16 research outputs found

    PPARalpha-mediated effects of dietary lipids on intestinal barrier gene expression

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>The selective absorption of nutrients and other food constituents in the small intestine is mediated by a group of transport proteins and metabolic enzymes, often collectively called 'intestinal barrier proteins'. An important receptor that mediates the effects of dietary lipids on gene expression is the peroxisome proliferator-activated receptor alpha (PPARα), which is abundantly expressed in enterocytes. In this study we examined the effects of acute nutritional activation of PPARα on expression of genes encoding intestinal barrier proteins. To this end we used triacylglycerols composed of identical fatty acids in combination with gene expression profiling in wild-type and PPARα-null mice. Treatment with the synthetic PPARα agonist WY14643 served as reference.</p> <p>Results</p> <p>We identified 74 barrier genes that were PPARα-dependently regulated 6 hours after activation with WY14643. For eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and oleic acid (OA) these numbers were 46, 41, and 19, respectively. The overlap between EPA-, DHA-, and WY14643-regulated genes was considerable, whereas OA treatment showed limited overlap. Functional implications inferred form our data suggested that nutrient-activated PPARα regulated transporters and phase I/II metabolic enzymes were involved in a) fatty acid oxidation, b) cholesterol, glucose, and amino acid transport and metabolism, c) intestinal motility, and d) oxidative stress defense.</p> <p>Conclusion</p> <p>We identified intestinal barrier genes that were PPARα-dependently regulated after acute activation by fatty acids. This knowledge provides a better understanding of the impact dietary fat has on the barrier function of the gut, identifies PPARα as an important factor controlling this key function, and underscores the importance of PPARα for nutrient-mediated gene regulation in intestine.</p

    The role of the small intestine in the development of dietary fat-induced obesity and insulin resistance in C57BL/6J mice

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Obesity and insulin resistance are two major risk factors underlying the metabolic syndrome. The development of these metabolic disorders is frequently studied, but mainly in liver, skeletal muscle, and adipose tissue. To gain more insight in the role of the small intestine in development of obesity and insulin resistance, dietary fat-induced differential gene expression was determined along the longitudinal axis of small intestines of C57BL/6J mice.</p> <p>Methods</p> <p>Male C57BL/6J mice were fed a low-fat or a high-fat diet that mimicked the fatty acid composition of a Western-style human diet. After 2, 4 and 8 weeks of diet intervention small intestines were isolated and divided in three equal parts. Differential gene expression was determined in mucosal scrapings using Mouse genome 430 2.0 arrays.</p> <p>Results</p> <p>The high-fat diet significantly increased body weight and decreased oral glucose tolerance, indicating insulin resistance. Microarray analysis showed that dietary fat had the most pronounced effect on differential gene expression in the middle part of the small intestine. By overrepresentation analysis we found that the most modulated biological processes on a high-fat diet were related to lipid metabolism, cell cycle and inflammation. Our results further indicated that the nuclear receptors Ppars, Lxrs and Fxr play an important regulatory role in the response of the small intestine to the high-fat diet. Next to these more local dietary fat effects, a secretome analysis revealed differential gene expression of secreted proteins, such as Il18, Fgf15, Mif, Igfbp3 and Angptl4. Finally, we linked the fat-induced molecular changes in the small intestine to development of obesity and insulin resistance.</p> <p>Conclusion</p> <p>During dietary fat-induced development of obesity and insulin resistance, we found substantial changes in gene expression in the small intestine, indicating modulations of biological processes, especially related to lipid metabolism. Moreover, we found differential expression of potential signaling molecules that can provoke systemic effects in peripheral organs by influencing their metabolic homeostasis. Many of these fat-modulated genes could be linked to obesity and/or insulin resistance. Together, our data provided various leads for a causal role of the small intestine in the etiology of obesity and/or insulin resistance.</p

    Traiter ou ne pas traiter?

