Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Stem Cells Int ; 2017: 9289213, 2017.
Article in English | MEDLINE | ID: mdl-28321259

ABSTRACT

Adipose-derived stem/stromal cells (ASCs) reside in the stromal vascular fraction (SVF) of adipose tissue (AT) and can be easily isolated. However, extraction of the SVF from lipoaspirate is a critical step in generating ASC, and semiautomated devices have been developed to enhance the efficacy and reproducibility of the outcomes and to decrease manipulation and contamination. In this study, we compared the reference method used in our lab for SVF isolation from lipoaspirate, with three medical devices: GID SVF-1™, Puregraft™, and Stem.pras®. Cell yield and their viability were evaluated as well as their phenotype with flow cytometry. Further on, we determined their proliferative potential using population doublings (PD), PD time (PDT), and clonogenicity assay (CFU-F). Finally, we checked their genetic stability using RT-qPCR for TERT mRNA assay and karyotyping as well as their multilineage potential including adipogenic, chondrogenic, and osteogenic differentiation. Our results demonstrate that all the devices allow the production of SVF cells with consistent yield and viability, in less time than the reference method. Expanded cells from the four methods showed no significant differences in terms of phenotype, proliferation capabilities, differentiation abilities, and genetic stability.

2.
J Nutr ; 145(8): 1754-62, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26108543

ABSTRACT

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.


Subject(s)
Colon/cytology , Dietary Fats/pharmacology , Fatty Acids, Unsaturated/pharmacology , Fatty Acids/pharmacology , Goblet Cells/drug effects , Animal Feed/analysis , Animals , Diet , Dietary Fats/administration & dosage , Fatty Acids/administration & dosage , Fatty Acids, Unsaturated/administration & dosage , Goblet Cells/metabolism , HT29 Cells , Humans , Mucin 5AC/genetics , Mucin 5AC/metabolism , Mucin-2/genetics , Mucin-2/metabolism , Plant Oils/administration & dosage , Plant Oils/chemistry , Rats , Rats, Wistar
3.
J Lipid Res ; 56(6): 1110-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25833686

ABSTRACT

Interleukin 17A (IL-17A) is a proinflammatory cytokine involved in the pathogenesis of chronic inflammatory diseases. In the field of immunometabolism, we have studied the impact of IL-17A on the lipid metabolism of human in vitro-generated monocyte-derived dendritic cells (DCs). Microarrays and lipidomic analysis revealed an intense remodeling of lipid metabolism induced by IL-17A in DCs. IL-17A increased 2-12 times the amounts of phospholipids, cholesterol, triglycerides, and cholesteryl esters in DCs. Palmitic (16:0), stearic (18:0), and oleic (18:ln-9c) acid were the main fatty acid chains present in DCs. They were strongly increased in response to IL-17A while their relative proportion remained unchanged. Capture of extracellular lipids was the major mechanism of lipid droplet accumulation, visualized by electron microscopy and Oil Red O staining. Besides this foamy phenotype, IL-17A induced a mixed macrophage-DC phenotype and expression of the nuclear receptor NR1H3/liver X receptor-α, previously identified in the context of atherosclerosis as the master regulator of cholesterol homeostasis in macrophages. These IL-17A-treated DCs were as competent as untreated DCs to stimulate allogeneic naive T-cell proliferation. Following this first characterization of lipid-rich DCs, we propose to call these IL-17A-dependent cells "foamy DCs" and discuss the possible existence of foamy DCs in atherosclerosis, a metabolic and inflammatory disorder involving IL-17A.


Subject(s)
Atherosclerosis/metabolism , Dendritic Cells/metabolism , Fatty Acids/metabolism , Interleukin-17/metabolism , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Differentiation/genetics , Cell Proliferation/genetics , Dendritic Cells/immunology , Foam Cells/metabolism , Gene Expression Regulation , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Lipid Droplets/immunology , Lipid Droplets/metabolism , Lipid Metabolism/immunology , Liver X Receptors , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Orphan Nuclear Receptors/biosynthesis , Orphan Nuclear Receptors/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
PLoS One ; 9(10): e110653, 2014.
Article in English | MEDLINE | ID: mdl-25337938

ABSTRACT

Inflammation is now recognized as a major factor contributing to type 2 diabetes (T2D). However, while the mechanisms and consequences associated with white adipose tissue inflammation are well described, very little is known concerning the situation in skeletal muscle. The aim of this study was to investigate, in vitro and in vivo, how skeletal muscle inflammation develops and how in turn it modulates local and systemic insulin sensitivity in different mice models of T2D and in humans, focusing on the role of the chemokine MCP1. Here, we found that skeletal muscle inflammation and macrophage markers are increased and associated with insulin resistance in mice models and humans. In addition, we demonstrated that intra-muscular TNFα expression is exclusively restricted to the population of intramuscular leukocytes and that the chemokine MCP1 was associated with skeletal muscle inflammatory markers in these models. Furthermore, we demonstrated that exposure of C2C12 myotubes to palmitate elevated the production of the chemokine MCP1 and that the muscle-specific overexpression of MCP1 in transgenic mice induced the local recruitment of macrophages and altered local insulin sensitivity. Overall our study demonstrates that skeletal muscle inflammation is clearly increased in the context of T2D in each one of the models we investigated, which is likely consecutive to the lipotoxic environment generated by peripheral insulin resistance, further increasing MCP1 expression in muscle. Consequently, our results suggest that MCP1-mediated skeletal muscle macrophages recruitment plays a role in the etiology of T2D.


