase 1/génétique , Protein Phosphatase 1/métabolisme , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , RT-PCR , Réponse aux protéines mal repliées
9.
FASEB J ; 28(9): 4100-10, 2014 Sep.
Article de Anglais | MEDLINE | ID: mdl-24928195

RÉSUMÉ

The importance of B-isoform of leptin receptor (LEPR-B) signaling in the hypothalamus, pancreas, or liver has been well characterized, but in the intestine, a unique site of entry for dietary nutrition into the body, it has been relatively ignored. To address this question, we characterized a mouse model deficient for LEPR-B specifically in intestinal epithelial cells (IECs). (IEC)LEPR-B-knockout (KO) and wild-type (WT) mice were generated by Cre-Lox strategy and fed a normal or high-fat diet (HFD). The analyses of the animals involved histology and immunohistochemistry of intestinal mucosa, indirect calorimetric measurements, whole-body composition, and expression and activities of nutrient transporters. (IEC)LEPR-B-KO mice exhibited a 2-fold increase in length of jejunal villi and have normal growth on a normal diet but were less susceptible (P<0.01) to HFD-induced obesity. No differences occurred in energy intake and expenditure between (IEC)LEPR-B-WT and -KO mice, but (IEC)LEPR-B-KO mice fed an HFD showed increased excreted fats (P<0.05). Activities of the Na(+)/glucose cotransporter SGLT-1 and GLUT2 were unaffected in LEPR-B-KO jejunum, while GLUT5-mediated fructose transport and PepT1-mediated peptide transport were substantially reduced (P<0.01). These data demonstrate that intestinal LEPR-B signaling is important for the onset of diet-induced obesity. They suggest that intestinal LEPR-B could be a potential per os target for prevention against obesity.


Sujet(s)
Alimentation riche en graisse/effets indésirables , Transporteurs de glucose par diffusion facilitée/métabolisme , Transporteur de glucose de type 2/métabolisme , Muqueuse intestinale/métabolisme , Obésité/étiologie , Récepteurs à la leptine/physiologie , Symporteurs/métabolisme , Animaux , Technique de Western , Composition corporelle , Poids , Prolifération cellulaire , Cellules cultivées , Ration calorique , Femelle , Transporteurs de glucose par diffusion facilitée/génétique , Transporteur de glucose de type 2/génétique , Transporteur de glucose de type 5 , Techniques immunoenzymatiques , Muqueuse intestinale/anatomopathologie , Leptine/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Transporteur-1 de peptides , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , RT-PCR , Symporteurs/génétique
10.
J Nutr Biochem ; 25(5): 557-64, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-24656388

RÉSUMÉ

Green tea containing polyphenols exerts antidiabetic and antiobesity effects, but the mechanisms involved are not fully understood. In this study, we first analyzed and compared polyphenol compounds [epigallocatechin gallate (EGCG), epigallocatechin (EGC)] in decoction of green tea leaves versus usual green tea extracts. Second, the effects of acute (30 min) or chronic (6 weeks) oral administration of green tea decoction (GTD) on intestinal glucose absorption were studied in vitro in Ussing chamber, ex vivo using isolated jejunal loops and in vivo through glucose tolerance tests. Finally, we explore in rat model fed normal or high-fat diet the effects of GTD on body weight, blood parameters and on the relative expression of glucose transporters SGLT-1, GLUT2 and GLUT4. GTD cooked for 15 min contained the highest amounts of phenolic compounds. In fasted rats, acute administration of GTD inhibited SGLT-1 activity, increased GLUT2 activity and improved glucose tolerance. Similarly to GTD, acute administration of synthetic phenolic compounds (2/3 EGCG+1/3 EGC) inhibited SGLT-1 activity. Chronic administration of GTD in rat fed high-fat diet reduced body weight gain, circulating triglycerides and cholesterol and improved glucose tolerance. GTD-treated rats for 6 weeks display significantly reduced SGLT-1 and increased GLUT2 mRNA levels in the jejunum mucosa. Moreover, adipose tissue GLUT4 mRNA levels were increased. These results indicate that GTD, a traditional beverage rich in EGCG and EGC reduces intestinal SGLT-1/GLUT2 ratio, a hallmark of regulation of glucose absorption in enterocyte, and enhances adipose GLUT4 providing new insights in its possible role in the control of glucose homeostasis.


