Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Int J Obes (Lond) ; 41(6): 917-925, 2017 06.
Article in English | MEDLINE | ID: mdl-28280270

ABSTRACT

BACKGROUND/OBJECTIVES: Circulating phospholipids and sphingolipids are implicated in obesity-related comorbidities such as insulin resistance and cardiovascular disease. How bariatric surgery affects these important lipid markers is poorly understood. We sought to determine whether Roux-en-Y gastric bypass (RYGB), which is associated with greater metabolic improvement, differentially affects the phosphosphingolipidome compared with adjustable gastric banding (AGB). SUBJECTS/METHODS: Fasting sera were available from 59 obese women (body mass index range 37-51 kg m-2; n=37 RYGB and 22 AGB) before surgery, then at 1 (21 RYGB, 12 AGB) and 3 months follow-up (19 RYGB, 12 AGB). HPLC-MS/MS was used to quantify 131 lipids from nine structural classes. DXA measurements and laboratory parameters were also obtained. The associations between lipids and clinical measurements were studied with P-values adjusted for the false discovery rate (FDR). RESULTS: Both surgical procedures rapidly induced weight loss and improved clinical profiles, with RYGB producing better improvements in fat mass, and serum total cholesterol, low-density lipoprotein-cholesterol (LDL-C) and orosomucoid (FDR <10%). Ninety-three (of 131) lipids were altered by surgery-the majority decreasing-with 29 lipids differentially affected by RYGB during the study period. The differential effect of the surgeries remained statistically significant for 20 of these lipids after adjusting for differences in weight loss between surgery types. The RYGB signature consisted of phosphatidylcholine species not exceeding 36 carbons, and ceramides and sphingomyelins containing C22 to C25 fatty acids. RYGB also led to a sustained increase in unsaturated ceramide and sphingomyelin species. The RYGB-specific lipid changes were associated with decreases in body weight, total and LDL-C, orosomucoid and increased HOMA-S (FDR <10%). CONCLUSIONS: Concomitant with greater metabolic improvement, RYGB induced early and sustained changes in phosphatidylcholines, sphingomyelins and ceramides that were independent of greater weight loss. These data suggest that RYGB may specifically alter sphingolipid metabolism, which, in part, could explain the better metabolic outcomes of this surgical procedure.


Subject(s)
Gastric Bypass , Gastroplasty , Obesity, Morbid/surgery , Phospholipids/blood , Sphingolipids/blood , Weight Loss/physiology , Adult , Biomarkers/blood , Ceramides/blood , Cholesterol/blood , Fasting/blood , Female , Follow-Up Studies , France , Humans , Lipid Metabolism , Obesity, Morbid/blood , Postoperative Period , Prospective Studies , Treatment Outcome
2.
Cell Mol Life Sci ; 64(19-20): 2452-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17876522

ABSTRACT

Lipid droplets have been considered for a long time as inert intracytoplasmic deposits formed within cells under various conditions. Recently, new tools and new approaches have been used to visualize and study these intracellular structures. This revealed new aspects of lipid droplets biology and pointed out their organized structure and dynamic composition. In adipocytes, the specialized cell type for the storage of energy as fat, lipid droplets are particularly well-developed organelles and exhibit unique properties. Also discussed in this paper is the view that lipid droplets, through specific candidate constituents, can play a role in sensing the level of their lipid stores by adipocytes.


Subject(s)
Adipocytes/metabolism , Lipid Metabolism , Animals , Fat Emulsions, Intravenous , Humans , Lipids , Organelles/metabolism , Triglycerides/metabolism
3.
Mol Cell Biol ; 25(21): 9621-31, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16227610

ABSTRACT

Caspases play important roles in apoptotic cell death and in some other functions, such as cytokine maturation, inflammation, or differentiation. We show here that the 5'-flanking region of the human CASP-2 gene contains three functional response elements for sterol regulatory element binding proteins (SREBPs), proteins that mediate the transcriptional activation of genes involved in cholesterol, triacylglycerol, and fatty acid synthesis. Exposure of several human cell lines to statins, lipid-lowering drugs that drive SREBP proteolytic activation, induced the CASP-2 gene to an extent similar to that for known targets of SREBP proteins. Adenoviral vector-mediated transfer of active SREBP-2 also induced expression of the CASP-2 gene and the caspase-2 protein and increased the cholesterol and triacylglycerol cellular content. These rises in lipids were strongly impaired following small interfering RNA-mediated silencing of the CASP-2 gene. Taken together, our results identify the human CASP-2 gene as a member of the SREBP-responsive gene battery that senses lipid levels in cells and raise the possibility that caspase-2 participates in the control of cholesterol and triacylglycerol levels.