Subject(s)
Chemokine CCL2/physiology , Insulin Resistance/immunology , Macrophages/immunology , Muscle, Skeletal/immunology , Myositis/immunology , Animals , Cell Line , Cell Movement , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myositis/metabolism
5.
Br J Nutr ; 112(4): 520-35, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-24932525

ABSTRACT

Dairy products derived from the milk of cows fed in pastures are characterised by higher amounts of conjugated linoleic acid and α-linolenic acid (ALA), and several studies have shown their ability to reduce cardiovascular risk. However, their specific metabolic effects compared with standard dairy in a high-fat diet (HFD) context remain largely unknown; this is what we determined in the present study with a focus on the metabolic and intestinal parameters. The experimental animals were fed for 12 weeks a HFD containing 20 % fat in the form of a pasture dairy cream (PDC) or a standard dairy cream (SDC). Samples of plasma, liver, white adipose tissue, duodenum, jejunum and colon were analysed. The PDC mice, despite a higher food intake, exhibited lower fat mass, plasma and hepatic TAG concentrations, and inflammation in the adipose tissue than the SDC mice. Furthermore, they exhibited a higher expression of hepatic PPARα mRNA and adipose tissue uncoupling protein 2 mRNA, suggesting an enhanced oxidative activity of the tissues. These results might be explained, in part, by the higher amounts of ALA in the PDC diet and in the liver and adipose tissue of the PDC mice. Moreover, the PDC diet was found to increase the proportions of two strategic cell populations involved in the protective function of the intestinal epithelium, namely Paneth and goblet cells in the small intestine and colon, compared with the SDC diet. In conclusion, a PDC HFD leads to improved metabolic outcomes and to a stronger gut barrier compared with a SDC HFD. This may be due, at least in part, to the protective mechanisms induced by specific lipids.


Subject(s)
Cattle/physiology , Diet/veterinary , Dietary Fats/therapeutic use , Functional Food , Milk , Obesity/physiopathology , Panniculitis/prevention & control , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animal Husbandry , Animals , Dairy Products/adverse effects , Dairy Products/analysis , Diet, High-Fat/adverse effects , Dietary Fats/adverse effects , Dietary Fats/analysis , Dietary Fats/metabolism , Female , Functional Food/analysis , Hypertriglyceridemia/etiology , Hypertriglyceridemia/prevention & control , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Lactation , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Milk/adverse effects , Milk/chemistry , Milk/metabolism , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Panniculitis/etiology , Plant Components, Aerial/chemistry , Plant Components, Aerial/growth & development , Poaceae/chemistry , Poaceae/growth & development , Random Allocation
6.
Nutr Res ; 33(11): 952-60, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24176235

ABSTRACT

Animal studies using a high-fat diet (HFD) have studied the effects of lipid overconsumption by comparing a defined HFD either with a natural-ingredient chow diet or with a defined low-fat diet (LFD), despite the dramatic differences between these control diets. We hypothesized that these differences in the control diet could modify the conclusions regarding the effects that an increase of fat in the diet has on several metabolic parameters. For 11 weeks, C57bl6/J mice were fed a low-fat chow diet (8% energy from fat), a typical semisynthetic LFD (12%), or a semisynthetic HFD (sy-HF) (40%). Conclusions about the effect of sy-HF on body weight gain, subcutaneous adipose tissue, insulin sensitivity, and adipose tissue inflammation were modified according to the control LFD. Conversely, conclusions about epididymal and retroperitoneal adipose tissue; fat intake effects on liver and muscular lipids, cholesterol, free fatty acids, and markers of low-grade inflammation; and of adipose tissue macrophage infiltration were the same regardless of the use of low-fat chow diet or semisynthetic LFD. For some physiological outcomes, conflicting conclusions were even reached about the effects of increased fat intake according to the chosen low-fat control. Some deleterious effects of sy-HF may not be explained by lipid overconsumption but rather by the overall quality of ingredients in a semisynthetic diet. According to the control LFD chosen, conclusions on the lipid-related effects of HFDs must be formulated with great care because some end points are profoundly affected by the ingredient composition of the diet rather than by fat content.


