Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Nutr Food Res ; 63(5): e1800788, 2019 03.
Article in English | MEDLINE | ID: mdl-30512227

ABSTRACT

SCOPE: Although about 90% of lycopene in dietary sources occurs in the linear all-trans conformation, a large proportion of the lycopene found in human tissues is of the cis-isomer type, notably (5Z)-lycopene. The biological effects of this (5Z) isomer have been under-researched. The aim of this study is to evaluate some biological functions of (5Z)-lycopene in adipocytes and to compare them with those of (all-E)-lycopene. METHODS AND RESULTS: (all-E)- and (5Z)-Lycopene displayed strong similarities in global gene expression profile and biological pathways impacted. Peroxisome proliferator-activated receptor (PPAR) signaling is identified as a major actor mediating the effects of lycopene isomers. Transactivation assays confirmed the ability of both isomers to transactivate PPARγ. In addition, the TNFα-induced proinflammatory cytokine mRNA expression in 3T3-L1 adipocytes is reduced by both isomers via a reduction in the phosphorylation levels of p65. Finally, lycopene isomers restore the TNF-α-blunted uptake of glucose by adipocytes via a modulation of AKT phosphorylation. CONCLUSION: These results show that lycopene isomers exert similar biological functions in adipocytes, linked to their ability to transactivate PPARγ. These findings add to our knowledge of lycopene effects in adipocyte biology and point to the possible use of lycopene in the prevention of obesity-related disorders.


Subject(s)
Adipocytes/drug effects , Adipocytes/physiology , Lycopene/chemistry , Lycopene/pharmacology , 3T3-L1 Cells , Animals , Cytokines/metabolism , Deoxyglucose/pharmacokinetics , Gene Expression Regulation/drug effects , Isomerism , Mice , NF-kappa B/metabolism , PPAR gamma/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism
2.
J Clin Endocrinol Metab ; 101(4): 1615-26, 2016 04.
Article in English | MEDLINE | ID: mdl-26829440

ABSTRACT

CONTEXT: Obesity alters adipose tissue's metabolic and endocrine functions and causes a chronic local and systemic low-grade inflammatory state to develop, generating obesity-associated complications. In the last decade, many entities contributing to and regulating this inflammatory state have been identified, among which are microRNAs. OBJECTIVE: This study aimed to identify microRNA regulated in inflamed adipocytes and adipose tissue, and its effect on adipocyte biology. DESIGN AND RESULTS: We screened the expression profile of TNFα-treated adipocytes (a major pro-inflammatory protein expressed in obese adipose tissue), and identified miR-155 as the most responsive microRNA. The involvement of TNFα on the basal miR-155 expression was confirmed in the adipose tissue of Tnfa−/− mice where miR-155 was significantly reduced. Also, mice overexpressing p65 or invalidated for p65 in adipose tissue respectively increased and decreased miR-155 expression, in line with the involvement of the nuclear factor κB (NF-κB) pathway in miR-155 induction. miR-155 expression was higher in obese subjects' adipose tissue than in that of normal-weight subjects, and correlated with TNFα expression and body mass index. Gain and loss of function of miR-155 showed its effect on adipocyte function, probably via its ability to target PPARγ mRNA 3'UTR. Interestingly, miR-155 overexpression also resulted in an increased inflammatory state in adipocytes. CONCLUSION: Altogether, these data are evidence of a proinflammatory loop mediated by NF-κB and miR-155 that could participate in the amplification of inflammatory status in adipocytes.


Subject(s)
Adipocytes/pathology , Adipose Tissue/pathology , Inflammation/etiology , MicroRNAs/genetics , Obesity/complications , Adipocytes/metabolism , Adipose Tissue/metabolism , Adult , Animals , Cells, Cultured , Humans , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
3.
J Lipid Res ; 56(6): 1100-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25914170