Subject(s)
Cysteine Endopeptidases/physiology , Sterol Regulatory Element Binding Protein 2/physiology , 5' Flanking Region , Binding Sites , Caspase 2 , Cell Line, Tumor , Cholesterol/biosynthesis , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/genetics , Gene Expression Regulation , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Response Elements , Sterol Regulatory Element Binding Protein 2/genetics , Triglycerides/biosynthesis
4.
Ann Pharm Fr ; 62(2): 87-91, 2004 Mar.
Article in French | MEDLINE | ID: mdl-15107725

ABSTRACT

During the last past Years, obesity had become a major public health problem, and new aspects of fat cells biology have been unraveled. First, since the discovery of leptin, adipocytes have been recognized as true endocrine cells secreting a variety of factors in a regulated manner. The role of these factors on the development of obesity-associated metabolic complications is becoming increasingly clear. Also, the process of fat cell differentiation has been uncovered, leading to the possibility of efficient targeting protein expression in adipose tIssue. Finally, lines of transgenic mice have been created, some of which are totally resistant to obesity. These models led to the identification of new potential adipose targets for the treatment of obesity.


Subject(s)
Adipocytes/pathology , Adipose Tissue/pathology , Obesity/pathology , Adipocytes/physiology , Adipose Tissue/physiopathology , Cell Differentiation/physiology , Endocrine Glands/physiology , Humans , Lipid Metabolism , Obesity/physiopathology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology
5.
Biochem Soc Trans ; 32(Pt 1): 103-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14748723

ABSTRACT

Adipose tissue is specialized in the storage of energy in the form of triacylglycerol. Within the fat cell, triacylglycerols are found in a well-defined structural compartment called the lipid droplet, which occupies the vast majority of the fat cell volume. However, many other lipids are present in the lipid droplet. These include sterols, carotenoids, cholecalciferol and lipophilic toxic pollutants of the environment such as dioxins and tocopherols. The topic of this article is the role of fat cell cholesterol in adipose tissue physiology and its potential implication in pathological states such as obesity.


Subject(s)
Adipose Tissue/metabolism , Cholesterol/metabolism , Obesity/metabolism , Adipose Tissue/pathology , Animals , DNA-Binding Proteins/metabolism , Humans , Obesity/pathology , Sterol Regulatory Element Binding Protein 2 , Transcription Factors/metabolism , Triglycerides/metabolism
6.
Horm Metab Res ; 35(4): 204-10, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12778362

ABSTRACT

In recent years, our view of adipose tissue has evolved from a passive sink for energy storage to an active tissue producing multiple molecules acting on various tissues in different aspects of energy homeostasis. The production of adipose-derived secretory products is tightly regulated as a function of adipocyte lipid accumulation, but the mechanisms by which fat cells are able to sense the levels of their triglyceride stores still remains largely unknown. This paper reviews new insights into this question taking cholesterol as a potential intracellular signaling molecule.


Subject(s)
Adipocytes/chemistry , Adipocytes/physiology , Cholesterol/metabolism , Signal Transduction/physiology , Triglycerides/physiology , Adipocytes/cytology , Cell Size/physiology , Cholesterol/physiology , DNA-Binding Proteins/physiology , Humans , Obesity/physiopathology , Sterol Regulatory Element Binding Protein 2 , Sterols/chemistry , Transcription Factors/physiology
7.
J Mol Endocrinol ; 29(2): 251-64, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12370125

