Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
J Endocrinol ; 215(3): 363-73, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23033362

ABSTRACT

Cancer cachexia induces loss of fat mass that accounts for a large part of the dramatic weight loss observed both in humans and in animal models; however, the literature does not provide consistent information regarding the set point of weight loss and how the different visceral adipose tissue depots contribute to this symptom. To evaluate that, 8-week-old male Wistar rats were subcutaneously inoculated with 1 ml (2×10(7)) of tumour cells (Walker 256). Samples of different visceral white adipose tissue (WAT) depots were collected at days 0, 4, 7 and 14 and stored at -80 °C (seven to ten animals/each day per group). Mesenteric and retroperitoneal depot mass was decreased to the greatest extent on day 14 compared with day 0. Gene and protein expression of PPARγ2 (PPARG) fell significantly following tumour implantation in all three adipose tissue depots while C/EBPα (CEBPA) and SREBP-1c (SREBF1) expression decreased over time only in epididymal and retroperitoneal depots. Decreased adipogenic gene expression and morphological disruption of visceral WAT are further supported by the dramatic reduction in mRNA and protein levels of perilipin. Classical markers of inflammation and macrophage infiltration (f4/80, CD68 and MIF-1α) in WAT were significantly increased in the later stage of cachexia (although showing a incremental pattern along the course of cachexia) and presented a depot-specific regulation. These results indicate that impairment in the lipid-storing function of adipose tissue occurs at different times and that the mesenteric adipose tissue is more resistant to the 'fat-reducing effect' than the other visceral depots during cancer cachexia progression.


Subject(s)
Adipose Tissue/metabolism , Cachexia/metabolism , Neoplasms/complications , Adipokines/blood , Adipose Tissue/pathology , Animals , Blotting, Western , Cachexia/blood , Cachexia/pathology , Male , Neoplasms/blood , Neoplasms/physiopathology , PPAR gamma/metabolism , Polymerase Chain Reaction , Rats , Rats, Wistar
2.
Cytokine ; 57(1): 9-16, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22099872

ABSTRACT

Cancer cachexia is a multifaceted syndrome whose aetiology is extremely complex and is directly related to poor patient prognosis and survival. Changes in lipid metabolism in cancer cachexia result in marked reduction of total fat mass, increased lipolysis, total oxidation of fatty acids, hyperlipidaemia, hypertriglyceridaemia, and hypercholesterolaemia. These changes are believed to be induced by inflammatory mediators, such as tumour necrosis factor-α (TNF-α) and other factors. Attention has recently been drawn to the current theory that cachexia is a chronic inflammatory state, mainly caused by the host's reaction to the tumour. Changes in expression of numerous inflammatory mediators, notably in white adipose tissue (WAT), may trigger several changes in WAT homeostasis. The inhibition of adipocyte differentiation by PPARγ is paralleled by the appearance of smaller adipocytes, which may partially account for the inhibitory effect of PPARγ on inflammatory gene expression. Furthermore, inflammatory modulation and/or inhibition seems to be dependent on the IKK/NF-κB pathway, suggesting that a possible interaction between NF-κB and PPARγ is required to modulate WAT inflammation induced by cancer cachexia. In this article, current literature on the possible mechanisms of NF-κB and PPARγ regulation of WAT cells during cancer cachexia are discussed. This review aims to assess the role of a possible interaction between NF-κB and PPARγ in the setting of cancer cachexia as well as its significant role as a potential modulator of chronic inflammation that could be explored therapeutically.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/pathology , Cachexia/complications , Cell Nucleus/metabolism , Inflammation/complications , Neoplasms/complications , Transcription Factors/metabolism , Animals , Cachexia/pathology , Humans , Inflammation/pathology , Neoplasms/pathology
3.
Int J Obes (Lond) ; 29 Suppl 1: S13-6, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15711576