ABSTRACT

A positive effect of all-trans retinoic acid (ATRA) on white adipose tissue (WAT) oxidative and thermogenic capacity has been described and linked to an in vivo fat-lowering effect of ATRA in mice. However, little is known about the effects of ATRA on mitochondria in white fat. Our objective has been to characterize the effect of ATRA on mitochondria biogenesis and oxidative phosphorylation (OXPHOS) capacity in mature white adipocytes. Transcriptome analysis, oxygraphy, analysis of mitochondrial DNA (mtDNA), and flow cytometry-based analysis of mitochondria density were performed in mature 3T3-L1 adipocytes after 24 h incubation with ATRA (2 µM) or vehicle. Selected genes linked to mitochondria biogenesis and function and mitochondria immunostaining were analyzed in WAT tissues of ATRA-treated as compared with vehicle-treated mice. ATRA upregulated the expression of a large set of genes linked to mtDNA replication and transcription, mitochondrial biogenesis, and OXPHOS in adipocytes, as indicated by transcriptome analysis. Oxygen consumption rate, mtDNA content, and staining of mitochondria were increased in the ATRA-treated adipocytes. Similar results were obtained in WAT depots of ATRA-treated mice. We conclude that ATRA impacts mitochondria in adipocytes, leading to increased OXPHOS capacity and mitochondrial content in these cells.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondrial Proteins/biosynthesis , Organelle Biogenesis , Tretinoin/metabolism , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , DNA, Mitochondrial/metabolism , Gene Expression Profiling , Gene Expression Regulation , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation
4.
Adipocyte ; 3(3): 180-9, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-25068084

ABSTRACT

Tumor necrosis factor α (TNFα) is a well-known mediator of inflammation in the context of obesity in adipose tissue. Its action appears to be directly linked to perturbations of the insulin pathway, leading to the development of insulin resistance. Visfatin has been suspected to be linked to insulin sensitivity, but the mechanism involved is still partly unknown. The aim of this study was to evaluate the role of visfatin in the impairment of the insulin pathway by TNFα activity in 3T3-L1 adipocytes and to unveil the mechanisms involved in such impairment. We demonstrated in 3T3-L1 adipocytes that visfatin was involved in TNFα-mediated insulin resistance in adipocytes. Indeed, after TNFα treatment in 3T3-L1 cells, visfatin was downregulated, leading to decreased nicotinamide adenine dinucleotide (NAD(+)) concentrations in cells. This decrease was followed by a decrease in Sirt1 activity, which was linked to an increase in PTP1B expression. The modulation of PTP1B by visfatin was likely responsible for the observed decreases in glucose uptake and Akt phosphorylation in 3T3-L1 adipocytes. Here, we demonstrated a complete pathway involving visfatin, NAD(+), Sirt1, and PTP1B that led to the perturbation of insulin signaling by TNFα in 3T3-L1 adipocytes.

5.
PLoS One ; 8(12): e83300, 2013.
Article in English | MEDLINE | ID: mdl-24367589

ABSTRACT

Epigenetic post-transcriptional modifications of histone tails are thought to help in coordinating gene expression during development. An epigenetic signature is set in pluripotent cells and interpreted later at the onset of differentiation. In pluripotent cells, epigenetic marks normally associated with active genes (H3K4me3) and with silent genes (H3K27me3) atypically co-occupy chromatin regions surrounding the promoters of important developmental genes. However, it is unclear how these epigenetic marks are recognized when cell differentiation starts and what precise role they play. Here, we report the essential role of the nuclear receptor peroxisome proliferator-activated receptor ß (PPARß, NR1C2) in Xenopus laevis early development. By combining loss-of-function approaches, large throughput transcript expression analysis by the mean of RNA-seq and intensive chromatin immunoprecipitation experiments, we unveil an important cooperation between epigenetic marks and PPARß. During Xenopus laevis gastrulation PPARß recognizes H3K27me3 marks that have been deposited earlier at the pluripotent stage to activate early differentiation genes. Thus, PPARßis the first identified transcription factor that interprets an epigenetic signature of pluripotency, in vivo, during embryonic development. This work paves the way for a better mechanistic understanding of how the activation of hundreds of genes is coordinated during early development.


Subject(s)
Cell Differentiation/genetics , Chromatin/genetics , Epigenesis, Genetic , Gastrulation/genetics , PPAR-beta/metabolism , Animals , Base Sequence , Blastula/cytology , Blastula/embryology , Gene Knockdown Techniques , Histones/chemistry , Histones/metabolism , Lysine/metabolism , Methylation , PPAR-beta/deficiency , PPAR-beta/genetics , RNA, Messenger/genetics , Transcriptome , Xenopus laevis/embryology , Xenopus laevis/genetics
6.
PLoS One ; 8(6): e66515, 2013.
Article in English | MEDLINE | ID: mdl-23824685