ABSTRACT

Catecholamines regulate white adipose tissue function and development by acting through beta- and alpha2-adrenergic receptors (ARs). Human adipocytes express mainly alpha 2A- but few or no beta 3-ARs while the reverse is true for rodent adipocytes. Our aim was to generate a mouse model with a human-like alpha2/beta-adrenergic balance in adipose tissue by creating transgenic mice harbouring the human alpha 2A-AR gene under the control of its own regulatory elements in a combined mouse beta 3-AR-/- and human beta 3-AR+/+ background. Transgenic mice exhibit functional human alpha 2A-ARs only in white fat cells. Interestingly, as in humans, subcutaneous adipocytes expressed higher levels of alpha2-AR than perigonadal fat cells, which are associated with a better antilipolytic response to epinephrine. High-fat-diet-induced obesity was observed in transgenic mice in the absence of fat cell size modifications. In addition, analysis of gene expression related to lipid metabolism in isolated adipocytes suggested reduced lipid mobilization and no changes in lipid storage capacity of transgenic mice fed a high-fat diet. Finally, the development of adipose tissue in these mice was not associated with significant modifications of glucose and insulin blood levels. Thus, these transgenic mice constitute an original model of diet-induced obesity for in vivo physiological and pharmacological studies with respect to the alpha2/beta-AR balance in adipose tissue.


Subject(s)
Adipose Tissue/metabolism , Receptors, Adrenergic, alpha-2/genetics , Adipocytes/cytology , Animals , Blood Glucose/analysis , Blood Pressure , Body Weight , Cell Size , Dietary Fats/pharmacology , Fatty Acids, Nonesterified/blood , Female , Gene Expression Regulation , Glucose Tolerance Test , Humans , Insulin/blood , Lipolysis/drug effects , Male , Mice , Mice, Transgenic , Middle Aged , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Adrenergic, beta/physiology , Tissue Distribution
8.
Biochem Biophys Res Commun ; 289(2): 564-7, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11716511

ABSTRACT

The regulation of resistin, a new adipose-derived circulating factor, is the subject of controversy. In particular, the question of its modulation in obesity led to opposite results reported by two different groups. In the current study, we assayed adipocyte resistin mRNA using fluorescent real-time RT-PCR. We studied the expression of resistin in mice which are differently sensitive to diet-induced obesity: the FVB/n strain, which poorly responds to high-fat diet and transgenic mice that express human alpha 2A-AR in adipose tissue in the absence of beta 3-adrenergic receptor (AR) under the FVB genetic background which are highly sensitive to high-fat diet and develop hyperplastic obesity. We observed that FVB mice, which have no significant increased body weight after an 8-week high-fat diet period, exhibited no alteration of resistin expression. In contrast, the transgenic mice developing high-fat diet-induced obesity exhibited markedly downregulated adipocyte resistin mRNA. We also showed that obesity induced by gold thioglucose injection in FVB/n mice reduces the expression of resistin in isolated adipocytes. This argues for decreased expression of resistin as a hallmark of obesity. Moreover, our data show that feeding a high-fat diet is not a primary determinant of resistin regulation.


Subject(s)
Diet , Hormones, Ectopic/metabolism , Intercellular Signaling Peptides and Proteins , Proteins , Adipose Tissue/metabolism , Animals , Body Weight , Dietary Fats , Fatty Acid Synthases/biosynthesis , Female , Hormones, Ectopic/biosynthesis , Lipoprotein Lipase/biosynthesis , Mice , Mice, Mutant Strains , Nerve Growth Factor , Obesity/genetics , RNA, Messenger/metabolism , Resistin , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
9.
Biochem J ; 357(Pt 3): 899-904, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11463364

ABSTRACT

We have previously shown that angiotensin II (Ang II) increases the expression of the gene encoding adipocyte fatty acid synthase (FAS). Here we investigate the mechanism responsible for increased FAS gene transcription by Ang II. We demonstrate that Ang II increased luciferase activity by 3-fold in 3T3-L1 adipocytes transfected with fusion constructs linking the FAS promoter to the luciferase reporter gene. Interestingly, we mapped the Ang II regulatory sequences to the insulin-responsive region (E box) in the proximal FAS promoter. The E box alone was able to mediate Ang II responsiveness when linked to a heterologous promoter. However, this response was lost when mutations that abolished the binding of the E box to its transcription factors were introduced. Using adenoviral overexpression of a dominant-negative form of adipocyte determination and differentiation factor 1 (ADD1), a transcription factor that binds to the insulin-responsive E box, we demonstrated that ADD1 was required for Ang II regulation of the FAS gene in 3T3-L1 adipocytes. Furthermore, ADD1 expression was also up-regulated by Ang II. With the use of transfections as well as glucose transport assays, we further demonstrated that Ang II stimulation of the FAS gene was dependent on glucose. In conclusion, this is the first report that Ang II regulates adipocyte FAS gene transcription via insulin response sequences in a glucose-dependent manner and that this regulation is mediated at least in part via the ADD1 transcription factor.