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARgamma) is a nuclear receptor regulating an array of diverse functions in a variety of cell types including regulation of genes associated with growth and differentiation. Its most notable function is to regulate development of adipose tissue, which involves coordinating expression of many hundreds of genes responsible for establishment of the mature adipocyte phenotype. Our recent studies have demonstrated a role for MEK/ERK signaling and CCAAT/enhancer binding proteins (C/EBP)beta in regulating expression of PPARgamma during adipogenesis. Furthermore, we have shown that cAMP-dependent signaling along with C/EBPbeta leads to the stimulation of PPARgamma activity by mechanisms that probably involve production of PPARgamma ligands. Additionally, we have recently demonstrated that phosphorylation of C/EBPbeta at a consensus ERK/GSK3 site is required for the PPARgamma-associated expression of adiponectin during the terminal stages of adipogenesis. GSK3beta also influences PPARgamma activity by regulating the turnover and subcellular localization of beta-catenin, a potent transcriptional activator of Wnt signaling. In fact, we have recently shown a crosstalk between PPARgamma and beta-catenin signaling. Specifically, activation of PPARgamma induces the degradation of beta-catenin during preadipocyte differentiation by mechanisms that require GSK3beta and the proteasome. In contrast, expression of a GSK3beta-phosphorylation-defective beta-catenin renders beta-catenin resistant to the degradatory action of PPARgamma. Interestingly, expression of the mutant beta-catenin blocks expression of adiponectin and C/EBPalpha in response to the activation of PPARgamma.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , MAP Kinase Signaling System/physiology , Obesity/metabolism , PPAR gamma/genetics , Adipose Tissue/cytology , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Cell Proliferation , Cytoskeletal Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , PPAR gamma/metabolism , Trans-Activators/metabolism , beta Catenin
4.
Obes Rev ; 5(4): 189-96, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15458394

ABSTRACT

This review highlights the presentations and discussions held during the 2003 Stock Conference in Lisbon focussed on the identification of new therapeutic targets for the treatment of obesity and identified through molecular and genetic studies. Transcription factors and their cofactors, signalling pathways and new insights provided by cellular and genetic studies were discussed as potential new avenues to modulate adipocyte formation and function.


Subject(s)
Adipocytes/physiology , Adipose Tissue/metabolism , Obesity/genetics , Obesity/therapy , Adipocytes/metabolism , Energy Metabolism/genetics , Energy Metabolism/physiology , Homeostasis , Humans , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology
5.
J Biol Chem ; 276(21): 18464-71, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11279134

ABSTRACT

The differentiation of 3T3-L1 preadipocytes is regulated in part by a cascade of transcriptional events involving activation of the CCAAT/enhancer-binding proteins (C/EBPs) and peroxisome proliferator-activated receptor gamma (PPARgamma) by dexamethasone (DEX), 3-isobutyl-1-methylxanthine (MIX), and insulin. In this study, we demonstrate that exposure of 3T3-L1 preadipocytes to DEX and insulin fails to induce adipogenesis as indicated by a lack of C/EBPalpha, PPARgamma2, and adipose protein 2/fatty acid-binding protein expression; however, PPARgamma1 is expressed. Treatment of these MIX-deficient cells with a PPARgamma ligand, troglitazone, induces C/EBPalpha expression and rescues the block in adipogenesis. In this regard, we also show that induction of C/EBPalpha gene expression by troglitazone in C3H10T1/2 cells ectopically expressing PPARgamma occurs in the absence of ongoing protein synthesis, suggesting a direct transactivation of the C/EBPalpha gene by PPARgamma. Furthermore, ectopic expression of a dominant negative isoform of C/EBPbeta (liver-enriched transcriptional inhibitory protein (LIP)) inhibits the induction of C/EBPalpha, PPARgamma2, and adipose protein 2/fatty acid-binding protein by DEX, MIX, and insulin in 3T3-L1 cells without affecting the induction of PPARgamma1 by DEX. Exposure of LIP-expressing preadipocytes to troglitazone along with DEX, MIX, and insulin induces differentiation into adipocytes. Additionally, we show that sustained expression of C/EBPalpha in these LIP-expressing adipocytes requires constant exposure to troglitazone. Taken together, these observations suggest that inhibition of C/EBPbeta activity not only blocks C/EBPalpha and PPARgamma2 expression, but it also renders the preadipocytes dependent on an exogenous PPARgamma ligand for their differentiation into adipocytes. We propose, therefore, an additional role for C/EBPbeta in regulating PPARgamma activity during adipogenesis, and we suggest an alternative means of inducing preadipocyte differentiation that relies on the dexamethasone-associated induction of PPARgamma1 expression.