ABSTRACT

Immune cell infiltration of expanding adipose tissue during obesity and its role in insulin resistance has been described and involves chemokines. However, studies so far have focused on a single chemokine or its receptor (especially CCL2 and CCL5) whereas redundant functions of chemokines have been described. The objective of this work was to explore the expression of chemokines in inflamed adipose tissue in obesity. Human and mouse adipocytes were analyzed for expression of chemokines in response to inflammatory signal (TNF-α) using microarrays and gene set enrichment analysis. Gene expression was verified by qRT-PCR. Chemokine protein was determined in culture medium with ELISA. Chemokine expression was investigated in human subcutaneous adipose tissue biopsies and mechanism of chemokine expression was investigated using chemical inhibitors and cellular and animal transgenic models. Chemokine encoding genes were the most responsive genes in TNF-α treated human and mouse adipocytes. mRNA and protein of 34 chemokine genes were induced in a dose-dependent manner in the culture system. Furthermore, expression of those chemokines was elevated in human obese adipose tissue. Finally, chemokine expression was reduced by NF-κB inactivation and elevated by NF-κB activation. Our data indicate that besides CCL2 and CCL5, numerous other chemokines such as CCL19 are expressed by adipocytes under obesity-associated chronic inflammation. Their expression is regulated predominantly by NF-κB. Those chemokines could be involved in the initiation of infiltration of leukocytes into obese adipose tissue.


Subject(s)
Adipose Tissue/metabolism , Chemokines/metabolism , NF-kappa B/metabolism , Adipose Tissue/cytology , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Obesity/metabolism , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
7.
Metallomics ; 5(5): 524-31, 2013 May.
Article in English | MEDLINE | ID: mdl-23503329

ABSTRACT

The white adipose tissue plays a major role in the development of obesity and associated metabolic complications by producing a variety of pro and anti-inflammatory adipokines. Recently, studies in humans or in animals have shown a beneficial effect of certain trace elements such as zinc on insulin resistance and adipokine secretion. The aim of our study was to test the effect of a zinc-nickel-cobalt solution (ZnNiCo) on adipocyte function and to identify potential health effects of this solution in the context of obesity and associated disorders. No impact of ZnNiCo on adipogenesis was observed in 3T3-L1 cells. Gene expression in murine and human adipocytes was examined in the presence of ZnNiCo using whole genome microarrays. This transcriptomic analysis indicated that ZnNiCo affected the expression levels of genes in adipocytes under basal conditions or incubated with TNF-α and showed a down regulation of several inflammatory genes belonging to the cytokine and chemokine families (P < 0.01). These data were confirmed in mice fed with a high fat diet supplemented with ZnNiCo (P < 0.05). A modulation of NF-κB activation (evaluated by ELISA; P < 0.05) by ZnNiCo could explain at least in part these observations. The trace elements present in ZnNiCo are able to modulate the expression level of several inflammation related transcripts in adipocytes. These studies suggest that ZnNiCo could play a role in the prevention of inflammation in adipose tissue in obesity.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Trace Elements/pharmacology , 3T3-L1 Cells , Adipocytes/pathology , Adipogenesis/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Chemokines/genetics , Chemokines/metabolism , Diet, High-Fat , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Response Elements/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Solutions , Transcriptome/genetics , Tumor Necrosis Factor-alpha/pharmacology
8.
Cell Biosci ; 2(1): 34, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-23046570

ABSTRACT

BACKGROUND: After liver injury, the repair process comprises activation and proliferation of hepatic stellate cells (HSCs), which produce extracellular matrix (ECM) proteins. Peroxisome proliferator-activated receptor beta/delta (PPARß/δ) is highly expressed in these cells, but its function in liver repair remains incompletely understood. This study investigated whether activation of PPARß/δ with the ligand GW501516 influenced the fibrotic response to injury from chronic carbon tetrachloride (CCl4) treatment in mice. Wild type and PPARß/δ-null mice were treated with CCl4 alone or CCl4 co-administered with GW501516. To unveil mechanisms underlying the PPARß/δ-dependent effects, we analyzed the proliferative response of human LX-2 HSCs to GW501516 in the presence or absence of PPARß/δ. RESULTS: We found that GW501516 treatment enhanced the fibrotic response. Compared to the other experimental groups, CCl4/GW501516-treated wild type mice exhibited increased expression of various profibrotic and pro-inflammatory genes, such as those involved in extracellular matrix deposition and macrophage recruitment. Importantly, compared to healthy liver, hepatic fibrotic tissues from alcoholic patients showed increased expression of several PPAR target genes, including phosphoinositide-dependent kinase-1, transforming growth factor beta-1, and monocyte chemoattractant protein-1. GW501516 stimulated HSC proliferation that caused enhanced fibrotic and inflammatory responses, by increasing the phosphorylation of p38 and c-Jun N-terminal kinases through the phosphoinositide-3 kinase/protein kinase-C alpha/beta mixed lineage kinase-3 pathway. CONCLUSIONS: This study clarified the mechanism underlying GW501516-dependent promotion of hepatic repair by stimulating proliferation of HSCs via the p38 and JNK MAPK pathways.

