Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nutr Metab (Lond) ; 10(1): 11, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23327542

ABSTRACT

BACKGROUND: GPR43 is a G-protein-coupled receptor that participates in adipocyte differentiation in mice and is over-expressed in adipose tissue of obese mice. The aim of this study was to investigate the implication of GPR43 in adipogenesis in humans and to determine the influence of obesity on its expression in human adipose tissue. FINDINGS: Preadipocytes were isolated from human omental adipose tissue and cultured during 13 days. One PPARγ agonist (troglitazone) and three GPR43 agonists (two physiological and one synthetic) were tested for their ability to induce differentiation. After 13 days, the three GPR43 agonists had no impact on aP2 expression, a marker of adipocyte differentiation, whereas troglitazone led to a huge over-expression of aP2 in these cells but tended to decrease GPR43 expression (p=0.06).GPR43 and inflammatory markers expression was also quantified in omental adipose tissue from lean and obese individuals. GPR43 expression in total adipose tissue was similar between obese patients and lean subjects and did not correlate with aP2 expression. In contrast, GPR43 expression positively correlated with TNFα mRNA. CONCLUSIONS: Our results suggest the absence of relationship between GPR43 and adipocyte differentiation in humans, unlike what was observed in mice. Furthermore, GPR43 expression is not increased in adipose tissue from obese subjects but could be related to TNFα-related inflammatory processes.

2.
Mol Nutr Food Res ; 57(2): 347-59, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23203768

ABSTRACT

SCOPE: Recent data suggest that gut microbiota contributes to the regulation of host lipid metabolism. We report how fermentable dietary fructo-oligosaccharides (FOS) control hepatic steatosis induced by n-3 PUFA depletion, which leads to hepatic alterations similar to those observed in non-alcoholic fatty liver disease patients. METHODS AND RESULTS: C57Bl/6J mice fed an n-3 PUFA-depleted diet for 3 months were supplemented with FOS during the last 10 days of treatment. FOS-treated mice exhibited higher caecal Bifidobacterium spp. and lower Roseburia spp. content. Microarray analysis of hepatic mRNA revealed that FOS supplementation reduced hepatic triglyceride accumulation through a proliferator-activated receptor α-stimulation of fatty acid oxidation and lessened cholesterol accumulation by inhibiting sterol regulatory element binding protein 2-dependent cholesterol synthesis. Cultured precision-cut liver slices confirmed the inhibition of fatty acid oxidation. FOS effects were related to a decreased hepatic micro-RNA33 expression and to an increased colonic glucagon-like peptide 1 production. CONCLUSIONS: The changes in gut microbiota composition by n-3 PUFA-depletion and prebiotics modulate hepatic steatosis by changing gene expression in the liver, a phenomenon that could implicate micro-RNA and gut-derived hormones. Our data underline the advantage of targeting the gut microbiota by colonic nutrients in the management of liver disease.


Subject(s)
Cholesterol/biosynthesis , Dietary Supplements , Fatty Acids, Omega-3/metabolism , Fatty Liver/pathology , Prebiotics , Animals , Bifidobacterium/growth & development , Energy Intake , Fatty Liver/metabolism , Gastrointestinal Tract/microbiology , Gene Expression Regulation , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Lipid Metabolism , Liver/metabolism , Male , Metagenome/physiology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Oligosaccharides/administration & dosage , Oxidative Stress/drug effects , PPAR alpha/genetics , PPAR alpha/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Nutr Rev ; 70(11): 631-41, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23110642

ABSTRACT

Coenzyme Q (CoQ), a lipophilic cofactor of the electron transport chain in the mitochondria, can be synthesized endogenously or provided by food. The aim of this review is to summarize the in vitro cell culture studies, the in vivo animal studies, and the human studies investigating the impact of CoQ supplementation on the occurrence of obesity and related disorders (diabetes, hypertension, lipemia, and atherosclerosis). The antioxidative properties of CoQ have been observed in different experimental models of atherosclerosis, obesity, and diabetes. The recent discovery of the anti-inflammatory effect of CoQ, mostly described in vitro, has generated increased interest in CoQ supplementation, but it needs to be confirmed in vivo in pathological situations. CoQ intervention studies in humans failed to show reproducible effects on body weight, fat mass, or glycemia, but CoQ supplementation does seem to have an antihypertensive effect. The molecular mechanism to explain this effect has only recently been discovered.