Subject(s)
Adipocytes/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adipocytes/cytology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Differentiation , Mice , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Transcription Factors/genetics , Transcriptional Activation
6.
J Biol Chem ; 276(1): 722-8, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11024036

ABSTRACT

The beta(3)-adrenergic receptor (beta(3)AR) is expressed predominantly in adipocytes, and it plays a major role in regulating lipolysis and adaptive thermogenesis. Its expression in a variety of adipocyte cell models is preceded by the appearance of CCAAT/enhancer-binding protein alpha (C/EBP alpha), which has been shown to regulate a number of other adipocyte-specific genes. Importantly, it has been demonstrated that several adipocyte cell lines that fail to express C/EBP alpha exhibit reduced insulin sensitivity, despite an apparent adipogenic phenotype. Here we show that transcription and function of the beta(3)AR correlates with C/EBP alpha expression in these adipocyte models. A 5.13-kilobase pair fragment of the mouse beta(3)AR promoter was isolated and sequenced. This fragment conferred a 50-fold increase in luciferase reporter gene expression in adipocytes. Two putative C/EBP binding sites exist at -3306 to -3298 and at -1462 to -1454, but only the more distal site is functional. Oligonucleotides corresponding to both the wild-type and mutated -3306 element were inserted upstream of a thymidine kinase luciferase construct. When cotransfected in fibroblasts with a C/EBP alpha expression vector, reporter gene expression increased 3-fold only in the wild-type constructs. The same mutation, when placed into the intact 5.13-kilobase pair promoter, reduced promoter activity in adipocytes from 50-fold to <10-fold. Electrophoretic mobility shift analysis demonstrated that the site at -3306 generated a specific protein-oligonucleotide complex that was supershifted by C/EBP alpha antibody, while a probe corresponding to a putative site at -1462 did not. These results define C/EBP alpha as a key transcriptional regulator of the mouse beta(3)AR gene during adipogenesis.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Receptors, Adrenergic, beta-3/genetics , 3T3 Cells , Adenylyl Cyclases/metabolism , Adipose Tissue/drug effects , Adrenergic beta-3 Receptor Agonists , Animals , COS Cells , DNA/genetics , DNA/metabolism , Dioxoles/pharmacology , Enzyme Activation/drug effects , Gene Expression Regulation, Developmental , Genes, Reporter , Mice , Molecular Sequence Data , Mutation , Organ Specificity , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics , Sequence Analysis, DNA , Transfection
7.
Am J Physiol Endocrinol Metab ; 279(4): E893-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11001773

ABSTRACT

The major leptin-containing membrane compartment was identified and characterized in rat adipose cells by means of equilibrium density and velocity sucrose gradient centrifugation. This compartment appears to be different from peptide-containing secretory granules present in neuronal, endocrine, and exocrine cells, as well as from insulin-sensitive GLUT-4-containing vesicles abundant in adipocytes. Exocytosis of both leptin- and GLUT-4-containing vesicles can be induced by insulin; however, only leptin secretion is responsive to serum stimulation. This latter effect is resistant to cycloheximide, suggesting that serum triggers the release of a stored pool of presynthesized leptin molecules. We conclude that regulated secretion of leptin and insulin-dependent translocation of GLUT-4 represent different pathways of membrane trafficking in rat adipose cells. NIH 3T3 cells ectopically expressing CAAT box enhancer binding protein-alpha and Swiss 3T3 cells expressing peroxisome proliferator-activated receptor-gamma undergo differentiation in vitro and acquire adipocyte morphology and insulin-responsive glucose uptake. Only the former cell line, however, is capable of leptin secretion. Thus different transcriptional mechanisms control the developmental onset of these two major and independent physiological functions in adipose cells.