Subject(s)
Adipose Tissue/metabolism , Adiposity , Biomedical Research/methods , Diet, High-Fat/adverse effects , Inflammation/etiology , Insulin Resistance , Lipid Metabolism/drug effects , Animals , Diet, Fat-Restricted/standards , Dietary Fats/administration & dosage , Energy Intake , Liver/drug effects , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Subcutaneous Fat/metabolism , Weight Gain
7.
PLoS One ; 8(5): e63997, 2013.
Article in English | MEDLINE | ID: mdl-23734181

ABSTRACT

The intake of ω-3 polyunsaturated fatty acids (PUFAs), which are abundant in marine fish meat and oil, has been shown to exert many beneficial effects. The mechanisms behind those effects are numerous, including interference with the arachidonic acid cascade that produces pro-inflammatory eicosanoids, formation of novel bioactive lipid mediators, and change in the pattern of secreted adipocytokines. In our study, we show that eicosapentaenoic acid (EPA) increases secreted adiponectin from 3T3-L1 adipocytes and in plasma of mice as early as 4 days after initiation of an EPA-rich diet. Using 3T3-L1 adipocytes, we report for the first time that 15-deoxy-δ(12,14)-PGJ3 (15d-PGJ3), a product of EPA, also increases the secretion of adiponectin. We demonstrate that the increased adiponectin secretion induced by 15d-PGJ3 is partially peroxisome proliferator-activated receptor-gamma (PPAR-γ)-mediated. Finally, we show that 3T3-L1 adipocytes can synthesize 15d-PGJ3 from EPA. 15d-PGJ3 was also detected in adipose tissue from EPA-fed mice. Thus, these studies provide a novel mechanism(s) for the therapeutic benefits of ω-3 polyunsaturated fatty acids dietary supplementation.


Subject(s)
Adipocytes/drug effects , Adiponectin/metabolism , PPAR gamma/metabolism , Prostaglandin D2/analogs & derivatives , 3T3-L1 Cells , Adipocytes/metabolism , Adiponectin/blood , Adiponectin/genetics , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Anilides/pharmacology , Animals , Dietary Fats/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Epididymis/drug effects , Epididymis/metabolism , Gas Chromatography-Mass Spectrometry , Male , Mice , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Prostaglandin D2/metabolism , Prostaglandin D2/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
8.
FASEB J ; 27(9): 3860-70, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23756648

ABSTRACT

Environmental contaminants are suspected to be involved in the epidemic incidence of metabolic disorders, food ingestion being a primarily route of exposure. We hypothesized that life-long consumption of a high-fat diet that contains low doses of pollutants will aggravate metabolic disorders induced by obesity itself. Mice were challenged from preconception throughout life with a high-fat diet containing pollutants commonly present in food (2,3,7,8-tetrachlorodibenzo-p-dioxin, polychlorinated biphenyl 153, diethylhexyl phthalate, and bisphenol A), added at low doses in the tolerable daily intake range. We measured several blood parameters, glucose and insulin tolerance, hepatic lipid accumulation, and gene expression in adult mice. Pollutant-exposed mice exhibited significant sex-dependent metabolic disorders in the absence of toxicity and weight gain. In males, pollutants increased the expression of hepatic genes (from 36 to 88%) encoding proteins related to cholesterol biosynthesis and decreased (40%) hepatic total cholesterol levels. In females, there was a marked deterioration of glucose tolerance, which may be related to the 2-fold induction of estrogen sulfotransferase and reduced expression of estrogen receptor α (25%) and estrogen target genes (>34%). Because of the very low doses of pollutants used in the mixture, these findings may have strong implications in terms of understanding the potential role of environmental contaminants in food in the development of metabolic diseases.


Subject(s)
Liver/drug effects , Liver/metabolism , Animals , Benzhydryl Compounds/toxicity , Blotting, Western , Body Weight/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Phenols/toxicity , Polychlorinated Biphenyls/toxicity , Polychlorinated Dibenzodioxins/toxicity , Reverse Transcriptase Polymerase Chain Reaction
9.
PLoS One ; 8(2): e56258, 2013.
Article in English | MEDLINE | ID: mdl-23437106

ABSTRACT

BACKGROUND: Two recent studies demonstrated that bariatric surgery induced remission of type 2 diabetes very soon after surgery and far too early to be attributed to weight loss. In this study, we sought to explore the mechanism/s of this phenomenon by testing the effects of proteins from the duodenum-jejunum conditioned-medium (CM) of db/db or Swiss mice on glucose uptake in vivo in Swiss mice and in vitro in both Swiss mice soleus and L6 cells. We studied the effect of sera and CM proteins from insulin resistant (IR) and insulin-sensitive subjects on insulin signaling in human myoblasts. METHODOLOGY/PRINCIPAL FINDINGS: db/db proteins induced massive IR either in vivo or in vitro, while Swiss proteins did not. In L6 cells, only db/db proteins produced a noticeable increase in basal (473)Ser-Akt phosphorylation, lack of GSK3ß inhibition and a reduced basal (389)Thr-p70-S6K1 phosphorylation. Human IR serum markedly increased basal (473)Ser-Akt phosphorylation in a dose-dependent manner. Human CM IR proteins increased by about twofold both basal and insulin-stimulated (473)Ser-Akt. Basal (9)Ser-GSK3ß phosphorylation was increased by IR subjects serum with a smaller potentiating effect of insulin. CONCLUSIONS: These findings show that jejunal proteins either from db/db mice or from insulin resistant subjects impair muscle insulin signaling, thus inducing insulin resistance.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Insulin Resistance , Insulin/metabolism , Jejunum/metabolism , Proteins/metabolism , Signal Transduction , Adult , Animals , Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Deoxyglucose/metabolism , Diabetes Mellitus, Experimental/enzymology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Phosphorylation/drug effects , Phosphoserine/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
10.
Nutr Metab (Lond) ; 10(1): 23, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23413782