9.
Mol Nutr Food Res ; 56(12): 1771-82, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23065818

ABSTRACT

SCOPE: Obesity is strongly associated with low-grade inflammation, notably due to an overproduction of proinflammatory markers by adipose tissue and adipocytes as well as a vitamin D deficiency. Whether these problems are interrelated has not been clearly established. METHODS AND RESULTS: In the present report, decreases in the levels of inflammatory markers such as IL-6, MCP-1, and IL-1ß (mRNA and protein level) in human adipocytes and in 3T3-L1 adipocytes were observed after 1,25-dihydroxyvitamin D3 (1,25-(OH)(2) D(3) ) treatment. Such treatment also decreased the expression of the TNF-α-mediated proinflammatory marker in 3T3-L1 and human adipocytes. A similar effect was observed in adipocyte-macrophage co-culture systems in which 1,25-(OH)(2) D(3) decreased proinflammatory marker expression under basal and TNF-α-stimulated conditions. The involvement of VDR and NF-κB was confirmed in these regulations. Incubation with 1,25-(OH)(2) D(3) also resulted in the dephosphorylation of p38, which is linked to the transcriptional induction of several Dusp family members. Functional consequences of the 1,25-(OH)(2) D(3) treatment on glucose uptake and AKT phosphorylation were observed. CONCLUSION: The improvement of both proinflammatory status and glucose uptake in adipocytes under 1,25-(OH)(2) D(3) effect suggests that low-grade inflammation could be linked to vitamin D deficiency. This observation offers new perspectives in the context of obesity and associated physiopathological disorders.


Subject(s)
Adipocytes/cytology , Glucose/metabolism , Inflammation/metabolism , Vitamin D/pharmacology , Vitamins/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Biomarkers/blood , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokines/genetics , Chemokines/metabolism , Coculture Techniques , Down-Regulation , Humans , Insulin Resistance , Interleukin-1beta/blood , Interleukin-1beta/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Mol Nutr Food Res ; 56(5): 725-32, 2012 May.
Article in English | MEDLINE | ID: mdl-22648619

ABSTRACT

SCOPE: Adipose tissue is infiltrated by an increasing number of macrophages during the development of obesity. These immune cells are suspected to be a major source of TNF-α that interferes with adipocyte function. Because lycopene possesses anti-inflammatory properties, we hypothesize that lycopene could reduce the production of TNF-α by macrophages and thus interfere in the cross-talk between macrophages and adipocytes. METHODS AND RESULTS: We demonstrated that physiological concentrations of lycopene were able to attenuate the lipopolysaccharide (LPS)-mediated induction of TNF-α in RAW 264.7 macrophages, at both the mRNA and protein levels. The molecular mechanism was studied. It appeared that the LPS-activation of both JNK and NF-κB signaling pathways was modulated by lycopene. The anti-inflammatory effects of lycopene on macrophages were accompanied by a decrease in LPS-stimulated macrophage migration in the presence of lycopene. Furthermore, lycopene decreased macrophage conditioned medium-induced proinflammatory cytokine, acute phase protein, and chemokine mRNA expression in 3T3-L1 adipocytes. CONCLUSION: These data indicate that lycopene displayed an anti-inflammatory effect on macrophages that beneficially impacted adipocyte function. Thus, these results suggest that lycopene could block the vicious cycle that occurs between adipocytes and macrophages in adipose tissue during obesity.