Subject(s)
Adipocytes/metabolism , Cell Compartmentation/physiology , Intracellular Fluid/metabolism , Leptin/metabolism , Muscle Proteins , 3T3 Cells , Adipocytes/cytology , Adipocytes/drug effects , Animals , Blood Proteins/pharmacology , CCAAT-Enhancer-Binding Protein-alpha/biosynthesis , Carrier Proteins/metabolism , Cell Differentiation/drug effects , Cell Fractionation , Cells, Cultured , Centrifugation, Density Gradient , Glucose/metabolism , Glucose Transporter Type 4 , Male , Mice , Monosaccharide Transport Proteins/metabolism , Organelles/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/biosynthesis , Transcription Factors/biosynthesis
8.
J Nutr ; 130(12): 3116S-3121S, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11110883

ABSTRACT

Recent advances regarding the biology of adipose tissue have identified the adipocyte as an important mediator in many physiologic and pathologic processes regarding energy metabolism. Consideration for a central role of adipose tissue in the development of obesity, cardiovascular disease and noninsulin-dependent diabetes mellitus has resulted in new incentives toward understanding the complexities of adipocyte differentiation. Current knowledge of this process includes a cascade of transcriptional events that culminate in the expression of peroxisome proliferator-activated receptor-gamma (PPARgamma) and CCAAT/enhancer binding protein-alpha (C/EBPalpha). These prominent adipogenic transcription factors have been shown to regulate, directly or indirectly, the gene expression necessary for the development of the mature adipocyte. Hormonal and nutritional signaling that impinges on these trans-acting factors provides a molecular link between lipids and lipid-related compounds and the gene expression important for glucose and lipid homeostasis. Knowledge concerning the transcriptional events mediating adipocyte differentiation provides a basis for understanding the physiologic processes associated with adipose tissue as well as for the development of therapeutic interventions in obesity and its related disorders.


Subject(s)
Adipocytes/physiology , Hormones/physiology , Obesity/physiopathology , Transcription, Genetic/physiology , Adipocytes/cytology , Adipose Tissue/physiopathology , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/physiology , Cell Differentiation/physiology , Energy Metabolism , Gene Expression , Hormones/blood , Humans , Lipid Metabolism , Obesity/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/physiology
9.
Biochem Biophys Res Commun ; 262(1): 216-22, 1999 Aug 19.
Article in English | MEDLINE | ID: mdl-10448095

ABSTRACT

We have recently demonstrated that STAT1, STAT5A, and STAT5B are induced during adipogenesis of cultured preadipocytes in a differentiation-dependent manner. Members of the C/EBP and PPAR families of transcription factors have also been shown to be induced during adipocyte differentiation and to play a significant role in the regulation of fat-specific genes. In this investigation, we have examined the ability of C/EBPs and PPARs to contribute to STAT protein expression during conversion of non-precursor fibroblasts to functionally mature adipocytes. For this study, NIH-3T3 fibroblasts engineered to ectopically co-express C/EBPbeta and C/EBPdelta under the control of a tetracycline-responsive, inducible expression system were utilized to assess STAT expression during controlled adipogenesis. Data presented here demonstrate that STAT1, STAT5A, and STAT5B, but not STAT3 and STAT6, were induced in a tetracycline-responsive manner during the differentiation of these engineered fibroblasts. The STAT protein accumulation resulting from C/EBP expression was tightly coupled to the morphological conversion of fibroblasts to adipocytes and represents an expression profile identical to that reported for mature adipocytes in vivo. Data are also presented demonstrating that STAT protein accumulation and adipocyte conversion occurred only during controlled conditions leading to the expression of PPARgamma and that the expression of these three STATs was tightly regulated in a PPARgamma ligand dose-response fashion. These data illustrate that the cascade of transcriptional events leading to adipogenesis regulate the STAT family of transcription factors and that the differentiation-dependent upregulation of STAT protein expression is regulated downstream of PPARgamma in a ligand-dependent manner.


Subject(s)
Adipocytes/cytology , Adipose Tissue/metabolism , DNA-Binding Proteins/biosynthesis , Milk Proteins , Receptors, Cytoplasmic and Nuclear/metabolism , Trans-Activators/biosynthesis , Transcription Factors/metabolism , 3T3 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/cytology , Animals , Blotting, Western , CCAAT-Enhancer-Binding Proteins , Cell Differentiation , Cell Size , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Gene Expression/drug effects , Ligands , Lipid Metabolism , Mice , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , STAT1 Transcription Factor , STAT5 Transcription Factor , Tetracycline/pharmacology , Time Factors , Transcription Factors/genetics , Up-Regulation/drug effects
10.
J Biol Chem ; 274(24): 17088-97, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10358062