ABSTRACT

BACKGROUND: Dietary intake of n-3 polyunsaturated fatty acids (PUFA) is primarily recognized to protect against cardiovascular diseases, cognitive dysfunctions and the onset of obesity and associated metabolic disorders. However, some of their properties such as bioavailability can depend on their chemical carriers. The objective of our study was to test the hypothesis that the nature of n-3 PUFA carrier results in different metabolic effects related to adiposity, oxidative stress and inflammation. METHODS: 4 groups of C57BL/6 mice were fed for 8 weeks low fat (LF) diet or high-fat (HF, 20%) diets. Two groups of high-fat diets were supplemented with long-chain n-3 PUFA either incorporated in the form of phospholipids (HF-ω3PL) or triacylglycerols (HF-ω3TG). RESULTS: Both HF-ω3PL and HF-ω3TG diets reduced the plasma concentrations of (i) inflammatory markers such as monocyte chemoattractant protein-1 (MCP-1) and interleukin 6 (IL-6), (ii) leptin and (iii) 4-hydroxy-2-nonenal (4-HNE), a marker of n-6 PUFA-derived oxidative stress compared with the control HF diet. Moreover, in both HF-ω3PL and HF-ω3TG groups, MCP-1 and IL-6 gene expressions were decreased in epididymal adipose tissue and the mRNA level of gastrointestinal glutathione peroxidase GPx2, an antioxidant enzyme, was decreased in the jejunum compared with the control HF diet. The type of n-3 PUFA carrier affected other outcomes. The phospholipid form of n-3 PUFA increased the level of tocopherols in epididymal adipose tissue compared with HF-ω3TG and resulted in smaller adipocytes than the two others HF groups. Adipocytes in the HF-ω3PL and LF groups were similar in size distribution. CONCLUSION: Supplementation of mice diet with long-chain n-3 PUFA during long-term consumption of high-fat diets had the same lowering effects on inflammation regardless of triacyglycerol or phospholipid carrier, whereas the location of these fatty acids on a PL carrier had a major effect on decreasing the size of adipocytes that was not observed with the triacyglycerol carrier. Altogether, these results would support the development functional foods containing LC n-3 PUFA in the form of PL in order to prevent some deleterious outcomes associated with the development of obesity.

11.
J Clin Endocrinol Metab ; 98(2): 802-10, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23284008

ABSTRACT

CONTEXT: The hypothesis of a limited expansion of sc adipose tissue during weight gain provides an attractive explanation for the reorientation of excess lipids toward ectopic sites, contributing to visceral adipose depots and metabolic syndrome. OBJECTIVE: Our objective was to define whether the characteristics of sc adipose tissue influence the partition of lipids toward abdominal fat depots during weight gain in healthy men. RESEARCH DESIGN AND METHODS: Forty-one healthy nonobese volunteers performed a 56-day overfeeding protocol (+760 kcal/d). Insulin sensitivity was estimated by euglycemic hyperinsulinemic clamp. Changes in abdominal visceral and sc adipose tissue depots were measured by magnetic resonance imaging. The fate of ingested lipids before and after overfeeding was investigated using a [d31]palmitate test meal, and gene expression was measured by real-time PCR in sc fat biopsies. RESULTS: Overfeeding led to a 2.5-kg body weight increase with large interindividual variations in abdominal sc and visceral adipose tissues. There was no relationship between the relative expansions of these 2 depots, but the increase in visceral depot was positively associated with the magnitude of the postprandial exogenous fatty acid release in the circulation during the test meal. The regulation of lipid storage-related genes (DGAT2, SREBP1c, and CIDEA) was defective in the sc fat of the subjects exhibiting the largest accumulation in visceral depot. CONCLUSIONS: Characteristics of sc adipose tissue appear therefore to contribute to the development of visceral fat depot, supporting the adipose tissue expandability theory and extending it to early stages of weight gain in nonobese subjects.


Subject(s)
Intra-Abdominal Fat/metabolism , Lipid Metabolism/physiology , Overnutrition/metabolism , Subcutaneous Fat/metabolism , Adult , Gene Expression , Glucose Clamp Technique , Humans , Insulin Resistance/physiology , Male , Obesity/metabolism
12.
J Lipid Res ; 53(10): 2069-2080, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22865918

ABSTRACT

Dietary intake of long-chain n-3 PUFA is now widely advised for public health and in medical practice. However, PUFA are highly prone to oxidation, producing potentially deleterious 4-hydroxy-2-alkenals. Even so, the impact of consuming oxidized n-3 PUFA on metabolic oxidative stress and inflammation is poorly described. We therefore studied such effects and hypothesized the involvement of the intestinal absorption of 4-hydroxy-2-hexenal (4-HHE), an oxidized n-3 PUFA end-product. In vivo, four groups of mice were fed for 8 weeks high-fat diets containing moderately oxidized or unoxidized n-3 PUFA. Other mice were orally administered 4-HHE and euthanized postprandially versus baseline mice. In vitro, human intestinal Caco-2/TC7 cells were incubated with 4-hydroxy-2-alkenals. Oxidized diets increased 4-HHE plasma levels in mice (up to 5-fold, P < 0.01) compared with unoxidized diets. Oxidized diets enhanced plasma inflammatory markers and activation of nuclear factor kappaB (NF-κB) in the small intestine along with decreasing Paneth cell number (up to -19% in the duodenum). Both in vivo and in vitro, intestinal absorption of 4-HHE was associated with formation of 4-HHE-protein adducts and increased expression of glutathione peroxidase 2 (GPx2) and glucose-regulated protein 78 (GRP78). Consumption of oxidized n-3 PUFA results in 4-HHE accumulation in blood after its intestinal absorption and triggers oxidative stress and inflammation in the upper intestine.