Subject(s)
Metabolic Syndrome/enzymology , Obesity/enzymology , Ubiquinone/physiology , Animals , Cell Membrane/metabolism , Electron Transport , Humans , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Mitochondria/metabolism , Obesity/drug therapy , Obesity/metabolism , Oxidation-Reduction , Ubiquinone/therapeutic use
4.
PLoS One ; 7(6): e37971, 2012.
Article in English | MEDLINE | ID: mdl-22761662

ABSTRACT

The gut microbiota has recently been proposed as a novel component in the regulation of host homeostasis and immunity. We have assessed for the first time the role of the gut microbiota in a mouse model of leukemia (transplantation of BaF3 cells containing ectopic expression of Bcr-Abl), characterized at the final stage by a loss of fat mass, muscle atrophy, anorexia and inflammation. The gut microbial 16S rDNA analysis, using PCR-Denaturating Gradient Gel Electrophoresis and quantitative PCR, reveals a dysbiosis and a selective modulation of Lactobacillus spp. (decrease of L. reuteri and L. johnsonii/gasseri in favor of L. murinus/animalis) in the BaF3 mice compared to the controls. The restoration of Lactobacillus species by oral supplementation with L. reuteri 100-23 and L. gasseri 311476 reduced the expression of atrophy markers (Atrogin-1, MuRF1, LC3, Cathepsin L) in the gastrocnemius and in the tibialis, a phenomenon correlated with a decrease of inflammatory cytokines (interleukin-6, monocyte chemoattractant protein-1, interleukin-4, granulocyte colony-stimulating factor, quantified by multiplex immuno-assay). These positive effects are strain- and/or species-specific since L. acidophilus NCFM supplementation does not impact on muscle atrophy markers and systemic inflammation. Altogether, these results suggest that the gut microbiota could constitute a novel therapeutic target in the management of leukemia-associated inflammation and related disorders in the muscle.


Subject(s)
Disease Models, Animal , Inflammation Mediators/metabolism , Inflammation/prevention & control , Lactobacillus/physiology , Leukemia, Experimental/complications , Muscular Atrophy/prevention & control , Acute Disease , Animals , Cells, Cultured , Dietary Supplements , Female , Fusion Proteins, bcr-abl/genetics , Gastrointestinal Tract/microbiology , Inflammation/etiology , Leukemia, Experimental/genetics , Leukemia, Experimental/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/microbiology , Liver Neoplasms/pathology , Metagenome , Mice , Mice, Inbred BALB C , Muscular Atrophy/etiology , Precursor Cells, B-Lymphoid/transplantation , Splenic Neoplasms/metabolism , Splenic Neoplasms/microbiology , Splenic Neoplasms/pathology
5.
PLoS One ; 6(8): e23365, 2011.
Article in English | MEDLINE | ID: mdl-21853118

ABSTRACT

Patients with non-alcoholic fatty liver disease are characterised by a decreased n-3/n-6 polyunsaturated fatty acid (PUFA) ratio in hepatic phospholipids. The metabolic consequences of n-3 PUFA depletion in the liver are poorly understood. We have reproduced a drastic drop in n-3 PUFA among hepatic phospholipids by feeding C57Bl/6J mice for 3 months with an n-3 PUFA depleted diet (DEF) versus a control diet (CT), which only differed in the PUFA content. DEF mice exhibited hepatic insulin resistance (assessed by euglycemic-hyperinsulinemic clamp) and steatosis that was associated with a decrease in fatty acid oxidation and occurred despite a higher capacity for triglyceride secretion. Microarray and qPCR analysis of the liver tissue revealed higher expression of all the enzymes involved in lipogenesis in DEF mice compared to CT mice, as well as increased expression and activation of sterol regulatory element binding protein-1c (SREBP-1c). Our data suggest that the activation of the liver X receptor pathway is involved in the overexpression of SREBP-1c, and this phenomenon cannot be attributed to insulin or to endoplasmic reticulum stress responses. In conclusion, n-3 PUFA depletion in liver phospholipids leads to activation of SREBP-1c and lipogenesis, which contributes to hepatic steatosis.