ABSTRACT

Molecular mechanisms coupling growth arrest and cell differentiation were examined during adipogenesis. Data are presented that document a cascade expression of members of two independent families of cyclin-dependent kinase inhibitors that define distinct states of growth arrest during 3T3-L1 preadipocyte differentiation. Exit from the cell cycle into a pre-differentiation state of post-mitotic growth arrest was characterized by significant increases in p21 and p27. During onset of irreversible growth arrest associated with terminal differentiation, the level of p21 declined with a concomitant, dramatic increase in p18 and a sustained level of p27. The expression of p18 and p21, regulated at the level of protein and mRNA accumulation, was directly coupled to differentiation. Stable cell lines were engineered to express adipogenic transcription factors to examine the active role of trans-acting elements in regulating these cell cycle inhibitors. Ectopic expression of peroxisome proliferator-activated receptor (PPAR) gamma in non-precursor fibroblastic cell lines resulted in conversion to adipocytes and a coordinated increase in p18 and p21 mRNA and protein expression in a PPARgamma ligand-associated manner. These data demonstrate a role for PPARgamma in mediating the differentiation-dependent cascade expression of cyclin-dependent kinase inhibitors, thereby providing a molecular mechanism coupling growth arrest and adipocyte differentiation.


Subject(s)
Adipocytes/cytology , Carrier Proteins/metabolism , Cell Cycle Proteins , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Enzyme Inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins , 3T3 Cells , Animals , CCAAT-Enhancer-Binding Proteins , Cell Cycle , Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p18 , Cyclin-Dependent Kinase Inhibitor p21 , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation , Mice , Mice, Inbred BALB C , Models, Biological , Nuclear Proteins/biosynthesis , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic
11.
Exp Cell Res ; 249(1): 177-87, 1999 May 25.
Article in English | MEDLINE | ID: mdl-10328964

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) plays a role in several disease states such as sepsis, cachexia, and non-insulin-dependent diabetes. TNF-alpha interferes with insulin signaling and inhibits differentiation-specific gene expression in adipose tissue and skeletal muscle. We have examined the mechanisms by which TNF-alpha, in comparison to basic fibroblast growth factor (bFGF), inhibits the insulin-like growth factor-I (IGF-I)-induced differentiation of C2C12 myoblasts. Adhesion of quiescent, suspended myoblasts to collagen in high concentrations of IGF-I (10 nM) induced these cells to proliferate during the initial 24 h postplating and in so doing transiently inhibited the expression of myogenin, an essential transcription factor controlling myoblast differentiation. Low doses of IGF-I (1 nM) were minimally mitogenic and enhanced muscle-specific gene expression. Quiescent myoblasts treated with bFGF in combination with IGF-I did not express myogenin, but expressed proliferating cell nuclear antigen and underwent DNA synthesis. In contrast, TNF-alpha in the presence or absence of 1 nM IGF-I, did not stimulate DNA synthesis in myoblasts. However, TNF-alpha inhibited myogenin mRNA and protein expression. Expression of the cyclin-dependent kinase inhibitor p21 correlated with myogenin expression and myoblast differentiation, but not with growth arrest. These results indicate that both TNF-alpha and bFGF inhibit myogenin expression but differentially influence myoblast proliferation.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Insulin-Like Growth Factor I/antagonists & inhibitors , Muscle, Skeletal/drug effects , Myogenin/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Adhesion , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Collagen , Depression, Chemical , Insulin-Like Growth Factor I/pharmacology , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myogenin/genetics , Phenotype , Transcription, Genetic/drug effects
12.
J Biol Chem ; 274(12): 7946-51, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10075691