Subject(s)
Adipocytes/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carotenoids/pharmacology , Inflammation/metabolism , Macrophages/drug effects , Macrophages/metabolism , Tumor Necrosis Factor-alpha/metabolism , 3T3-L1 Cells/drug effects , Adipocytes/metabolism , Animals , Biomarkers/metabolism , Cell Movement/drug effects , Cells, Cultured , Chemokines/genetics , Culture Media, Conditioned/pharmacology , Cytokines/genetics , Cytokines/metabolism , Inflammation/drug therapy , Lipopolysaccharides/toxicity , Lycopene , MAP Kinase Signaling System/drug effects , Mice , Tumor Necrosis Factor-alpha/genetics
11.
PLoS One ; 7(1): e29837, 2012.
Article in English | MEDLINE | ID: mdl-22253797

ABSTRACT

BACKGROUND: Polyphenols are the most abundant antioxidants in the human diet and are widespread constituents of fruits and beverages, such as tea, coffee or wine. Epidemiological, clinical and animal studies support a role of polyphenols in the prevention of various diseases, such as cardiovascular diseases, cancers or neurodegenerative diseases. Recent findings suggest that polyphenols could interact with cellular signaling cascades regulating the activity of transcription factors and consequently affecting the expression of genes. However, the impact of polyphenol on the expression of microRNA, small non-coding RNAs, has not yet been studied. The aim of this study was to investigate the impact of dietary supplementation with polyphenols at nutritional doses on miRNA expression in the livers of apolipoprotein E-deficient mice (apoE⁻/⁻) jointly with mRNA expression profiling. METHODOLOGY/PRINCIPAL FINDINGS: Using microarrays, we measured the global miRNA expression in the livers of wild-type (C57B6/J) mice or apoE⁻/⁻ mice fed diets supplemented with one of nine different polyphenols or a control diet. This analysis revealed that knock-out of the apoE gene induced significant modulation in the expression of miRNA. Moreover, changes in miRNA expression were observed after polyphenol supplementation, and five miRNAs (mmu-miR-291b-5p, mmu-miR-296-5p, mmu-miR-30c-1*, mmu-miR-467b* and mmu-miR-374*) were identified as being commonly modulated by these polyphenols. We also observed that these polyphenols counteracted the modulation of miRNA expression induced by apoE mutation. Pathway analyses on these five miRNA-target genes revealed common pathways, some of which were also identified from a pathway analysis on mRNA profiles. CONCLUSION: This in vivo study demonstrated for the first time that polyphenols at nutritional doses modulate the expression of miRNA in the liver. Even if structurally different, all polyphenols induced a similar miRNA expression profile. Common pathways were identified from both miRNA-target and mRNA analysis, revealing cellular functions that could be regulated by polyphenols at both the miRNA and mRNA level.


Subject(s)
Apolipoproteins E/deficiency , Diet , Gene Expression Regulation/drug effects , MicroRNAs/genetics , Polyphenols/pharmacology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/genetics , Animals , Apolipoproteins E/metabolism , Cluster Analysis , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Profiling , Humans , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
12.
PLoS One ; 6(6): e20644, 2011.
Article in English | MEDLINE | ID: mdl-21673813

ABSTRACT

Evidence from cell culture studies indicates that ß-carotene-(BC)-derived apocarotenoid signaling molecules can modulate the activities of nuclear receptors that regulate many aspects of adipocyte physiology. Two BC metabolizing enzymes, the BC-15,15'-oxygenase (Bcmo1) and the BC-9',10'-oxygenase (Bcdo2) are expressed in adipocytes. Bcmo1 catalyzes the conversion of BC into retinaldehyde and Bcdo2 into ß-10'-apocarotenal and ß-ionone. Here we analyzed the impact of BC on body adiposity of mice. To genetically dissect the roles of Bcmo1 and Bcdo2 in this process, we used wild-type and Bcmo1(-/-) mice for this study. In wild-type mice, BC was converted into retinoids. In contrast, Bcmo1(-/-) mice showed increased expression of Bcdo2 in adipocytes and ß-10'-apocarotenol accumulated as the major BC derivative. In wild-type mice, BC significantly reduced body adiposity (by 28%), leptinemia and adipocyte size. Genome wide microarray analysis of inguinal white adipose tissue revealed a generalized decrease of mRNA expression of peroxisome proliferator-activated receptor γ (PPARγ) target genes. Consistently, the expression of this key transcription factor for lipogenesis was significantly reduced both on the mRNA and protein levels. Despite ß-10'-apocarotenoid production, this effect of BC was absent in Bcmo1(-/-) mice, demonstrating that it was dependent on the Bcmo1-mediated production of retinoids. Our study evidences an important role of BC for the control of body adiposity in mice and identifies Bcmo1 as critical molecular player for the regulation of PPARγ activity in adipocytes.