Sujet(s)
Alimentation riche en graisse/effets indésirables , Thé , Prise de poids/effets des médicaments et des substances chimiques , Administration par voie orale , Animaux , Caféine/analyse , Caféine/pharmacologie , Catéchine/analogues et dérivés , Catéchine/analyse , Catéchine/pharmacologie , Glucose/métabolisme , Hyperglycémie provoquée , Transporteur de glucose de type 2/génétique , Transporteur de glucose de type 2/métabolisme , Transporteur de glucose de type 4/génétique , Lipides/sang , Mâle , Polyphénols/analyse , Polyphénols/pharmacologie , Transport des protéines/effets des médicaments et des substances chimiques , Rat Wistar , Transporteur-1 sodium-glucose/antagonistes et inhibiteurs , Transporteur-1 sodium-glucose/génétique , Transporteur-1 sodium-glucose/métabolisme , Thé/composition chimique
11.
Gastroenterology ; 144(4): 771-80, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23313268

RÉSUMÉ

BACKGROUND & AIMS: Glucose is absorbed into intestine cells via the sodium glucose transporter 1 (SGLT-1) and glucose transporter 2 (GLUT2); various peptides and hormones control this process. Apelin is a peptide that regulates glucose homeostasis and is produced by proximal digestive cells; we studied whether glucose modulates apelin secretion by enterocytes and the effects of apelin on intestinal glucose absorption. METHODS: We characterized glucose-related luminal apelin secretion in vivo and ex vivo by mass spectroscopy and immunologic techniques. The effects of apelin on (14)C-labeled glucose transport were determined in jejunal loops and in mice following apelin gavage. We determined levels of GLUT2 and SGLT-1 proteins and phosphorylation of AMPKα2 by immunoblotting. The net effect of apelin on intestinal glucose transepithelial transport was determined in mice. RESULTS: Glucose stimulated luminal secretion of the pyroglutaminated apelin-13 isoform ([Pyr-1]-apelin-13) in the small intestine of mice. Apelin increased specific glucose flux through the gastric epithelial barrier in jejunal loops and in vivo following oral glucose administration. Conversely, pharmacologic apelin blockade in the intestine reduced the increased glycemia that occurs following oral glucose administration. Apelin activity was associated with phosphorylation of AMPKα2 and a rapid increase of the GLUT2/SGLT-1 protein ratio in the brush border membrane. CONCLUSIONS: Glucose amplifies its own transport from the intestinal lumen to the bloodstream by increasing luminal apelin secretion. In the lumen, active apelin regulates carbohydrate flux through enterocytes by promoting AMPKα2 phosphorylation and modifying the ratio of SGLT-1:GLUT2. The glucose-apelin cycle might be pharmacologically handled to regulate glucose absorption and assess better control of glucose homeostasis.


Sujet(s)
Glucides/pharmacocinétique , Glucose/métabolisme , Protéines et peptides de signalisation intercellulaire/métabolisme , Absorption intestinale/effets des médicaments et des substances chimiques , Absorption intestinale/physiologie , Analyse de variance , Animaux , Transport biologique/effets des médicaments et des substances chimiques , Transport biologique/physiologie , Technique de Western , Chromatographie en phase liquide/méthodes , Modèles animaux de maladie humaine , Glucose/pharmacologie , Transporteur de glucose de type 2/métabolisme , Immunohistochimie , Mâle , Spectrométrie de masse , Souris , Souris de lignée C57BL , Microscopie confocale , Répartition aléatoire , Valeurs de référence , Transporteur-1 sodium-glucose/métabolisme
12.
Inflamm Bowel Dis ; 17(3): 747-57, 2011 Mar.
Article de Anglais | MEDLINE | ID: mdl-20684014