ABSTRACT

Adipocyte differentiation is regulated by at least two major transcription factors, CCAAT/enhancer-binding protein alpha (C/EBPalpha) and peroxisome proliferator-activated receptor gamma (PPARgamma). Expression of PPARgamma in fibroblasts converts them to fat-laden cells with an adipocyte-like morphology. Here, we investigate the ability of PPARgamma to confer insulin-sensitive glucose transport to a variety of murine fibroblast cell lines. When cultured in the presence of a PPARgamma ligand, Swiss-3T3 and BALB/c-3T3 cells ectopically expressing PPARgamma accumulate lipid droplets, express C/EBPalpha, aP2, insulin-responsive aminopeptidase, and glucose transporter isoform 4 (GLUT4), and exhibit highly insulin-responsive 2-deoxyglucose uptake. In contrast, PPARgamma-expressing NIH-3T3 cells, despite similar lipid accumulation, adipocyte morphology, and aP2 expression, do not express C/EBPalpha or GLUT4 and fail to acquire insulin sensitivity. In cells ectopically expressing PPARgamma, the development of insulin-responsive glucose uptake correlates with C/EBPalpha expression. Furthermore, ectopic expression of C/EBPalpha in NIH-3T3 cells converts them to the adipocyte phenotype and restores insulin-sensitive glucose uptake. We propose that the pathway(s) leading to fat accumulation and morphological changes are distinct from that leading to insulin-dependent glucose transport. Our results suggest that although PPARgamma is sufficient to trigger the adipogenic program, C/EBPalpha is required for establishment of insulin-sensitive glucose transport.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Glucose/metabolism , Insulin/pharmacology , Muscle Proteins , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Biological Transport/drug effects , CCAAT-Enhancer-Binding Proteins , Cell Differentiation/drug effects , Fibroblasts/metabolism , Glucose Transporter Type 4 , Mice , Mice, Inbred BALB C , Monosaccharide Transport Proteins/metabolism
13.
Ann N Y Acad Sci ; 892: 134-45, 1999 Nov 18.
Article in English | MEDLINE | ID: mdl-10842659

ABSTRACT

Adipocyte differentiation is regulated by at least two families of transcription factors, CCAAT/enhancer binding proteins (C/EBPs) and peroxisome proliferator-activated receptors (PPARs). Induction of PPAR gamma gene transcription during the differentiation of preadipocytes into adipocytes in vitro occurs following an initial phase of cell proliferation and requires a direct involvement of C/EBP beta, C/EBP delta, and glucocorticoids. Ectopic expression of PPAR gamma in non-adipogenic, Swiss 3T3 fibroblasts promotes their conversion into adipocytes as indicated by the accumulation of lipid droplets and the induction of C/EBP alpha, aP2, insulin-responsive aminopeptidase (IRAP), and glucose transporter 4 (GLUT4) expression. These PPAR gamma-expressing Swiss cells also exhibit a high level of insulin-responsive glucose uptake that is comparable to that expressed in 3T3-L1 adipocytes. In contrast, PPAR gamma-expressing NIH-3T3 fibroblasts, despite similar lipid accumulation, adipocyte morphology, and aP2 expression, do not synthesize C/EBP alpha and fail to acquire insulin sensitivity. In Swiss 3T3 cells ectopically expressing PPAR gamma, the development of insulin-responsive glucose uptake correlates with C/EBP alpha expression. Furthermore, ectopic expression of C/EBP alpha in NIH-3T3 cells induces PPAR gamma expression and adipogenesis, but also restores insulin-sensitive glucose transport. These results suggest that although PPAR gamma is sufficient to trigger the adipogenic program, C/EBP alpha is required for establishment of insulin-sensitive glucose transport in adipocytes.


Subject(s)
3T3 Cells/metabolism , Adipocytes/metabolism , DNA-Binding Proteins/metabolism , Glucose/metabolism , Muscle Proteins , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sialoglycoproteins/metabolism , Thiazolidinediones , Transcription Factors/metabolism , Transcription, Genetic , Animals , CCAAT-Enhancer-Binding Proteins , Chromans/pharmacology , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Hypoglycemic Agents/pharmacology , Interleukin 1 Receptor Antagonist Protein , Mice , Monosaccharide Transport Proteins/metabolism , Nuclear Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Thiazoles/pharmacology , Transcription Factors/genetics , Transcription, Genetic/drug effects , Troglitazone
14.
J Cell Biochem ; Suppl 32-33: 59-67, 1999.
Article in English | MEDLINE | ID: mdl-10629104