Subject(s)
Fatty Acids, Omega-3/metabolism , Fatty Liver/genetics , Genome/genetics , Insulin Resistance/genetics , Liver/metabolism , Animals , Cannabinoid Receptor Modulators/metabolism , Cholesterol/biosynthesis , Diet , Endoplasmic Reticulum Stress/genetics , Fatty Liver/pathology , Feeding Behavior , Gene Expression Regulation , Lipid Metabolism/genetics , Liver/pathology , Liver X Receptors , Mice , Oligonucleotide Array Sequence Analysis , Orphan Nuclear Receptors/metabolism , Oxidation-Reduction , Phospholipids/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
6.
Nutr Metab (Lond) ; 8(1): 44, 2011 Jun 27.
Article in English | MEDLINE | ID: mdl-21707971

ABSTRACT

BACKGROUND: Western diet is characterized by an insufficient n-3 polyunsaturated fatty acid (PUFA) consumption which is known to promote the pathogenesis of several diseases. We have previously observed that mice fed with a diet poor in n-3 PUFA for two generations exhibit hepatic steatosis together with a decrease in body weight. The gut microbiota contributes to the regulation of host energy metabolism, due to symbiotic relationship with fermentable nutrients provided in the diet. In this study, we have tested the hypothesis that perturbations of the gut microbiota contribute to the metabolic alterations occurring in mice fed a diet poor in n-3 PUFA for two generations (n-3/- mice). METHODS: C57Bl/6J mice fed with a control or an n-3 PUFA depleted diet for two generations were supplemented with prebiotic (inulin-type Fructooligosaccharides, FOS, 0.20 g/day/mice) during 24 days. RESULTS: n-3/-mice exhibited a marked drop in caecum weight, a decrease in lactobacilli and an increase in bifidobacteria in the caecal content as compared to control mice (n-3/+ mice). Dietary supplementation with FOS for 24 days was sufficient to increase caecal weight and bifidobacteria count in both n-3/+ and n-3/-mice. Moreover, FOS increased lactobacilli content in n-3/-mice, whereas it decreased their level in n-3/+ mice. Interestingly, FOS treatment promoted body weight gain in n-3/-mice by increasing energy efficiency. In addition, FOS treatment decreased fasting glycemia and lowered the higher expression of key factors involved in the fatty acid catabolism observed in the liver of n-3/-mice, without lessening steatosis. CONCLUSIONS: the changes in the gut microbiota composition induced by FOS are different depending on the type of diet. We show that FOS may promote lactobacilli and counteract the catabolic status induced by n-3 PUFA depletion in mice, thereby contributing to restore efficient fat storage.

7.
J Nutr Biochem ; 22(8): 712-22, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21115338

ABSTRACT

Inulin-type fructans (ITF) are nondigestible/fermentable carbohydrates which are able - through the modification of the gut microbiota - to counteract high-fat (HF) diet-induced obesity, endotoxemia and related-metabolic alterations. However, their influence on adipose tissue metabolism has been poorly studied until now. The aim of this study was to assess the influence of ITF supplementation on adipose tissue metabolism, by focusing on a G protein-coupled receptor (GPR), GPR43, as a potential link between gut fermentation processes and white adipose tissue development. Male C57bl6/J mice were fed a standard diet or an HF diet without or with ITF (0.2 g/day per mouse) during 4 weeks. The HF diet induced an accumulation of large adipocytes, promoted peroxisome proliferator activated receptor gamma (PPARγ)-activated differentiation factors and led to a huge increase in GPR43 expression in the subcutaneous adipose tissue. All those effects were blunted by ITF treatment, which modulated the gut microbiota in favor of bifidobacteria at the expense of Roseburia spp. and of Clostridium cluster XIVa. The dietary modulation of GPR43 expression seems independent of endotoxemia, in view of data obtained in vivo (acute and chronic lipopolysaccharides treatment). In conclusion, ITF, which promote gut fermentation, paradoxically counteract GPR43 overexpression induced in the adipose tissue by an HF diet, a phenomenon that correlates with a beneficial effect on adiposity and with potential decrease in PPARγ-activated processes.