Subject(s)
Aldehydes/pharmacokinetics , Diet, High-Fat , Fatty Acids, Omega-3/metabolism , Inflammation/metabolism , Intestinal Mucosa/metabolism , Oxidative Stress , Aldehydes/administration & dosage , Animals , Biomarkers/metabolism , Caco-2 Cells , Endoplasmic Reticulum Chaperone BiP , Glutathione Peroxidase/metabolism , Heat-Shock Proteins/metabolism , Humans , Intestinal Absorption/physiology , Lipid Peroxidation , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction
13.
PLoS One ; 7(7): e40306, 2012.
Article in English | MEDLINE | ID: mdl-22808131

ABSTRACT

Dioxins are persistent organic pollutants interfering with endocrine systems and causing reproductive and developmental disorders. The objective of our project was to determine the impact of an in utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on reproductive function of male and female offspring in the rat with a special emphasis on the immature period. We used a low dose of TCDD (unique exposure by oral gavage of 200 ng/kg at 15.5 days of gestation) in order to mirror a response to an environmental dose of TCDD not altering fertility of the progeny. We choose a global gene expression approach using Affymetrix microarray analysis, and testes of 5 days and ovaries of 14 days of age. Less than 1% of the expressed genes in gonads were altered following embryonic TCDD exposure; specifically, 113 genes in ovaries and 56 in testes with 7 genes common to both sex gonads. It included the repressor of the aryl hydrocarbon receptor (Ahrr), the chemokines Ccl5 and Cxcl4 previously shown to be regulated by dioxin in testis, Pgds2/Hpgds and 3 others uncharacterized. To validate and extend the microarray data we realized real-time PCR on gonads at various developmental periods of interest (from 3 to 25 days for ovaries, from 5 to the adult age for testes). Overall, our results evidenced that both sex gonads responded differently to TCDD exposure. For example, we observed induction of the canonic battery of TCDD-induced genes coding enzymes of the detoxifying machinery in ovaries aged of 3-14 days of age (except Cyp1a1 induced at 3-10 days) but not in testes of 5 days (except Ahrr). We also illustrated that inflammatory pathway is one pathway activated by TCDD in gonads. Finally, we identified several new genes targeted by TCDD including Fgf13 in testis and one gene, Ptgds2/Hpgds regulated in the two sex gonads.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Profiling , Ovary/growth & development , Polychlorinated Dibenzodioxins/toxicity , Prenatal Exposure Delayed Effects/genetics , Sex Characteristics , Testis/growth & development , Animals , Animals, Newborn , Chemokines/genetics , Chemokines/metabolism , Crosses, Genetic , Embryo, Mammalian/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Liver/drug effects , Liver/metabolism , Male , Oligonucleotide Array Sequence Analysis , Ovary/drug effects , Ovary/metabolism , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pregnancy , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley , Reproduction/drug effects , Reproduction/genetics , Software , Testis/drug effects , Testis/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects
14.
J Clin Endocrinol Metab ; 97(9): 3326-32, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22745235

ABSTRACT

CONTEXT: Dietary fibers have been associated with a reduced incidence of type 2 diabetes mellitus in epidemiological studies; however, the precise mechanisms are unknown. OBJECTIVE: The objective of the study was to evaluate the efficacy and site of action of an insoluble dietary fiber derived from maize (HAM-RS2) in improving insulin resistance in subjects at increased risk of type 2 diabetes mellitus. DESIGN: This study was a randomized, controlled crossover, dietary intervention study. SETTING: The study was conducted at the Centre for Diabetes, Endocrinology, and Research, Royal Surrey County Hospital, Guildford, United Kingdom. PARTICIPANTS: Fifteen men and women with insulin resistance participated in the study. INTERVENTION: The intervention included 40 g/d HAM-RS2 compared with a matched placebo for 8 wk. MAIN OUTCOME MEASURES: After each supplement, participants underwent a two-step hyperinsulinemic-euglycemic clamp study with the addition of glucose tracers; a meal tolerance test; arteriovenous sampling across forearm muscle tissue; and a sc adipose tissue biopsy for assessment of gene expression. RESULTS: There was enhanced uptake of glucose into the forearm muscle measured by arteriovenous sampling (65 ± 15% increase after resistant starch; P < 0.001). Adipose tissue function was also affected, with enhanced fatty acid suppression after HAM-RS2 treatment and an increase in gene expression for hormone sensitive lipase (P = 0.005), perilipin (P = 0.011), lipoprotein lipase (P = 0.014), and adipose triglyceride lipase (P = 0.03) in biopsy samples. There was no effect on the insulin sensitivity of hepatic glucose production or plasma lipids after HAM-RS2. CONCLUSION: HAM-RS2 improved peripheral but not hepatic insulin resistance and requires further study as an intervention in patients with or at risk for type 2 diabetes.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/metabolism , Dietary Fiber/pharmacology , Insulin Resistance/physiology , Metabolic Syndrome/diet therapy , Metabolic Syndrome/metabolism , Muscles/drug effects , Muscles/metabolism , Adult , Aged , Biomarkers/analysis , Biomarkers/metabolism , Biopsy , Cytokines/metabolism , Diet , Female , Gene Expression/drug effects , Glucose/metabolism , Glucose Clamp Technique , Humans , Lipid Metabolism/drug effects , Liver/metabolism , Male , Membrane Potentials/drug effects , Metabolic Syndrome/physiopathology , Middle Aged , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Single-Blind Method , Zea mays/chemistry
15.
Am J Physiol Endocrinol Metab ; 302(3): E374-86, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22094473