RÉSUMÉ

BACKGROUND: In human pathology, the "creeping fat" (CF) of the mesentery is unique to Crohn's disease (CD). CF is usually referred to as an ectopic extension of mesenteric adipose tissue (MAT). However, since no animal model developing CF has ever been established, very little is known about this type of fat-depot expansion and its role in the development of the disease. METHODS: We developed and standardized an experimental protocol in mice that reproducibly induces CF development when a severe colonic inflammation is obtained by intracolonic instillation of DNBS. RESULTS: Macro-microscopic observations revealed a fatty appearance of CF. Yet when compared to MAT from the same animals, CF contains very little triglycerides, few adipocytes, and we observed a very low expression and protein levels of both adipose markers (hormone-sensitive lipase, perilipin) and adipocytokines (leptin, adiponectin). The decreased expression of perilipin in CF was also observed by immunohistochemistry. Conversely, the expression of proinflammatory and fibrous markers (Pref-1) was much higher in CF than in MAT. These observations were fully consistent with those made on CF recovered from five CD patients and compared with subcutaneous and mesenteric fat from the same patients. CONCLUSIONS: Altogether, this work reports an original experimental mice model of CF. In this model we establish for the first time that CF only occurs in severe colonic inflammation and shows an inflammatory, fibrous but not an adipose pattern.


Sujet(s)
Tissu adipeux/anatomopathologie , Colite/anatomopathologie , Maladie de Crohn/anatomopathologie , Mésentère , Tissu adipeux/métabolisme , Animaux , Technique de Western , Poids , Colite/induit chimiquement , Colite/métabolisme , Maladie de Crohn/métabolisme , 1-Fluoro-2,4-dinitro-benzène/analogues et dérivés , 1-Fluoro-2,4-dinitro-benzène/toxicité , Test ELISA , Humains , Techniques immunoenzymatiques , Lipides , Mâle , Souris , Souris de lignée BALB C , Myeloperoxidase/métabolisme , ARN messager/génétique , RT-PCR
13.
Am J Physiol Gastrointest Liver Physiol ; 299(1): G179-85, 2010 Jul.
Article de Anglais | MEDLINE | ID: mdl-20448142

RÉSUMÉ

L-glutamine is the primary metabolic fuel for enterocytes. Glutamine from the diet is transported into the absorptive cells by two sodium-dependent neutral amino acid transporters present at the apical membrane: ASCT2/SLC1A5 and B(0)AT1/SLC6A19. We have demonstrated that leptin is secreted into the stomach lumen after a meal and modulates the transport of sugars after binding to its receptors located at the brush border of the enterocytes. The present study was designed to address the effect of luminal leptin on Na(+)-dependent glutamine (Gln) transport in rat intestine and identify the transporters involved. We found that 0.2 nM leptin inhibited uptake of Gln and phenylalanine (Phe) (substrate of B(0)AT1) using everted intestinal rings. In Ussing chambers, 10 mM Gln absorption followed as Na(+)-induced short-circuit current was inhibited by leptin in a dose-dependent manner (maximum inhibition at 10 nM; I(C50) = approximately 0.1 nM). Phe absorption was also decreased by leptin. Western blot analysis after 3-min incubation of the intestinal loops with 10 mM Gln, showed marked increase of ASCT2 and B(0)AT1 protein in the brush-border membrane that was reduced by rapid preincubation of the intestinal lumen with 1 nM leptin. Similarly, the increase in ASCT2 and B(0)AT1 gene expression induced by 60-min incubation of the intestine with 10 mM Gln was strongly reduced after a short preincubation period with leptin. Altogether these data demonstrate that, in rat, leptin controls the active Gln entry through reduction of both B(0)AT1 and ASCT2 proteins traffic to the apical plasma membrane and modulation of their gene expression.