Subject(s)
Adipocytes/drug effects , Angiotensin II/pharmacology , CCAAT-Enhancer-Binding Proteins/physiology , DNA-Binding Proteins/physiology , Fatty Acid Synthases/genetics , Insulin/pharmacology , Transcription Factors , 3T3 Cells , Adipocytes/enzymology , Animals , Drug Interactions , Gene Expression Regulation, Enzymologic/drug effects , Glucose/pharmacology , Mice , Promoter Regions, Genetic/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Sterol Regulatory Element Binding Protein 1 , Transcription, Genetic/drug effects
11.
J Biol Chem ; 276(15): 11512-6, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11278421

ABSTRACT

Fatty acid synthase (FAS), a nutritionally regulated lipogenic enzyme, is transcriptionally controlled by ADD1/SREBP1c (adipocyte determination and differentiation 1/sterol regulatory element-binding protein 1c), through insulin-mediated stimulation of ADD1/SREBP1c expression. Progesterone exerts lipogenic effects on adipocytes, and FAS is highly induced in breast tumor cell lines upon progesterone treatment. We show here that progesterone up-regulates ADD1/SREBP1c expression in the MCF7 breast cancer cell line and the primary cultured preadipocyte from rat parametrial adipose tissue. In MCF7, progesterone induced ADD1/SREBP1c and Metallothionein II (a well known progesterone-regulated gene) mRNAs, with comparable potency. In preadipocytes, progesterone increased ADD1/SREBP1c mRNA dose-dependently, but not SREBP1a or SREBP2. Run-on experiments demonstrated that progesterone action on ADD1/SREBP1c was primarily at the transcriptional level. The membrane-bound and mature nuclear forms of ADD1/SREBP1 protein accumulated in preadipocytes cultured with progesterone, and FAS induction could be abolished by adenovirus-mediated overexpression of a dominant negative form of ADD1/SREBP1 in these cells. Finally, in the presence of insulin, progesterone was unable to up-regulate ADD1/SREBP1c mRNA in preadipocytes, whereas its effect was restored after 24 h of insulin deprivation. Together these results demonstrate that ADD1/SREBP1c is controlled by progesterone, which, like insulin, acts by increasing ADD1/SREBP1c gene transcription. This provides a potential mechanism for the lipogenic actions of progesterone on adipose tissue.


Subject(s)
Adipocytes/drug effects , CCAAT-Enhancer-Binding Proteins/genetics , Cell Differentiation/drug effects , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Progesterone/pharmacology , Transcription Factors , Adipocytes/cytology , Lipolysis , RNA, Messenger/genetics , Sterol Regulatory Element Binding Protein 1
12.
J Biol Chem ; 276(20): 16904-10, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11278795

ABSTRACT

Enlarged fat cells exhibit modified metabolic capacities, which could be involved in the metabolic complications of obesity at the whole body level. We show here that sterol regulatory element-binding protein 2 (SREBP-2) and its target genes are induced in the adipose tissue of several models of rodent obesity, suggesting cholesterol imbalance in enlarged adipocytes. Within a particular fat pad, larger adipocytes have reduced membrane cholesterol concentrations compared with smaller fat cells, demonstrating that altered cholesterol distribution is characteristic of adipocyte hypertrophy per se. We show that treatment with methyl-beta-cyclodextrin, which mimics the membrane cholesterol reduction of hypertrophied adipocytes, induces insulin resistance. We also produced cholesterol depletion by mevastatin treatment, which activates SREBP-2 and its target genes. The analysis of 40 adipocyte genes showed that the response to cholesterol depletion implicated genes involved in cholesterol traffic (caveolin 2, scavenger receptor BI, and ATP binding cassette 1 genes) but also adipocyte-derived secretion products (tumor necrosis factor alpha, angiotensinogen, and interleukin-6) and proteins involved in energy metabolism (fatty acid synthase, GLUT 4, and UCP3). These data demonstrate that altering cholesterol balance profoundly modifies adipocyte metabolism in a way resembling that seen in hypertrophied fat cells from obese rodents or humans. This is the first evidence that intracellular cholesterol might serve as a link between fat cell size and adipocyte metabolic activity.