Subject(s)
Adiposity/drug effects , beta Carotene/pharmacology , beta-Carotene 15,15'-Monooxygenase/metabolism , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Animals , Dietary Supplements , Dioxygenases , Down-Regulation/drug effects , Female , Mice , Mice, Inbred C57BL , Oxygenases/genetics , Oxygenases/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Retinoids/blood , Retinoids/metabolism , beta-Carotene 15,15'-Monooxygenase/genetics
13.
Mol Nutr Food Res ; 55(4): 578-84, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21462325

ABSTRACT

SCOPE: Carotenoids are mainly stored in adipose tissue. However, nothing is known regarding the uptake of carotenoids by adipocytes. Thus, our study explored the mechanism by which lycopene and lutein, two major human plasma carotenoids, are transported. METHODS AND RESULTS: CD36 was a putative candidate for this uptake, 3T3-L1 cells were treated with sulfosuccinimidyl oleate, a CD36-specific inhibitor. sulfosuccinimidyl oleate-treated cells showed a significant decrease in both lycopene and lutein uptake as compared to control cells. Their uptake was also decreased by partial inhibition of CD36 expression using siRNA, whereas the overexpression of CD36 in Cos-1 cells increased their uptake. Finally, the effect of CD36 on carotenoid uptake was confirmed ex vivo in cultures of adipose tissue explants from CD36(-/-) mice, which exhibited reduced carotenoid uptake as compared to wild-type mice explants. CONCLUSION: For the first time, we report the involvement of a transporter, CD36, in carotenoid uptake by adipocytes and adipose tissue.


Subject(s)
Adipose Tissue, White/metabolism , CD36 Antigens/physiology , Carotenoids/metabolism , Lutein/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, White/drug effects , Animals , Biological Transport/drug effects , CD36 Antigens/chemistry , CD36 Antigens/genetics , COS Cells , Chlorocebus aethiops , Humans , Lycopene , Male , Mice , Mice, Knockout , Oleic Acids/pharmacology , Organ Culture Techniques , RNA Interference , RNA, Small Interfering , Succinimides/pharmacology
14.
J Nutr Biochem ; 21(12): 1207-13, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20149624

ABSTRACT

Intestine is the gateway for newly absorbed tocopherols. This organ also plays a crucial role in cholesterol metabolism. Because tocopherols are known to impact cholesterol metabolism in the liver, we hypothesized that tocopherols could also modulate cholesterol metabolism in the intestine. This study aimed to verify this hypothesis and to unveil the mechanisms involved, using Caco-2 cells as a model of the human intestinal cell. Both α- and γ-tocopherol significantly (P<.05) decreased endogenous cholesterol synthesis and apo-AI-mediated cholesterol secretion in Caco-2 cells. Tocopherols down-regulated (P<.05) up to half of the genes involved in the cholesterol synthesis pathway, together with CYP27A1, which is involved in oxysterol production. The activity of this enzyme, as well as the levels of intracellular oxysterols, was significantly diminished by tocopherols. Finally, tocopherols significantly reduced ABCA1 mRNA levels in Caco-2 cells. We conclude that tocopherols impair the endogenous synthesis and apo-AI-mediated secretion of cholesterol in Caco-2 cells. This effect involves a down-regulation of genes involved in the cholesterol synthesis pathway, resulting in down-regulation of CYP27A1 which, in turn, diminishes oxysterol concentrations. The outcome is a decrease of LXR activity, resulting in down-regulation of ABCA1. These data reinforce the effect of α- and γ-tocopherol on cholesterol metabolism via gene expression regulation.


Subject(s)
Antioxidants/pharmacology , Apolipoprotein A-I/metabolism , Cholesterol/biosynthesis , alpha-Tocopherol/pharmacology , gamma-Tocopherol/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Caco-2 Cells , Cholestanetriol 26-Monooxygenase/genetics , Cholestanetriol 26-Monooxygenase/metabolism , Cholesterol/genetics , Cholesterol/metabolism , Down-Regulation , Humans , Intestinal Mucosa/metabolism , Microarray Analysis
15.
Endocrinology ; 150(12): 5318-25, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19833717