Sujet(s)
Système ASC de transport d'acides aminés/métabolisme , Systèmes de transport d'acides aminés neutres/métabolisme , Glutamine/métabolisme , Intestin grêle/métabolisme , Leptine/métabolisme , Système ASC de transport d'acides aminés/génétique , Systèmes de transport d'acides aminés neutres/génétique , Animaux , Transport biologique , Régulation de l'expression des gènes , Techniques in vitro , Muqueuse intestinale/métabolisme , Intestin grêle/effets des médicaments et des substances chimiques , Mâle , Potentiels de membrane , Antigènes mineurs d'histocompatibilité , Peptides/pharmacologie , Phénylalanine/métabolisme , Transport des protéines , ARN messager/métabolisme , Rats , Rat Wistar , Récepteurs à la leptine/antagonistes et inhibiteurs , Récepteurs à la leptine/métabolisme , Protéines recombinantes/métabolisme , Séreuse/métabolisme , Sodium/métabolisme , Facteurs temps
14.
Mol Cell Biol ; 30(11): 2636-50, 2010 Jun.
Article de Anglais | MEDLINE | ID: mdl-20351171

RÉSUMÉ

The homeostatic self-renewal of the colonic epithelium requires coordinated regulation of the canonical Wnt/beta-catenin and Notch signaling pathways to control proliferation and lineage commitment of multipotent stem cells. However, the molecular mechanisms by which the Wnt/beta-catenin and Notch1 pathways interplay in controlling cell proliferation and fate in the colon are poorly understood. Here we show that NADPH oxidase 1 (NOX1), a reactive oxygen species (ROS)-producing oxidase that is highly expressed in colonic epithelial cells, is a pivotal determinant of cell proliferation and fate that integrates Wnt/beta-catenin and Notch1 signals. NOX1-deficient mice reveal a massive conversion of progenitor cells into postmitotic goblet cells at the cost of colonocytes due to the concerted repression of phosphatidylinositol 3-kinase (PI3K)/AKT/Wnt/beta-catenin and Notch1 signaling. This conversion correlates with the following: (i) the redox-dependent activation of the dual phosphatase PTEN, causing the inactivation of the Wnt pathway effector beta-catenin, and (ii) the downregulation of Notch1 signaling that provokes derepression of mouse atonal homolog 1 (Math1) expression. We conclude that NOX1 controls the balance between goblet and absorptive cell types in the colon by coordinately modulating PI3K/AKT/Wnt/beta-catenin and Notch1 signaling. This finding provides the molecular basis for the role of NOX1 in cell proliferation and postmitotic differentiation.


Sujet(s)
Prolifération cellulaire , Côlon/cytologie , Cellules souches multipotentes/physiologie , NADH, NADPH oxidoreductases/métabolisme , Récepteur Notch1/métabolisme , Transduction du signal/physiologie , Protéines de type Wingless/métabolisme , Animaux , Cellules Caco-2 , Cadhérines/métabolisme , Différenciation cellulaire/physiologie , Lignage cellulaire , Côlon/physiologie , Cellules épithéliales/cytologie , Cellules épithéliales/physiologie , Humains , Muqueuse intestinale/cytologie , Souris , Souris de lignée C57BL , Souris knockout , Cellules souches multipotentes/cytologie , NADH, NADPH oxidoreductases/génétique , NADPH Oxidase 1 , Phosphohydrolase PTEN/métabolisme , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Espèces réactives de l'oxygène/métabolisme , Récepteur Notch1/génétique , Protéines de type Wingless/génétique , bêta-Caténine/métabolisme
15.
PLoS One ; 4(11): e7935, 2009 Nov 30.
Article de Anglais | MEDLINE | ID: mdl-19956534