Subject(s)
Adipocytes/physiology , Adipose Tissue/physiology , Cholesterol/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Glucose/metabolism , Receptors, Cell Surface , Transcription Factors/genetics , beta-Cyclodextrins , 3T3 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipose Tissue/cytology , Animals , Carboxypeptidase H , Carboxypeptidases/deficiency , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Carrier Proteins/physiology , Cell Membrane/physiology , Cells, Cultured , Cyclodextrins/pharmacology , Energy Metabolism , Epididymis , Gene Expression Regulation/drug effects , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hypertrophy , Insulin/pharmacology , Male , Membrane Lipids/physiology , Mice , Mice, Knockout , Mice, Obese , Rats , Rats, Zucker , Receptors, LDL/genetics , Receptors, Leptin , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Sterol Regulatory Element Binding Protein 2
13.
J Lipid Res ; 42(1): 106-13, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11160371

ABSTRACT

Fatty acid synthase (FAS), a key lipogenic enzyme, is expressed in the two major sites of fatty acid production in the body, that is, the liver and the adipose tissue. Surprisingly, the relative contribution of these sites to lipogenesis is highly variable among species. For example, besides the situation in rodents, where liver and fat are equally active, lipogenesis in some mammals such as the pig occurs principally in adipose tissue, whereas in avian species, the liver is the main lipogenic site. We addressed the question concerning the factors determining the site of fatty acid synthesis. We show that the expression of adipocyte determination and differentiation-dependent factor 1/sterol regulatory element-binding protein (ADD-1/SREBP-1) mRNA, but not SREBP-2, is linked to FAS protein content or activity in adipose tissues and livers of pig, chicken, and rabbit. Tissue differences in ADD-1/SREBP-1 mRNA expression between species were paralleled by commensurate variations in the nuclear concentration of SREBP-1 protein. Moreover, overexpression of ADD-1/SREBP-1 by adenoviral gene transfer induces FAS in chicken adipocytes, where lipogenesis is normally low. Conversely, the expression of a dominant negative form of ADD-1/SREBP-1 in pig adipocytes downregulates FAS expression. These results reinforce the role of ADD-1/SREBP-1 as a key regulator of lipogenesis, by extending its importance to nonrodent mammals and birds. Furthermore, they establish that differential expression of ADD-1/SREBP-1 is a key determinant of the site of fatty acid synthesis in the body.-Gondret, F., P. Ferré, and I. Dugail. ADD-1/SREBP-1 is a major determinant of tissue differential lipogenic capacity in mammalian and avian species. J. Lipid Res. 2001. 42: 106;-113.


Subject(s)
Birds/metabolism , CCAAT-Enhancer-Binding Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Lipids/biosynthesis , Mammals/metabolism , Adipose Tissue/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins/genetics , Chickens , DNA-Binding Proteins/genetics , Fatty Acid Synthases/drug effects , Fatty Acid Synthases/metabolism , Liver/cytology , Liver/metabolism , Male , Molecular Sequence Data , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rabbits , Sequence Alignment , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Swine , Tissue Distribution , Transcription Factors/genetics , Transduction, Genetic
14.
Nutr Metab Cardiovasc Dis ; 11(4 Suppl): 118-21, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11894744

ABSTRACT

Dietary digestible carbohydrates are able to modulate lipogenesis, by modifying the expression of genes coding for key lipogenic enzymes, like fatty acid synthase. The overall objective of the Nutrigene project (FAIR-CT97-3011) was to study the efficiency of various carbohydrates to modulate the lipogenic capacity and relevant gene expression in rat and human species (control and obese subjects) and to understand the underlying molecular mechanisms involved in the regulation of lipogenic genes by carbohydrates. Key cellular mediators (namely SREBP-1c and 2, AMP activated protein kinase, cholesterol content) of the regulation of lipogenic gene expression by glucose and/or insulin were identified and constitute new putative targets in the development of plurimetabolic syndrome associated with obesity. In humans, hepatic lipogenesis and triglyceride synthesis, assessed in vivo by the use of stable isotopes, was promoted by a high-carbohydrate diet in non obese subjects, and in non alcoholic steatotic patients, but was not modified in the adipose tissue of obese subjects. Non digestible/fermentable carbohydrates, such as fructans, were shown to decrease hepatic lipogenesis in non obese rats, and to lessen hepatic steatosis and body weight in obese Zucker rats. If confirmed in obese humans, this would allow the development of functional food able to counteract the metabolic disturbances linked to obesity.