    Get PDF
    La décision de traiter ou non repose sur le risque fracturaired'un patient, son adhésion à la prise en charge, l'efficacité dutraitement et son profil d'effets indésirables, et sur le remboursementde ce dernier. Différents algorithmes, dont l'outil FRAX,permettent d'évaluer le risque fracturaire. Ce dernier outil aquelques limites: absence de quantification du type de fracturesantérieures, mauvaise appréciation du risque lié à une corticothérapiesystémique active, absence de validation prospective.Le seuil thérapeutique peut être fixe indépendant de l'âgeou varier avec l'âge. Les analyses coût-efficacité montrent quepour un même profil de risque, plus la personne est âgée, plusgrand est le bénéfice économique. Le jugement clinique peutnous guider dans certaines situations. La fracture non traumatique,l'âge avancé, la corticothérapie, un T-score abaissé sontles principaux facteurs de risque utilisés en pratique. Dans l'approchediagnostique, la recherche de la fracture vertébrale sousjacenteest impérative, idéalement par IVA. Les quelques exemplesci-dessous montrent les limites des algorithmes et dujugement clinique.Sans facteur de risque pour l'ostéoporose, mais avec un T-scoreà -3.2 DS, à quel âge va-t-on débuter un traitement chez cettefemme ? Avant ou après 60 ans ? Certaines situations cliniquessemblent claires et posent l'indication à traiter: la fracture de lahanche, la fracture vertébrale spontanée, la corticothérapie aulong cours. Mais si pour ces trois situations la densitométrieosseuse donne un T-score à -0.5 DS, ou si le patient a 35 ans,est-ce que chaque clinicien sera d'accord de traiter ? On saitpar exemple que le risque fracturaire sous corticothérapie aulong cours semble faible chez la femme préménopausée et chezl'homme avant 50 ans. Que faire après une fracture du poignetà 50 ans : ne pas traiter si le T-score est à -1.5 DS et traiter si leT-score est à -3 DS ? L'antécédent de fracture du poignet n'estpas un facteur de risque aussi fort de la fracture subséquenteque la hanche, la vertèbre ou l'humérus. Et chez cette femme de80 ans ayant eu une fracture de côte sur un effort de toux, avecun T-score à -2.5 DS ? Ou cette autre femme de 83 ans, sansfacteur de risque particulier pour l'ostéoporose mais avec unT-score à -3.1 DS ? Ces deux dernières femmes bénéficient d'untraitement en terme économique et le praticien respecte les indicationsau remboursement. Mais certains modèles préconisentde ne pas traiter les personnes très âgées si leur risque fracturairen'est pas très élevé.Dans toutes ces situations, le partage de la décision entre lepraticien et son patient prime sur les éventuelles propositionsissues d'algorithmes qui doivent encore être améliorés

    PPARalpha-mediated effects of dietary lipids on intestinal barrier gene expression

    No full text
    Background: The selective absorption of nutrients and other food constituents in the small intestine is mediated by a group of transport proteins and metabolic enzymes, often collectively called ‘intestinal barrier proteins’. An important receptor that mediates the effects of dietary lipids on gene expression is the peroxisome proliferator-activated receptor alpha (PPARα), which is abundantly expressed in enterocytes. In this study we examined the effects of acute nutritional activation of PPARα on expression of genes encoding intestinal barrier proteins. To this end we used triacylglycerols composed of identical fatty acids in combination with gene expression profiling in wild-type and PPARα-null mice. Treatment with the synthetic PPARα agonist WY14643 served as reference

    Role of small intestine in the development of dietary fat-induced obesity and insulin resistance in C57BL/6J mice.

    No full text
    Obesity and insulin resistance are two major risk factors underlying the metabolic syndrome. To gain more insight in the role of the small intestine in the etiology of these metabolic disorders, a microarray study was performed on small intestines (SI) of C57BL/6J mice that were fed a high fat diet mimicking the fatty acid composition of a Western-style human diet. The mice became obese and developed dietary fat-induced glucose intolerance. For gene expression profiling, the small intestines were subdivided in three equal parts along the longitudinal axis. The most pronounced effects of dietary fat were detected in part 2 of the small intestine. The biological processes that were most extensively modulated on a high fat diet were related to lipid metabolism, especially β- and ω-fatty acid oxidation seemed to play an important role, cell cycle and inflammation/immune response. An additional secretome analysis revealed differentially expressed secreted proteins, such as Il18, Ffgf15, Mif, Igfbp3 and Angptl4, which might provoke systemic effects in peripheral organs by influencing their metabolic homeostasis. Furthermore, many of the dietary fat-modulated genes and biological processes in small intestine were previously already associated with obesity and/or insulin resistance. Together, the data of this exploratory study provided various leads for an essential role of the small intestine in development of obesity and/or insulin resistance