RÉSUMÉ

BACKGROUND AND AIMS: The small intestine is the major site of absorption of dietary sugars. The rate at which they enter and exit the intestine has a major effect on blood glucose homeostasis. In this study, we determine the effects of luminal leptin on activity/expression of GLUT2 and GLUT5 transporters in response to sugars intake and analyse their physiological consequences. METHODOLOGY: Wistar rats, wild type and AMPKalpha(2) (-/-) mice were used. In vitro and in vivo isolated jejunal loops were used to quantify transport of fructose and galactose in the absence and the presence of leptin. The effects of fructose and galactose on gastric leptin release were determined. The effects of leptin given orally without or with fructose were determined on the expression of GLUT2/5, on some gluconeogenesis and lipogenic enzymes in the intestine and the liver. PRINCIPAL FINDINGS: First, in vitro luminal leptin activating its receptors coupled to PKCbetaII and AMPKalpha, increased insertion of GLUT2/5 into the brush-border membrane leading to enhanced galactose and fructose transport. Second in vivo, oral fructose but not galactose induced in mice a rapid and potent release of gastric leptin in gastric juice without significant changes in plasma leptin levels. Moreover, leptin given orally at a dose reproducing comparable levels to those induced by fructose, stimulated GLUT5-fructose transport, and potentiated fructose-induced: i) increase in blood glucose and mRNA levels of key gluconeogenesis enzymes; ii) increase in blood triglycerides and reduction of mRNA levels of intestinal and hepatic Fasting-induced adipocyte factor (Fiaf) and iii) increase in SREBP-1c, ACC-1, FAS mRNA levels and dephosphorylation/activation of ACC-1 in liver. CONCLUSION/SIGNIFICANCE: These data identify for the first time a positive regulatory control loop between gut leptin and fructose in which fructose triggers release of gastric leptin which, in turn, up-regulates GLUT5 and concurrently modulates metabolic functions in the liver. This loop appears to be a new mechanism (possibly pathogenic) by which fructose consumption rapidly becomes highly lipogenic and deleterious.


Sujet(s)
Transporteurs de glucose par diffusion facilitée/métabolisme , Transporteur de glucose de type 2/métabolisme , Transporteur de glucose de type 5/métabolisme , Leptine/métabolisme , Foie/métabolisme , Animaux , Glycémie/métabolisme , Fructose/métabolisme , Néoglucogenèse , Homéostasie , Mâle , Souris , Souris de lignée C57BL , Rats , Rat Wistar
16.
Diabetes ; 58(9): 2032-8, 2009 Sep.
Article de Anglais | MEDLINE | ID: mdl-19502416

RÉSUMÉ

OBJECTIVE: An increased expression of RELM-beta (resistin-like molecule-beta), a gut-derived hormone, is observed in animal models of insulin resistance/obesity and intestinal inflammation. Intestinal sugar absorption is modulated by dietary environment and hormones/cytokines. The aim of this study was to investigate the effect of RELM-beta on intestinal glucose absorption. RESEARCH DESIGN AND METHODS: Oral glucose tolerance test was performed in mice and rats in the presence and the absence of RELM-beta. The RELM-beta action on glucose transport in rat jejunal sacs, everted rings, and mucosal strips was explored as well as downstream kinases modulating SGLT-1 and GLUT2 glucose transporters. RESULTS: Oral glucose tolerance test carried out in rodents showed that oral administration of RELM-beta increased glycemia. Studies in rat jejunal tissue indicated that mucosal RELM-beta promoted absorption of glucose from the gut lumen. RELM-beta had no effect on paracellular mannitol transport, suggesting a transporter-mediated transcellular mechanism. In studies with jejunal mucosa mounted in Ussing chamber, luminal RELM-beta inhibited SGLT-1 activity in line with a diminished SGLT-1 abundance in brush border membranes (BBMs). Further, the potentiating effect of RELM-beta on jejunal glucose uptake was associated with an increased abundance of GLUT2 at BBMs. The effects of RELM-beta were associated with an increased amount of protein kinase C betaII in BBMs and an increased phosphorylation of AMP-activated protein kinase (AMPK). CONCLUSIONS: The regulation of SGLT-1 and GLUT2 by RELM-beta expands the role of gut hormones in short-term AMPK/protein kinase C mediated control of energy balance.