Subject(s)
Adipose Tissue/metabolism , Dietary Carbohydrates/metabolism , Fatty Acid Synthases/metabolism , Gene Expression Regulation/physiology , Lipids/biosynthesis , Obesity/genetics , Adipose Tissue/physiopathology , Animals , Fatty Acid Synthases/genetics , Gene Expression Regulation/genetics , Humans , Liver/metabolism , Rats
15.
Biochem J ; 344 Pt 3: 873-80, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10585876

ABSTRACT

We show that Id (inhibitor of DNA binding) 2 and Id3, dominant negative members of the helix-loop-helix (HLH) family, interact with the adipocyte determination and differentiation factor 1 (ADD1)/sterol regulatory element-binding protein (SREBP) 1c, a transcription factor of the basic HLH-leucine zipper family that controls the expression of several key genes of adipose metabolism. Gel mobility-shift assays performed with in vitro-translated ADD1, Id2 or Id3 proteins and a fatty acid synthase (FAS) promoter oligonucleotide showed evidence for a marked inhibition of the formation of DNA-ADD1 complexes by Id2 or Id3 proteins. Co-immunoprecipitation studies using in vitro-translated proteins demonstrated further the physical interaction of Id and ADD1/SREBP-1c proteins in the absence of DNA. Using the FAS gene as a model of an ADD1-regulated promoter in transiently transfected isolated rat adipocytes or mature 3T3-L1 adipocytes, a potent inhibition of the activity of the FAS-chloramphenicol acetyltransferase reporter gene was observed by overexpression of Id2 or Id3. Reciprocally, co-transfection of Id3 antisense and ADD1 expression vectors in preadipocytes potentiated the ADD1/SREBP-1c effect on the FAS promoter activity. Finally, in the non adipogenic NIH-3T3 cell line, most of the ADD1-mediated trans-activation of the FAS promoter was counteracted by co-transfection of Id2 or Id3 expression vectors. Previous studies have indicated Id gene expression to be down-regulated during adipogenesis [Moldes, Lasnier, Fève, Pairault and Djian (1997) Mol. Cell. Biol. 17, 1796-1804]. We here demonstrated that there was a dramatic rise of Id2 and Id3 mRNA levels when 3T3-L1 adipocytes or isolated rat fat cells were exposed to lipolytic and anti-lipogenic agents, forskolin and isoproterenol. Taken together, our data show that Id products are functionally involved in modulating ADD1/SREBP-1c transcriptional activity, and thus lipogenesis in adipocytes.


Subject(s)
CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/antagonists & inhibitors , Fatty Acid Synthases/genetics , Neoplasm Proteins , Nuclear Proteins/antagonists & inhibitors , Promoter Regions, Genetic , Repressor Proteins , Transcription Factors/antagonists & inhibitors , 3T3 Cells , Adipocytes , Animals , Cell Differentiation/genetics , Cyclic AMP/pharmacology , DNA-Binding Proteins/genetics , Gene Expression Regulation, Enzymologic , Genes, Reporter , Inhibitor of Differentiation Protein 1 , Inhibitor of Differentiation Protein 2 , Inhibitor of Differentiation Proteins , Mice , RNA, Messenger/metabolism , Rats , Signal Transduction/genetics , Sterol Regulatory Element Binding Protein 1 , Transcription Factors/genetics , fas Receptor/genetics
16.
Mol Cell Biol ; 19(5): 3760-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10207099

ABSTRACT

The transcription of genes encoding proteins involved in the hepatic synthesis of lipids from glucose is strongly stimulated by carbohydrate feeding. It is now well established that in the liver, glucose is the main activator of the expression of this group of genes, with insulin having only a permissive role. While ADD1/SREBP-1 has been implicated in lipogenic gene expression through temporal association with food intake and ectopic gain-of-function experiments, no genetic evidence for a requirement for this factor in glucose-mediated gene expression has been established. We show here that the transcription of ADD1/SREBP-1c in primary cultures of hepatocytes is controlled positively by insulin and negatively by glucagon and cyclic AMP, establishing a link between this transcription factor and carbohydrate availability. Using adenovirus-mediated transfection of a powerful dominant negative form of ADD1/SREBP-1c in rat hepatocytes, we demonstrate that this factor is absolutely necessary for the stimulation by glucose of L-pyruvate kinase, fatty acid synthase, S14, and acetyl coenzyme A carboxylase gene expression. These results demonstrate that ADD1/SREBP-1c plays a crucial role in mediating the expression of lipogenic genes induced by glucose and insulin.