ABSTRACT

Low-grade inflammation observed in obesity is a risk factor for cardiovascular disease. Recent studies revealed that this would be linked to gut-derived endotoxemia during fat digestion in high-fat diets, but nothing is known about the effect of lipid composition. The study was designed to test the impact of oil composition of high-fat diets on endotoxin metabolism and inflammation in mice. C57/Bl6 mice were fed for 8 wk with chow or isocaloric isolipidic diets enriched with oils differing in fatty acid composition: milk fat, palm oil, rapeseed oil, or sunflower oil. In vitro, adipocytes (3T3-L1) were stimulated or not with lipopolysaccharide (LPS; endotoxin) and incubated with different fatty acids. In mice, the palm group presented the highest level of IL-6 in plasma (P < 0.01) together with the highest expression in adipose tissue of IL-1ß and of LPS-sensing TLR4 and CD14 (P < 0.05). The higher inflammation in the palm group was correlated with a greater ratio of LPS-binding protein (LBP)/sCD14 in plasma (P < 0.05). The rapeseed group resulted in higher sCD14 than the palm group, which was associated with lower inflammation in both plasma and adipose tissue despite higher plasma endotoxemia. Taken together, our results reveal that the palm oil-based diet resulted in the most active transport of LPS toward tissues via high LBP and low sCD14 and the greatest inflammatory outcomes. In contrast, a rapeseed oil-based diet seemed to result in an endotoxin metabolism driven toward less inflammatory pathways. This shows that dietary fat composition can contribute to modulate the onset of low-grade inflammation through the quality of endotoxin receptors.


Subject(s)
Adipose Tissue, White/immunology , Cytokines/metabolism , Diet, High-Fat/adverse effects , Metabolic Diseases/etiology , Metabolic Diseases/immunology , Receptors, Immunologic/metabolism , 3T3-L1 Cells , Acute-Phase Proteins , Adipose Tissue, White/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Carrier Proteins/blood , Cytokines/blood , Fatty Acids, Monounsaturated , Fatty Acids, Nonesterified/adverse effects , Fatty Acids, Nonesterified/blood , Gram-Negative Bacteria/immunology , Gram-Negative Bacteria/isolation & purification , Gram-Positive Bacteria/immunology , Gram-Positive Bacteria/isolation & purification , Intestines/immunology , Intestines/microbiology , Intestines/pathology , Lipopolysaccharide Receptors/blood , Lipopolysaccharide Receptors/metabolism , Male , Membrane Glycoproteins/blood , Metabolic Diseases/metabolism , Metabolic Diseases/microbiology , Mice , Mice, Inbred C57BL , Palm Oil , Plant Oils/adverse effects , Random Allocation , Rapeseed Oil , Sunflower Oil , Toll-Like Receptor 4/metabolism
16.
J Clin Endocrinol Metab ; 96(10): E1675-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21816778

ABSTRACT

CONTEXT: GPIHBP1 is a new endothelial binding site for lipoprotein lipase (LPL), the key enzyme for intravascular lipolysis of triglyceride-rich lipoproteins (TGRL). We have identified two new missense mutations of the GPIHBP1 gene, C89F and G175R, by systematic sequencing in a cohort of 376 hyperchylomicronemic patients without mutations on the LPL, APOC2, or APOA5 gene. OBJECTIVE: Phenotypic expression and functional consequences of these two mutations were studied. DESIGN: We performed clinical and genotypic studies of probands and their families. GPIHBP1 functional alterations were studied in CHO pgsA-745 transfected cells. RESULTS: Probands are an adult with a homozygous G175R mutation and a child with a hemizygous C89F neomutation and a deletion of the second allele. C89F mutation was associated with a C14F signal peptide polymorphism on the same haplotype. Both patients had resistant hyperchylomicronemia, low LPL activity, and history of acute pancreatitis. In CHO pgsA-745 cells, both G175R and C14F variants reduce the expression of GPIHBP1 at the cell surface. C89F mutation is responsible for a drastic LPL-binding defect to GPIHBP1. C14F may further potentiate C89F effect. CONCLUSIONS: The emergence of hyperchylomicronemia in the generation after a neomutation further establishes a critical role for GPIHBP1 in TGRL physiopathology in humans. Our results highlight the crucial role of C65-C89 disulfide bond in LPL binding by GPIHBP1 Ly6 domain. Furthermore, we first report a mutation of the hydrophobic C-terminal domain that impairs GPIHBP1 membrane targeting.