Sujet(s)
Transporteur de glucose de type 2/métabolisme , Glucose/pharmacocinétique , Hormones de sécrétion ectopique/métabolisme , Jéjunum/métabolisme , Transporteur-1 sodium-glucose/métabolisme , AMP-Activated Protein Kinases/métabolisme , Animaux , Transport biologique actif/effets des médicaments et des substances chimiques , Transport biologique actif/physiologie , Hyperglycémie provoquée , Hormones de sécrétion ectopique/pharmacologie , Protéines et peptides de signalisation intercellulaire , Absorption intestinale/effets des médicaments et des substances chimiques , Absorption intestinale/physiologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Jéjunum/effets des médicaments et des substances chimiques , Mâle , Souris , Souris de lignée C57BL , Phosphorylation/effets des médicaments et des substances chimiques , Phosphorylation/physiologie , Protéine kinase C/métabolisme , Protein kinase C beta , Rats , Rat Wistar , Protéines recombinantes/pharmacologie , Sodium/métabolisme
17.
Biol Cell ; 101(10): 573-86, 2009 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-19392661

RÉSUMÉ

BACKGROUND INFORMATION: The TSPO (18 kDa translocator protein) is a mitochondrial transmembrane protein involved in cholesterol transport in organs that synthesize steroids and bile salts. Different natural and synthetic high-affinity TSPO ligands have been characterized through their ability to stimulate cholesterol transport, but also to stimulate other physiological functions including cell proliferation, apoptosis and calcium-dependent transepithelial ion secretion. Here, we investigate the localization and functions of TSPO in the small intestine. RESULTS: TSPO was present in enterocyte mitochondria but not in rat intestinal goblet cells. Enterocyte cytoplasm also contained the endogenous TSPO ligand, polypeptide DBI (diazepam-binding inhibitor). Whereas intestinal TSPO had high affinity for the synthetic ligand PK 11195, the pharmacological profile of TSPO in the duodenum was distinct from the jejunum and ileum. Specifically, benzodiazepine Ro5-4864 and protoporphyrin IX showed 5-13-fold lower affinity for duodenal TSPO. The mRNA and protein ratios of TSPO to other mitochondrial membrane proteins VDAC (voltage-dependent anion channel) and ANT (adenine nucleotide transporter) were significantly different. PK 11195 stimulated calcium-dependent chloride secretion in the duodenum and calcium-dependent chloride absorption in the ileum, but did not affect jejunum ion transport. CONCLUSIONS: The functional differences in subpopulations of TSPO in different regions of the intestine could be related to structural organization of mitochondrial protein complexes that mediate the ability of TSPO to modulate either chloride secretion or absorption in the duodenum and ileum respectively.


Sujet(s)
Protéines de transport/métabolisme , Entérocytes/métabolisme , Intestin grêle/métabolisme , Mitochondries/métabolisme , Récepteurs GABA-A/métabolisme , Glandes surrénales/métabolisme , Animaux , Protéines de transport/composition chimique , Protéines de transport/génétique , Inhibiteur de la liaison au diazépam/composition chimique , Inhibiteur de la liaison au diazépam/métabolisme , Duodénum/composition chimique , Duodénum/cytologie , Duodénum/métabolisme , Entérocytes/composition chimique , Entérocytes/ultrastructure , Antagonistes du récepteur GABA-A , Iléum/composition chimique , Iléum/cytologie , Iléum/métabolisme , Concentration inhibitrice 50 , Muqueuse intestinale/composition chimique , Muqueuse intestinale/métabolisme , Muqueuse intestinale/ultrastructure , Intestin grêle/composition chimique , Intestin grêle/cytologie , Transport des ions/physiologie , Isoquinoléines/métabolisme , Isoquinoléines/pharmacologie , Jéjunum/composition chimique , Jéjunum/cytologie , Jéjunum/métabolisme , Ligands , Mâle , Membranes/métabolisme , Mitochondries/composition chimique , Mitochondries/ultrastructure , Protéines de transport de la membrane mitochondriale/antagonistes et inhibiteurs , Pore de transition de perméabilité mitochondriale , Protéines mitochondriales/composition chimique , Protéines mitochondriales/génétique , Protéines mitochondriales/métabolisme , Myocarde , Spécificité d'organe , Dosage par compétition , Rats , Rat Wistar/métabolisme , Récepteurs GABA-A/composition chimique , Récepteurs GABA-A/génétique , Glande submandibulaire/métabolisme
18.
J Ethnopharmacol ; 121(3): 419-24, 2009 Jan 30.
Article de Anglais | MEDLINE | ID: mdl-19061948