ABSTRACT

Adiponectin is a well-known adipokine secreted by adipocytes that presents insulin-sensitizing properties. The regulation of expression of this adipokine by micronutrients is largely unknown. We demonstrate here that adiponectin expression is induced in adipocytes after exposure to tocopherols via the peroxisome proliferator-activated receptor gamma (PPARgamma) pathway. Vitamin E force feeding resulted in an induction of adiponectin in mice at both mRNA and protein levels. Adiponectin mRNA and protein secretion were also increased by vitamin E (alpha- and gamma-tocopherol) in 3T3-L1 cells, together with PPARgamma mRNA, independent of an antioxidant effect. In transient transfections, both alpha- and gamma-vitamers induced the luciferase gene reporter under the control of a human adiponectin promoter via a PPAR-responsive element. The induction of adiponectin by tocopherols seems to be PPARgamma dependent, because it was blocked by the specific antagonist GW9662. Finally, we showed that intracellular concentrations of a PPARgamma endogenous ligand, 15-deoxy-Delta12,14-prostaglandin J2, increased after treatment with tocopherols in 3T3-L1 cells. In summary, vitamin E up-regulates adiponectin expression via a mechanism that implicates PPARgamma together with its endogenous ligand 15-deoxy-Delta12,14-prostaglandin J2. The induction of adiponectin via an original molecular mechanism could be considered as the basis for the beneficial effect of vitamin E on insulin sensitivity.


Subject(s)
Adipocytes/drug effects , Adiponectin/genetics , PPAR gamma/metabolism , Vitamin E/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adiponectin/blood , Adiponectin/metabolism , Anilides/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , Promoter Regions, Genetic/genetics , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transfection , Vitamin E/administration & dosage , Vitamins/administration & dosage , Vitamins/pharmacology
16.
Genes Nutr ; 4(3): 179-87, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19557453

ABSTRACT

Recent epidemiological data suggest that beta-carotene may be protective against metabolic diseases in which adipose tissue plays a key role. Adipose tissue constitutes the major beta-carotene storage tissue and its functions have been shown to be modulated in response to beta-carotene breakdown products, especially retinal produced after cleavage by beta-carotene 15,15'-monooxygenase (BCMO1), and retinoic acid arising from oxidation of retinal. However, the possibility exists that beta-carotene in its intact form can also affect adipocyte function. Development of a knock out model and identification of a loss-of-function mutation have pointed out BCMO1 as being probably the sole enzyme responsible for provitamin A conversion into retinal in mammals. The utilisation of BCMO1(-/-)mice should provide insights on beta-carotene effect on its own in the future. In humans, intervention studies have highlighted the huge interindividual variation of beta-carotene conversion efficiency, possibly due to genetic polymorphisms, which might impact on response to beta-carotene. This brief review discusses the processes involved in beta-carotene conversion and the effect of cleavage products on body fat and adipose tissue function.

17.
Food Chem Toxicol ; 46(12): 3832-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18957315

ABSTRACT

Epidemiological studies have suggested that lycopene has protective effects against various diseases including cardiovascular diseases. However, mechanistic studies to understand these effects are difficult due to the insolubility of lycopene in aqueous culture medium. The objective of the present study was to use LDL or BSA as physiological vehicles for lycopene and to compare them with various classical vehicles. Among tested vehicles, only LDL, BSA, THF/BHT, beadlets, and liposomes were able to solubilise lycopene. No cytotoxicity was observed with these vehicles. LDL and BSA allowed good stability of lycopene during incubation (52% and 43% for 2microM lycopene solutions), but remained less efficient than THF/BHT or beadlets (67% and 62%). Incubation of adipocytes (3T3-L1) with the different vehicles for 24 and 48h showed that beadlets best delivered lycopene to cells. Finally, whatever the vehicle used, intracellular localization of lycopene was the same: lipid droplets (32-51%), plasma membrane (32-37%) and nuclear membrane (19-29%). As a conclusion, LDL or BSA display comparable properties to THF/BHT or beadlets. It is the first time that lycopene carried by physiological vehicles is shown to reach different subcellular compartments supporting molecular effects in adipocyte, such as cell signaling or nuclear receptor interacting.


Subject(s)
Adipocytes/metabolism , Carotenoids/metabolism , Lipoproteins, LDL/pharmacology , Serum Albumin/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Cell Survival/drug effects , Culture Media , Drug Delivery Systems , L-Lactate Dehydrogenase/metabolism , Lipoproteins, LDL/toxicity , Lycopene , Mice , Oxidation-Reduction , Pharmaceutical Vehicles , Serum Albumin/toxicity , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...