Subject(s)
Carrier Proteins/genetics , Chylomicrons/blood , Chylomicrons/genetics , Hyperlipoproteinemia Type I/blood , Hyperlipoproteinemia Type I/genetics , Adult , Animals , Apolipoprotein A-V , Apolipoprotein C-II/genetics , Apolipoprotein C-II/metabolism , Apolipoproteins A/genetics , Apolipoproteins A/metabolism , CHO Cells , Cohort Studies , Cricetinae , Cricetulus , DNA/genetics , Humans , Infant , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Liver/enzymology , Male , Mutation/genetics , Mutation/physiology , Mutation, Missense , Pancreatitis/complications , Pancreatitis/genetics , Pedigree , Receptors, Lipoprotein
17.
J Clin Endocrinol Metab ; 96(6): E962-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21430024

ABSTRACT

CONTEXT: Previous studies suggest a role for fibroblast growth factor receptor 1 (FGFR1) in the regulation of energy balance. OBJECTIVE: Our objective was to investigate whether FGFR1 is an obesity gene by genetic association and functional studies. DESIGN: The study was designed to genotype common FGFR1 single-nucleotide polymorphisms (SNP) in large cohorts, confirm significant results in additional cohorts, and measure FGFR1 expression in human adipose tissue and in rodent hypothalamus. SETTING: General community and referral centers for specialized care was the setting for the study. PARTICIPANTS: We genotyped FGFR1 SNP in 2438 obese and 2115 lean adults and 985 obese and 532 population-based children. Results were confirmed in 928 obese and 2738 population-based adults and 487 obese and 441 lean children. Abdominal sc adipose tissue was investigated in 202 subjects. We also investigated diet-induced, obese fasting, and fed rats. MAIN OUTCOME MEASURES: We analyzed the association between FGFR1 SNP and obesity. In secondary analyses, we related adipose FGFR1 expression to genotype, obesity, and degree of fat cell differentiation and related hypothalamic FGFR1 to energy balance. RESULTS: FGFR1 rs7012413*T was nominally associated with obesity in all four cohorts; metaanalysis odds ratio = 1.17 (95% confidence interval = 1.10-1.25), and P = 1.8 × 10(-6), which was P = 7.0 × 10(-8) in the recessive model. rs7012413*T was associated with FGFR1 expression in adipose tissue (P < 0.0001). In this organ, but not in skeletal muscle, FGFR1 mRNA (P < 0.0001) and protein (P < 0.05) were increased in obesity. In rats, hypothalamic expression of FGFR1 declined after fasting (P < ]0.001) and increased after diet-induced obesity (P < 0.05). CONCLUSIONS: FGFR1 is a novel obesity gene that may promote obesity by influencing adipose tissue and the hypothalamic control of appetite.


Subject(s)
Adipose Tissue/metabolism , Muscle, Skeletal/metabolism , Obesity/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adolescent , Adult , Animals , Appetite/genetics , Blotting, Western , Chi-Square Distribution , Child , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Haplotypes , Humans , Hypothalamus/metabolism , Male , Obesity/metabolism , Odds Ratio , Polymorphism, Single Nucleotide , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , White People
18.
Am J Clin Nutr ; 92(4): 975-84, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20739421

ABSTRACT

BACKGROUND: The mechanisms underlying body weight evolution after diet-induced weight loss are poorly understood. OBJECTIVE: We aimed to identify and characterize differences in the subcutaneous adipose tissue (SAT) transcriptome of subjects with different weight changes after energy restriction-induced weight loss during 6 mo on 4 different diets. DESIGN: After an 8-wk low-calorie diet (800 kcal/d), we randomly assigned weight-reduced obese subjects from 8 European countries to receive 4 diets that differed in protein and glycemic index content. In addition to anthropometric and plasma markers, SAT biopsies were taken at the beginning [clinical investigation day (CID) 2] and end (CID3) of the weight follow-up period. Microarray analysis was used to define SAT gene expression profiles at CID2 and CID3 in 22 women with continued weight loss (successful group) and in 22 women with weight regain (unsuccessful group) across the 4 dietary arms. RESULTS: Differences in SAT gene expression patterns between successful and unsuccessful groups were mainly due to weight variations rather than to differences in dietary macronutrient content. An analysis of covariance with total energy intake as a covariate identified 1338 differentially expressed genes. Cellular growth and proliferation, cell death, cellular function, and maintenance were the main biological processes represented in SAT from subjects who regained weight. Mitochondrial oxidative phosphorylation was the major pattern associated with continued weight loss. CONCLUSIONS: The ability to control body weight loss independent of energy intake or diet composition is reflected in the SAT transcriptome. Although cell proliferation may be detrimental, a greater mitochondrial energy gene expression is suggested as being beneficial for weight control. This trial was registered at clinicaltrials.gov as NCT00390637.