Subject(s)
Adipogenesis , Inulin/pharmacology , PPAR gamma/genetics , Prebiotics/analysis , Receptors, G-Protein-Coupled/genetics , Adipose Tissue/metabolism , Animals , Body Weight , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Diet, High-Fat/adverse effects , Dietary Fats/administration & dosage , Electrophoresis, Polyacrylamide Gel , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fructans/pharmacology , Gene Expression Regulation , Immunoblotting , Male , Mice , Mice, Inbred C57BL , PPAR gamma/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors
8.
J Nutr ; 140(3): 509-14, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20089787

ABSTRACT

Magnesium (Mg) deficiency is a common nutritional disorder that is linked to an inflammatory state characterized by increased plasma acute phase protein and proinflammatory cytokine concentrations. Recent studies have shown that changes in the composition of gut microbiota composition participate in systemic inflammation. In this study, therefore, we assessed the potential role of gut microbiota in intestinal and systemic inflammation associated with Mg deficiency in mice. For this purpose, mice were fed a control or Mg-deficient diet (500 mg vs. 70 mg Mg/kg) for 4 or 21 d. Compared with the mice fed the control diet, mice fed the Mg-deficient diet for 4 d had a lower gut bifidobacteria content (-1.5 log), a 36-50% lower mRNA content of factors controlling gut barrier function in the ileum (zonula occludens-1, occludin, proglucagon), and a higher mRNA content (by approximately 2-fold) in the liver and/or intestine of tumor necrosis factor-alpha, interleukin-6, CCAAT/enhancer binding protein homologous protein, and activating transcription factor 4, reflecting inflammatory and cellular stress. In contrast, mice fed the Mg-deficient diet for 21 d had a higher cecal bifidobacteria content compared with the control group, a phenomenon accompanied by restoration of the intestinal barrier and the absence of inflammation. In conclusion, we show that Mg deficiency, independently of any other changes in nutrient intake, modulates the concentration of bifidobacteria in the gut, a phenomenon that may time-dependently affect inflammation and metabolic disorders in mice.


Subject(s)
Bifidobacterium/physiology , Colon/microbiology , Inflammation/metabolism , Magnesium/metabolism , Animals , Body Weight , Colon/drug effects , Male , Mice , Mice, Inbred C57BL , Nutrition Disorders
9.
Am J Clin Nutr ; 90(5): 1236-43, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19776140

ABSTRACT

BACKGROUND: We have previously shown that gut microbial fermentation of prebiotics promotes satiety and lowers hunger and energy intake in humans. In rodents, these effects are associated with an increase in plasma gut peptide concentrations, which are involved in appetite regulation and glucose homeostasis. OBJECTIVE: Our aim was to examine the effects of prebiotic supplementation on satiety and related hormones during a test meal for human volunteers by using a noninvasive micromethod for blood sampling to measure plasma gut peptide concentrations. DESIGN: This study was a randomized, double-blind, parallel, placebo-controlled trial. A total of 10 healthy adults (5 men and 5 women) were randomly assigned to groups that received either 16 g prebiotics/d or 16 g dextrin maltose/d for 2 wk. Meal tolerance tests were performed in the morning to measure the following: hydrogen breath test, satiety, glucose homeostasis, and related hormone response. RESULTS: We show that the prebiotic treatment increased breath-hydrogen excretion (a marker of gut microbiota fermentation) by approximately 3-fold and lowered hunger rates. Prebiotics increased plasma glucagon-like peptide 1 and peptide YY concentrations, whereas postprandial plasma glucose responses decreased after the standardized meal. The areas under the curve for plasma glucagon-like peptide 1 and breath-hydrogen excretion measured after the meal (0-60 min) were significantly correlated (r = 0.85, P = 0.007). The glucose response was inversely correlated with the breath-hydrogen excretion areas under the curve (0-180 min; r = -0.73, P = 0.02). CONCLUSION: Prebiotic supplementation was associated with an increase in plasma gut peptide concentrations (glucagon-like peptide 1 and peptide YY), which may contribute in part to changes in appetite sensation and glucose excursion responses after a meal in healthy subjects.