Subject(s)
CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Glucose/pharmacology , Lipids/biosynthesis , Liver/metabolism , Nuclear Proteins/metabolism , Transcription Factors , Transcriptional Activation/genetics , Adenoviridae/genetics , Animals , Cells, Cultured , Cyclic AMP/pharmacology , Female , Glucagon/pharmacology , Glycolysis/genetics , Histocytochemistry , Insulin/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sterol Regulatory Element Binding Protein 1 , Transfection
17.
J Biol Chem ; 273(44): 29164-71, 1998 Oct 30.
Article in English | MEDLINE | ID: mdl-9786926

ABSTRACT

Elevated lipogenesis is a key determinant of exaggerated fat deposition in adipose tissue of obese Zucker rats. We previously delineated a region in the fatty-acid synthase promoter, which was responsible for obesity-related overexpression of the fatty-acid synthase (FAS) gene, by negatively regulating the activity of the downstream promoter in lean but not obese rat fat cells. The present study aimed to identify the transcriptional factors acting on this target region. First, functional analysis of mutated FAS promoter constructs in transiently transfected lean and obese rat adipocytes showed that the activity of the obesity-related region relied on the presence of a transcriptionally inactive sterol regulatory element at -150, which counteracted activation through the downstream E-box. Adenovirus-mediated overexpression of a dominant negative form of adipocyte determination and differentiation factor 1 (ADD1) was used to neutralize endogenous ADD1/ sterol regulatory element-binding protein (SREBP) transcriptional activity in fat cells, by producing inactive dimers unable to bind target DNA. With this system, we observed that overexpression of FAS in obese rat adipocytes was ADD1/SREBP-dependent. SREBP isoforms expression was assessed in lean and obese rat fat cells and showed no differences in the level of ADD1/SREBP1 mRNA. In addition, equivalent amounts of immunoreactive ADD1/SREBP1 were found in nuclear extracts from lean and obese rat fat cells. In contrast, immunoreactive SREBP2, which was very low in nuclear extracts from lean rats, was induced in obese rat fat cells. Finally, using in vitro binding studies, we showed that SREBP2 was able to displace ADD1/SREBP1 binding from the sterol regulatory element (SRE) site. Thus, we propose a mechanism for obesity-related overexpression of FAS gene in rat adipocyte. ADD1/SREBP1-activated transcription proceeding from the E-box motif is counterbalanced by a negative SRE site acting by limiting the availability of ADD1/SREBP1 in normal fat cells. The negative effect of this site is abolished in obese rat adipocyte nuclei where SREBP2 is induced and can substitute for ADD1/SREBP1 binding to the inactive SRE. These results provide evidence for the implication of SREBPs in the dysregulation of adipocyte metabolism characteristic of the obese state.


Subject(s)
Adipose Tissue/enzymology , CCAAT-Enhancer-Binding Proteins , Fatty Acid Synthases/genetics , Obesity/genetics , Transcription, Genetic , Animals , Binding, Competitive , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Male , Nuclear Proteins/metabolism , Obesity/enzymology , Protein Binding , Rats , Rats, Zucker , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Obes Res ; 6(2): 122-7, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9545018

ABSTRACT

The massive obesity caused in rodents by the disruption of the leptin-receptor signal through genetic defects at the level of either leptin (OB) or leptin receptor (OB-R) has raised the question of the relevance of these genes to morbid obesity in humans. In this study, we screened a large population of massively obese subjects for the presence of a leptin receptor mutation homologous to that of fa/fa rats, a single base substitution changing glutamine 269, a highly conserved glutamine found at position 270 in the human sequence. After polymerase chain reaction (PCR) amplification of a DNA region encompassing the end of exon 5, intron 5, and the beginning of exon 6, we performed restriction fragment length polymorphism analysis. Within the limitations of this approach where only mutations introducing restriction sites (5 of 8 possibilities) could be assessed, no evidence of mutation at the codon gln 270 was found in 343 massively obese subjects. However, a new OB-R gene variant in intron 5 was revealed by MaeII digestion of the PCR products. MaeII/hOB-R genotyping revealed no difference in the distribution of the genotypes between obese subjects and a group of 79 unrelated nonobese control subjects. In addition, no significant association between various obesity-related metabolic phenotypes and the presence of MaeII/hOB-R alleles was found. Thus, our results did not support a significant role for the MaeII/hOB-R gene variant in the development of the obese phenotype in the population we studied.