RÉSUMÉ

AIM OF THE STUDY: Nigella sativa L. (Ranunculaceae) seeds have been used traditionally for centuries, notably for treating diabetes. MATERIALS AND METHODS: We studied the effects of the crude aqueous extract of Nigella sativa seeds on intestinal glucose absorption in vitro using a short-circuit current technique and in vivo using an oral glucose tolerance test. RESULTS: The aqueous extract of Nigella sativa (0.1 pg/ml to 100 ng/ml) exerted dose-dependent inhibition of sodium-dependent glucose transport across isolated rat jejunum. Maximal inhibition exceeded 80% and IC50 was close to 10 pg/ml. An oral glucose tolerance test was carried out in rats after the initial dose and after a 6-week treatment of Nigella sativa (2 g/(kg day)), and compared to metformin (300 mg/(kg day)). Chronic Nigella sativa treatment improved glucose tolerance as efficiently as metformin. Nigella sativa and metformin also reduced body weight without any toxic effect. CONCLUSIONS: To our knowledge, this is the first demonstration that Nigella sativa directly inhibits the electrogenic intestinal absorption of glucose in vitro. Together with the observed improvement of glucose tolerance and body weight in rats after chronic oral administration in vivo, these effects further validate the traditional use of Nigella sativa seeds against diabetes.


Sujet(s)
Poids/effets des médicaments et des substances chimiques , Glucose/métabolisme , Hypoglycémiants/pharmacologie , Absorption intestinale/effets des médicaments et des substances chimiques , Nigella sativa , Extraits de plantes/pharmacologie , Animaux , Relation dose-effet des médicaments , Femelle , Hyperglycémie provoquée , Mâle , Metformine/pharmacologie , Rats , Rat Sprague-Dawley , Graines
19.
Diabetes ; 56(10): 2494-500, 2007 Oct.
Article de Anglais | MEDLINE | ID: mdl-17626888

RÉSUMÉ

OBJECTIVE: Orexins are neuropeptides involved in energy homeostasis. We investigated the effect of orexin A (OxA) and orexin B (OxB) on intestinal glucose transport in the rat. RESEARCH DESIGN AND METHODS AND RESULTS: Injection of orexins led to a decrease in the blood glucose level in oral glucose tolerance tests (OGTTs). Effects of orexins on glucose entry were analyzed in Ussing chambers using the Na(+)-dependent increase in short-circuit current (Isc) to quantify jejunal glucose transport. The rapid and marked increase in Isc induced by luminal glucose was inhibited by 10 nmol/l OxA or OxB (53 and 59%, respectively). Response curves to OxA and OxB were not significantly different with half-maximal inhibitory concentrations at 0.9 and 0.4 nmol/l, respectively. On the one hand, OxA-induced inhibition of Isc was reduced by the neuronal blocker tetrodotoxin (TTX) and by a cholecystokinin (CCK) 2R antagonist, indicating involvement of neuronal and endocrine CCK-releasing cells. The OX(1)R antagonist SB334867 had no effect on OxA-induced inhibition, which is likely to occur via a neuronal and/or endocrine OX(2)R. On the other hand, SB334867 induced a significant right shift of the concentration-effect curve for OxB. This OxB-preferring OX(1)R pathway was not sensitive to TTX or to CCKR antagonists, suggesting that OxB may act directly on enterocytic OX(1)R. These distinct effects of OxA and OxB are consistent with the expression of OX(1)R and OX(2)R mRNA in the epithelial and nonepithelial tissues, respectively. CONCLUSIONS: Our data delineate a new function for orexins as inhibitors of intestinal glucose absorption and provide a new basis for orexin-induced short-term control of energy homeostasis.