Subject(s)
Appetite/physiology , Dietary Fiber/pharmacology , Dietary Supplements , Eating/physiology , Incretins/biosynthesis , Satiety Response/physiology , Adult , Appetite/drug effects , Blood Glucose/metabolism , Breath Tests , Double-Blind Method , Female , Glucagon-Like Peptide 1/blood , Humans , Hydrogen/analysis , Male , Pancreatic Polypeptide/blood , Peptide YY/blood
10.
Biochem Pharmacol ; 78(11): 1391-400, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19632207

ABSTRACT

BACKGROUND: Diabetes and obesity are metabolic disorders induced by an excessive dietary intake of fat, usually related to inflammation and oxidative stress. AIMS: The aim of the study is to investigate the effect of the antioxidant coenzyme Q10 (CoQ10) on hepatic metabolic and inflammatory disorders associated with diet-induced obesity and glucose intolerance. METHODS: C57bl6/j mice were fed for 8 weeks, either a control diet (CT) or a high-fat diet plus 21% fructose in the drinking water (HFF). CoQ10 supplementation was performed in this later condition (HFFQ). RESULTS: HFF mice exhibit increased energy consumption, fat mass development, fasting glycaemia and insulinemia and impaired glucose tolerance. HFF treatment promoted the expression of genes involved in reactive oxygen species production (NADPH oxidase), inflammation (CRP, STAMP2) and metabolism (CPT1alpha) in the liver. CoQ10 supplementation decreased the global hepatic mRNA expression of inflammatory and metabolic stresses markers without changing obesity and tissue lipid peroxides compared to HFF mice. HFF diets paradoxically decreased TBARS (reflecting lipid peroxides) levels in liver, muscle and adipose tissue versus CT group, an effect related to vitamin E content of the diet. CONCLUSION: In conclusion, HFF model promotes glucose intolerance and obesity by a mechanism independent on the level of tissue peroxides. CoQ10 tends to decrease hepatic stress gene expression, independently of any modulation of lipid peroxidation, which is classically considered as its most relevant effect.


Subject(s)
Dietary Fats/administration & dosage , Liver/drug effects , Obesity/drug therapy , Oxidative Stress/drug effects , Ubiquinone/analogs & derivatives , Animals , Biomarkers/metabolism , Body Weight/drug effects , Energy Metabolism/drug effects , Fructose/administration & dosage , Glucose/metabolism , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Homeostasis , Inflammation/drug therapy , Inflammation/metabolism , Lipid Peroxides/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , RNA, Messenger/biosynthesis , Reactive Oxygen Species/metabolism , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
11.
Br J Nutr ; 102(3): 462-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19161640

ABSTRACT

The mechanism, by which a high-fat (HF) diet could impair glucose metabolism, is not completely understood but could be related to inflammation, lipotoxicity and oxidative stress. Lipid peroxides have been proposed as key mediators of intracellular metabolic response. The purpose of the present study was to analyse, in mice fed with a HF diet, the possible association between obesity and glucose tolerance on the one hand, and between oxidative stress and lipid peroxidation on the other hand. The present results show that a HF diet (70 % energy as fat), v. a high-carbohydrate chow diet (control), increases body weight and fat mass development, and impairs glycaemia and insulinaemia within 4 weeks. It also promotes the expression of NADPH oxidase in the liver--signing both oxidative and inflammatory stress--but decreases thiobarbituric acid-reactive substances content in the liver as well as in epididymal, subcutaneous and visceral adipose tissues. HF diet, with elevated vitamin E content, induces high concentration of alpha-tocopherol in liver and adipose tissues, which contributes to the protection against lipid peroxidation. Thus, lipid peroxidation in key organs is not necessarily related to the development of metabolic disorders associated with diabetes and obesity.


Subject(s)
Diabetes Mellitus/metabolism , Dietary Fats/adverse effects , Lipid Peroxidation/physiology , Obesity/metabolism , Adipose Tissue/metabolism , Animals , Antioxidants/analysis , Biomarkers/analysis , Diabetes Mellitus/immunology , Dietary Supplements , Fatty Acids/analysis , Inflammation , Insulin Resistance , Lipids/analysis , Liver/chemistry , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , NADPH Oxidases/analysis , NADPH Oxidases/genetics , Oxidative Stress , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Thiobarbituric Acid Reactive Substances/analysis , Vitamin E/administration & dosage , Vitamins/administration & dosage , alpha-Tocopherol/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...