    PPARalpha-mediated effects of dietary lipids on intestinal barrier gene expression-1

    No full text
    Lated along the proximal-distal axis of the small intestine of wild-type mice that received the control diet (white, open bars), or were acutely treated (6 hr) with WY14643 (black, closed bars) (n = 4 per group). Small intestines were divided into 10 equal parts; part 1 refers to the most proximal part (duodenum), part 10 refers to the most distal (terminal ileum). Messenger RNA levels were standardized to cyclophilin; part 1 of the non-treated mice was arbitrarily set to 1. Significance of control versus treated wild-type mice was determined per segment using an unpaired student's -test. * p-value < 0.05. Data are presented as mean ± standard error.<p><b>Copyright information:</b></p><p>Taken from "PPARalpha-mediated effects of dietary lipids on intestinal barrier gene expression"</p><p>http://www.biomedcentral.com/1471-2164/9/231</p><p>BMC Genomics 2008;9():231-231.</p><p>Published online 19 May 2008</p><p>PMCID:PMC2408604.</p><p></p

    PPARalpha-mediated effects of dietary lipids on intestinal barrier gene expression-0

    No full text
    genes for each treatment. A) Overlap between OA, EPA and DHA, B) Overlap between EPA, DHA, and WY14643.<p><b>Copyright information:</b></p><p>Taken from "PPARalpha-mediated effects of dietary lipids on intestinal barrier gene expression"</p><p>http://www.biomedcentral.com/1471-2164/9/231</p><p>BMC Genomics 2008;9():231-231.</p><p>Published online 19 May 2008</p><p>PMCID:PMC2408604.</p><p></p

    PPARalpha-mediated effects of dietary lipids on intestinal barrier gene expression-2

    No full text
    Ed genes in sections isolated along the proximal-distal axis of the small intestine from PPARα-null mice (white, open bars) and wild-type mice (black, closed bars) that were acutely treated (6 hr) with the 4 agonists (n = 4 per group). The small intestine was divided into 10 equal parts; part 1 refers to the most proximal part (duodenum), part 10 refers to the most distal (terminal ileum). Messenger RNA levels were standardized to cyclophilin; part 1 of the PPARα-null mice was arbitrarily set to 1. White bars represent the PPARα-null mice, black bars represent the wild-type mice. Significance of treated WT versus treated KO mice was determined per segment using an unpaired student's -test. * p-value < 0.05. Data are presented as mean ± standard error. A) fatty acid transport protein 4 (Fatp4). B) ATP-binding cassette, sub-family D, member 3 (Abcd3; ALD). C) cytochrome P450, family 2, subfamily c, polypeptide 65 (Cyp2c65). D) cytochrome P450, family 4, subfamily f, polypeptide 16 (Cyp4f16).<p><b>Copyright information:</b></p><p>Taken from "PPARalpha-mediated effects of dietary lipids on intestinal barrier gene expression"</p><p>http://www.biomedcentral.com/1471-2164/9/231</p><p>BMC Genomics 2008;9():231-231.</p><p>Published online 19 May 2008</p><p>PMCID:PMC2408604.</p><p></p

    A cholesterol-free, high-fat diet suppresses gene expression of cholesterol transporters in murine small intestine

    No full text
    Transporters present in the epithelium of the small intestine determine the efficiency by which dietary and biliary cholesterol are taken up into the body and thus control whole-body cholesterol balance. Niemann-Pick C1 Like Protein 1 (Npc1l1) transports cholesterol into the enterocyte, whereas ATP-binding cassette transporters Abca1 and Abcg5/Abcg8 are presumed to be involved in cholesterol efflux from the enterocyte toward plasma HDL and back into the intestinal lumen, respectively. Abca1, Abcg5, and Abcg8 are well-established liver X receptor (LXR) target genes. We examined the effects of a high-fat diet on expression and function of cholesterol transporters in the small intestine in mice. Npc1l1, Abca1, Abcg5, and Abcg8 were all downregulated after 2, 4, and 8 wk on a cholesterol-free, high-fat diet. The high-fat diet did not affect biliary cholesterol secretion but diminished fractional cholesterol absorption from 61 to 42% (
    corecore