Sujet(s)
Glucose/métabolisme , Absorption intestinale , Muqueuse intestinale/physiologie , Protéines et peptides de signalisation intracellulaire/physiologie , Neurones/physiologie , Neuropeptides/physiologie , Animaux , Transit gastrointestinal , Hyperglycémie provoquée , Hypothalamus/physiologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Protéines et peptides de signalisation intracellulaire/génétique , Jéjunum/effets des médicaments et des substances chimiques , Jéjunum/innervation , Jéjunum/physiologie , Mâle , Neuropeptides/génétique , Orexines , ARN/génétique , ARN/isolement et purification , Rats , Rat Wistar , RT-PCR , Tétrodotoxine/pharmacologie
20.
Biol Cell ; 99(11): 639-47, 2007 Nov.
Article de Anglais | MEDLINE | ID: mdl-17561806

RÉSUMÉ

BACKGROUND INFORMATION: TSPO (translocator protein), known previously as PBR (peripheral-type benzodiazepine receptor), is a 18 kDa protein expressed in the mitochondrial membrane of a variety of tissues. TSPO has been reported to be over-expressed in human colorectal tumours and cancer cell lines, but its function is not well characterized. RESULTS: We investigated the expression and function of TSPO in the human colon cancer cells HT-29. Immunohistochemical studies revealed that TSPO is localized in mitochondria, and its endogenous ligand, the polypeptide diazepam-binding inhibitor, in the cytosol. Radioligand binding studies using the specific high-affinity drug ligand [(3)H]PK 11195 and membrane fraction demonstrated saturable binding, with K(d) and B(max) values of 13.5+/-1.5 nM and 10.1+/-1.0 pmol/mg respectively. PK 11195 induced a rapid and transient dose-dependent rise in intracellular [Ca(2+)], which was unaffected by extracellular Ca(2+), but was blocked by the PTP (permeability transition pore) inhibitor, cyclosporin A, and by the TSPO partial agonist, flunitrazepam. Using HT-29 clone 19A cell line, which forms cell monolayers, we demonstrated that TSPO ligand stimulated a Ca(2+)-dependent transepithelial Cl(-) secretion. This secretion was inhibited: (i) after removal of extracellular Cl(-); (ii) by apical addition of the Cl(-) channel blocker NPPB [5-nitro-2-(3-phenylpropylamino)-benzoate]; and (iii) by basolateral addition of the Na(+)-K(+)-2Cl(-) co-transporter inhibitor bumetanide. Furthermore, the intracellular Ca(2+) chelator BAPTA/AM [bis-(o-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid tetrakis(acetoxymethyl ester)] and cyclosporin A abolished the rise in PK 11195-induced Cl(-) secretion. CONCLUSIONS: These findings indicate that TSPO is located in mitochondrial membranes of HT-29 and reveal that its activation induces a rise in cytosolic Ca(2+), leading to the stimulation of Cl(-) secretion.


Sujet(s)
Antinéoplasiques/pharmacologie , Calcium/métabolisme , Chlorures/métabolisme , Isoquinoléines/pharmacologie , Membranes mitochondriales/métabolisme , Protéines mitochondriales/biosynthèse , Protéines tumorales/biosynthèse , Récepteurs GABA/biosynthèse , Inhibiteurs de l'angiogenèse/pharmacologie , Lignée cellulaire tumorale , Tumeurs du côlon , Ciclosporine/pharmacologie , Antienzymes/pharmacologie , Flunitrazépam/pharmacologie , Modulateurs GABA/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Ligands , Mitochondries/métabolisme , Protéines mitochondriales/antagonistes et inhibiteurs , Protéines tumorales/antagonistes et inhibiteurs , Nitro-benzoates/pharmacologie , Symporteurs des ions sodium-potassium-chlorure
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...