Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 303(5): G589-99, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22700822

ABSTRACT

We studied the effect of dietary fat type, varying in polyunsaturated-to-saturated fatty acid ratios (P/S), on development of metabolic syndrome. C57Bl/6J mice were fed purified high-fat diets (45E% fat) containing palm oil (HF-PO; P/S 0.4), olive oil (HF-OO; P/S 1.1), or safflower oil (HF-SO; P/S 7.8) for 8 wk. A low-fat palm oil diet (LF-PO; 10E% fat) was used as a reference. Additionally, we analyzed diet-induced changes in gut microbiota composition and mucosal gene expression. The HF-PO diet induced a higher body weight gain and liver triglyceride content compared with the HF-OO, HF-SO, or LF-PO diet. In the intestine, the HF-PO diet reduced microbial diversity and increased the Firmicutes-to-Bacteroidetes ratio. Although this fits a typical obesity profile, our data clearly indicate that an overflow of the HF-PO diet to the distal intestine, rather than obesity itself, is the main trigger for these gut microbiota changes. A HF-PO diet-induced elevation of lipid metabolism-related genes in the distal small intestine confirmed the overflow of palm oil to the distal intestine. Some of these lipid metabolism-related genes were previously already associated with the metabolic syndrome. In conclusion, our data indicate that saturated fat (HF-PO) has a more stimulatory effect on weight gain and hepatic lipid accumulation than unsaturated fat (HF-OO and HF-SO). The overflow of fat to the distal intestine on the HF-PO diet induced changes in gut microbiota composition and mucosal gene expression. We speculate that both are directly or indirectly contributive to the saturated fat-induced development of obesity and hepatic steatosis.


Subject(s)
Dietary Fats/pharmacology , Fatty Acids/pharmacology , Fatty Liver/metabolism , Intestines/drug effects , Metabolic Syndrome/metabolism , Obesity/metabolism , Animals , Fatty Liver/genetics , Gene Expression/drug effects , Intestinal Mucosa/metabolism , Intestines/microbiology , Liver/metabolism , Metabolic Syndrome/genetics , Metagenome , Mice , Mice, Inbred C57BL , Obesity/genetics
2.
Br J Nutr ; 105(7): 1005-11, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21205428

ABSTRACT

There is increased interest in the potential protective role of dietary Ca in the development of metabolic disorders related to the metabolic syndrome. Ca-induced intestinal precipitation of fatty acids and bile acids as well as systemic metabolic effects of Ca on adipose tissue is proposed to play a causal role. In this experiment, we have studied all these aspects to validate the suggested protective effect of Ca supplementation, independent of other dietary changes, on the development of diet-induced obesity and insulin resistance. In our diet intervention study, C57BL/6J mice were fed high-fat diets differing in Ca concentrations (50 v. 150 mmol/kg). Faecal excretion analyses showed an elevated precipitation of intestinal fatty acids (2·3-fold; P < 0·01) and bile acids (2-fold; P < 0·01) on the high-Ca diet. However, this only led to a slight reduction in fat absorption (from 98 to 95 %; P < 0·01), mainly in the distal small intestine as indicated by gene expression changes. We found no effect on body-weight gain. Lipolysis and lipogenesis-related parameters in adipose tissue also showed no significant changes on the high-Ca diet, indicating no systemic effects of dietary Ca on adiposity. Furthermore, early gene expression changes of intestinal signalling molecules predicted no protective effect of dietary Ca on the development of insulin resistance, which was confirmed by equal values for insulin sensitivity on both diets. Taken together, our data do not support the proposed protective effect of dietary Ca on the development of obesity and/or insulin resistance, despite a significant increase in faecal excretion of fatty acids and bile acids.


Subject(s)
Bile Acids and Salts/metabolism , Calcium, Dietary/pharmacology , Dietary Fats/metabolism , Fatty Acids/metabolism , Insulin Resistance , Intestine, Small/drug effects , Obesity/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Dietary Fats/adverse effects , Dietary Supplements , Feces/chemistry , Gene Expression/drug effects , Intestinal Absorption , Intestine, Small/metabolism , Lipogenesis/drug effects , Lipolysis/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/chemically induced , Trace Elements/pharmacology
3.
BMC Genomics ; 9: 231, 2008 May 19.
Article in English | MEDLINE | ID: mdl-18489776

ABSTRACT

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 (PPARalpha), which is abundantly expressed in enterocytes. In this study we examined the effects of acute nutritional activation of PPARalpha 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 PPARalpha-null mice. Treatment with the synthetic PPARalpha agonist WY14643 served as reference. RESULTS: We identified 74 barrier genes that were PPARalpha-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 PPARalpha 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. CONCLUSION: We identified intestinal barrier genes that were PPARalpha-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 PPARalpha as an important factor controlling this key function, and underscores the importance of PPARalpha for nutrient-mediated gene regulation in intestine.


Subject(s)
Dietary Fats, Unsaturated/pharmacology , Intestine, Small/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Animals , Biological Transport, Active , Cholesterol/metabolism , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Fatty Acids/metabolism , Gastrointestinal Motility , Gene Expression Profiling , Gene Expression Regulation/drug effects , Intestinal Absorption/drug effects , Intestinal Absorption/genetics , Intestinal Absorption/physiology , Intestine, Small/drug effects , Male , Mice , Mice, Knockout , Oleic Acid/pharmacology , Oligonucleotide Array Sequence Analysis , Oxidation-Reduction , Oxidative Stress , PPAR alpha/agonists , PPAR alpha/deficiency , Pyrimidines/pharmacology
4.
BMC Med Genomics ; 1: 14, 2008 May 06.
Article in English | MEDLINE | ID: mdl-18457598

ABSTRACT

BACKGROUND: 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. METHODS: 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. RESULTS: 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. CONCLUSION: 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.

5.
Am J Physiol Gastrointest Liver Physiol ; 294(5): G1171-80, 2008 May.
Article in English | MEDLINE | ID: mdl-18356535

ABSTRACT

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% (P < 0.05). In an acute experiment in which triacylglycerols of unsaturated fatty acids were given by gavage, we found that this downregulation occurs within a 6-h time frame. Studies in LXRalpha-null mice, confirmed by in vitro data, showed that fatty acid-induced downregulation of cholesterol transporters is LXRalpha independent and associated with a posttranslational increase in 3-hydroxy-3-methylglutaryl-coenzyme A reductase activity that reflects induction of cholesterol biosynthesis as well as with a doubling of neutral fecal sterol loss. This study highlights the induction of adaptive changes in small intestinal cholesterol metabolism during exposure to dietary fat.


Subject(s)
Cholesterol/metabolism , Dietary Fats/pharmacology , Intestine, Small/metabolism , Membrane Transport Proteins/genetics , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , Animals , Bile Acids and Salts/analysis , Cholesterol/blood , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Dietary Fats/administration & dosage , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Feces/chemistry , Gene Expression Regulation/drug effects , Hydroxycholesterols/pharmacology , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Intestine, Small/cytology , Intestine, Small/drug effects , Lipoproteins/genetics , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Oligonucleotide Array Sequence Analysis , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...