Subject(s)
Adipose Tissue/physiology , Diet, Reducing , Energy Intake/physiology , Gene Expression Profiling , Obesity/diet therapy , Adipose Tissue/physiopathology , Adult , Blood Pressure , Female , Follow-Up Studies , Humans , Models, Genetic , Oligonucleotide Array Sequence Analysis , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Treatment Failure , Treatment Outcome , Weight Loss/physiology
19.
Am J Physiol Endocrinol Metab ; 298(6): E1161-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20233941

ABSTRACT

Apelin, an adipocyte-secreted factor upregulated by insulin, is increased in adipose tissue (AT) and plasma with obesity. Apelin was recently identified as a new player in the control of glucose homeostasis. However, the regulation of apelin and APJ (apelin receptor) expression in skeletal muscle in relation to insulin resistance or type 2 diabetes is not known. Thus we studied apelin and APJ expression in AT and muscle in different mice models of obesity and in type 2 diabetic patients. In insulin-resistant high-fat (HF)-fed mice, apelin and APJ expression were increased in AT compared with control. This was not the case in AT of highly insulin-resistant db/db mice. In skeletal muscle, apelin expression was similar in control and HF-fed mice and decreased in db/db mice. APJ expression was decreased in both HF-fed and db/db mice. Control subjects and type 2 diabetic patients were subjected to a hyperinsulinemic-euglycemic clamp, and tissues biopsies were obtained before and at the end of the clamp. There was no significant difference in basal apelin and APJ expression in AT and muscle between control and diabetic patients. However, apelin plasma levels were significantly increased in diabetic patients. During the clamp, hyperinsulinemia increased apelin and APJ expression in AT of control but not in diabetic subjects. In muscle, only APJ mRNA levels were increased in control but also in diabetic patients. Taken together, these data show that apelin and APJ expression in mice and humans is regulated in a tissue-dependent manner and according to the severity of insulin resistance.


Subject(s)
Adipose Tissue/physiology , Carrier Proteins/biosynthesis , Diabetes Mellitus, Type 2/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Muscle, Skeletal/physiology , Receptors, G-Protein-Coupled/biosynthesis , Adipokines , Adipose Tissue/metabolism , Adult , Animals , Apelin , Apelin Receptors , Carrier Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Female , Gene Expression Regulation , Glucose Clamp Technique , Humans , Insulin/blood , Insulin Resistance/physiology , Intercellular Signaling Peptides and Proteins/genetics , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Middle Aged , Muscle, Skeletal/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
Diabetes ; 59(1): 61-70, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19808894

ABSTRACT

OBJECTIVE: Activation of extracellular signal-regulated kinase-(ERK)-1/2 by cytokines in adipocytes is involved in the alterations of adipose tissue functions participating in insulin resistance. This study aims at identifying proteins regulating ERK1/2 activity, specifically in response to inflammatory cytokines, to provide new insights into mechanisms leading to abnormal adipose tissue function. RESEARCH DESIGN AND METHODS: Kinase activities were inhibited with pharmacological inhibitors or siRNA. Lipolysis was monitored through glycerol production. Gene expression in adipocytes and adipose tissue of obese mice and subjects was measured by real-time PCR. RESULTS: IkappaB kinase-(IKK)-beta inhibition prevented mitogen-activated protein (MAP) kinase kinase (MEK)/ERK1/2 activation in response to interleukin (IL)-1beta and tumor necrosis factor (TNF)-alpha but not insulin in 3T3-L1 and human adipocytes, suggesting that IKKbeta regulated a MAP kinase kinase kinase (MAP3K) involved in ERK1/2 activation induced by inflammatory cytokines. We show that the MAP3K8 called Tpl2 was expressed in adipocytes and that IL-1beta and TNF-alpha activated Tpl2 and regulated its expression through an IKKbeta pathway. Pharmacological inhibition or silencing of Tpl2 prevented MEK/ERK1/2 activation by these cytokines but not by insulin, demonstrating its involvement in ERK1/2 activation specifically in response to inflammatory stimuli. Importantly, Tpl2 was implicated in cytokine-induced lipolysis and in insulin receptor substrate-1 serine phosphorylation. Tpl2 mRNA expression was upregulated in adipose tissue of obese mice and patients and correlated with TNF-alpha expression. CONCLUSIONS: Tpl2 is selectively involved in inflammatory cytokine-induced ERK1/2 activation in adipocytes and is implicated in their deleterious effects on adipocyte functions. The deregulated expression of Tpl2 in adipose tissue suggests that Tpl2 may be a new actor in adipose tissue dysfunction in obesity.


Subject(s)
3T3-L1 Cells/cytology , Adipocytes/cytology , Interleukin-1beta/pharmacology , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins/genetics , Tumor Necrosis Factor-alpha/pharmacology , Adipocytes/physiology , Animals , Cell Differentiation , Enzyme Activation , Fasting , Humans , I-kappa B Kinase/metabolism , Lipolysis , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Obese , Mitogen-Activated Protein Kinase 3/drug effects , Obesity, Morbid/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Thinness , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...