Subject(s)
Carrier Proteins/genetics , Introns , Mutation , Obesity, Morbid/genetics , Receptors, Cell Surface , Adult , Animals , Deoxyribonucleases, Type II Site-Specific , Diabetes Mellitus/genetics , Exons , Genotype , Glutamine/genetics , Humans , Middle Aged , Obesity , Polymerase Chain Reaction , Rats , Receptors, Leptin , Sequence Homology
19.
Diabetes ; 46(9): 1393-9, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9287037

ABSTRACT

Thiazolidinediones are potent antidiabetic compounds, in both animal and human models, which act by enhancing peripheral sensitivity to insulin. Thiazolidinediones are high-affinity ligands for peroxisome proliferator-activated receptor-gamma, a key factor for adipocyte differentiation, and they are efficient promoters of adipocyte differentiation in vitro. Thus, it could be questioned whether a thiazolidinedione therapy aimed at improving insulin sensitivity would promote the recruitment of new adipocytes in vivo. To address this problem, we have studied the in vivo effect of pioglitazone on glucose metabolism and gene expression in the adipose tissue of an animal model of obesity with insulin resistance, the obese Zucker (fa/fa) rat. Pioglitazone markedly improves insulin action in the obese Zucker (fa/fa) rat, but doubles its weight gain after 4 weeks of treatment. The drug induces a large increase of glucose utilization in adipose tissue, where it stimulates the expression of genes involved in lipid metabolism such as the insulin-responsive GLUT, fatty acid synthase, and phosphoenolpyruvate carboxykinase genes, but decreases the expression of the ob gene. These changes are related to both an enhanced adipocyte differentiation, as shown by the large increase in the number of small adipocytes in the retroperitoneal fat pad, and a direct effect of pioglitazone on specific gene expression (phosphoenolpyruvate carboxykinase and ob genes) in mature adipocytes.


Subject(s)
Adipocytes/metabolism , Glucose/metabolism , Muscle Proteins , Obesity/metabolism , Thiazoles/therapeutic use , Thiazolidinediones , Animals , Body Weight/drug effects , Cell Differentiation , Fatty Acid Synthases/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glucose Transporter Type 4 , Insulin Resistance , Leptin , Lipid Metabolism , Lipid Mobilization/drug effects , Monosaccharide Transport Proteins/genetics , Obesity/pathology , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Pioglitazone , Proteins/genetics , Rats , Rats, Mutant Strains
20.
J Biol Chem ; 271(35): 21297-302, 1996 Aug 30.
Article in English | MEDLINE | ID: mdl-8702907

ABSTRACT

We have previously shown that the proximal 2-kb sequence of the fatty acid synthase (FAS) promoter transfected into rat adipocytes was highly sensitive to the cellular context, displaying an overactivity in obese (fa/fa) versus lean Zucker rat adipocytes. Using deletional analysis, we show here that FAS promoter activity mainly depends on a region from -200 to -126. This sequence exerts a strong negative effect on FAS promoter in adipocytes from lean rats but not in those from obese rats, resulting in a marked overtranscriptional activity in the latter cells. This region, fused to a heterologous promoter, the E1b TATA box, induced differential levels of gene reporter activity in lean and obese rat adipocytes, indicating it harbors fa-responsive element(s). Whatever the rat genotype, adipocyte nuclear proteins were shown to footprint the same protected sequence within the fa-responsive region, and supershift analysis demonstrated that Sp1 or Sp1-like proteins were bound to this DNA subregion. Compelling evidence that the Sp1 binding site contained in this sequence was implicated in the differential promoter activity in lean versus obese rats, was provided by the observation that mutations at this Sp1 site induced a 2.5-fold increase in FAS promoter activity in adipocytes from lean rats, whereas they had no effect in adipocytes from obese rats.


Subject(s)
Adipocytes/enzymology , Fatty Acid Synthases/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites , DNA , Molecular Sequence Data , Mutagenesis , Nuclear Proteins/metabolism , Rats , Rats, Zucker , Regulatory Sequences, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...