ABSTRACT

The adipocyte is now known to play an active role in many physiological and pathological processes regarding energy metabolism. Consideration of adipose tissue as an endocrine organ that secretes a variety of unrelated bioactive molecules has broadened our appreciation of adipocyte function to exceed the once considered passive role in lipid metabolism. Growing interest in this tissue has lead to significant advances regarding the molecular basis for adipocyte differentiation. Several diverse families of transcription factors are currently under active investigation for their roles in mediating this complex process. Knowledge concerning the sequence of transcriptional events during adipogenesis and the interplay among adipogenic transcription factors provides a basis for understanding the physiological processes associated with adipose tissue as well as for the development of therapeutic intervention of adipocyte related diseases. J. Cell. Biochem. Suppls. 32/33:59-67, 1999.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Gene Expression Regulation , Milk Proteins , Transcription, Genetic/genetics , Animals , CCAAT-Enhancer-Binding Proteins , Cell Differentiation , DNA-Binding Proteins/metabolism , MEF2 Transcription Factors , Myogenic Regulatory Factors , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Response Elements/genetics , STAT5 Transcription Factor , Trans-Activators/metabolism , Transcription Factors/metabolism
15.
J Cell Physiol ; 174(3): 285-92, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9462690

ABSTRACT

Fas, which functions to initiate a signal causing apoptosis, is expressed in epithelia, thus, suggesting a role in controlling cell number during states of cell and matrix turnover. In view of this, we hypothesized that cell-matrix interactions may be an important determinant of Fas expression in epithelial cells. To investigate this, we examined the effect of insoluble extracellular matrix molecules on Fas expression in murine lung epithelial (MLE) cells, a transformed mouse lung epithelial cell line. We report that 1) insoluble extracellular matrices increased Fas mRNA in a time and concentration-dependent manner; 2) induced increases in Fas mRNA were associated with concomitantly increased Fas protein; and 3) nonspecific adherence to a polylysine substrate did not induce Fas mRNA. Consistent with these findings, Fas-induced apoptosis was significantly enhanced in cultures plated on type IV collagen. Employing rat hepatocytes, we confirmed that the insoluble extracellular matrix also increases Fas expression in primary epithelial cells. By amplifying Fas-mediated apoptosis, these data suggest a mechanism whereby the extracellular matrix regulates the fate of specific epithelial cell populations.


Subject(s)
Apoptosis/physiology , Cell Communication , Epithelial Cells/metabolism , Epithelial Cells/physiology , Extracellular Matrix/physiology , fas Receptor/biosynthesis , Animals , Cell Line, Transformed , Cells, Cultured , Extracellular Matrix Proteins/physiology , Liver/cytology , Lung/cytology , Mice , RNA, Messenger/biosynthesis , Rats , Solubility , fas Receptor/genetics
16.
J Clin Invest ; 101(1): 22-32, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9421462

ABSTRACT

To define the molecular mechanisms that control GLUT4 expression during adipogenesis, NIH-3T3 fibroblasts ectopically expressing different adipogenic transcription factors (C/EBPbeta, C/EBPdelta, C/EBPalpha, and PPARgamma) under the control of a tetracycline-responsive inducible (C/EBPs) or a constitutive retroviral (PPARgamma) expression system were used. Enhanced production of C/EBPbeta (beta2 cell line), C/EBPbeta together with C/EBPdelta (beta/delta39 cell line), C/EBPalpha (alpha1 cell line), or PPARgamma (Pgamma2 cell line) in cells exposed to dexamethasone and the PPARgamma ligand ciglitazone (a thiazolidinedione) resulted in expression of GLUT4 mRNA as well as other members of the adipogenic gene program, including aP2 and adipsin. Focusing our studies on the beta/delta39 cells, we have demonstrated that C/EBPbeta along with C/EBPdelta in the presence of dexamethasone induces PPARgamma, adipsin, and aP2 mRNA production; however, GLUT4 mRNA is only expressed in cells exposed to ciglitazone. In addition, enhanced expression of a ligand-activated form of PPARgamma in the beta/delta39 fibroblasts stimulates synthesis of GLUT4 protein and gives rise to a population of adipocytic cells that take up glucose in direct response to insulin. C/EBPalpha is not expressed in the beta/delta39 cells under conditions that stimulate the adipogenic program. This observation suggests that PPARgamma alone or in combination with C/EBPbeta and C/EBPdelta is capable of activating GLUT4 gene expression.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/metabolism , Monosaccharide Transport Proteins/biosynthesis , Muscle Proteins , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazolidinediones , Transcription Factors/metabolism , 3T3 Cells , Adaptor Protein Complex 2 , Adaptor Protein Complex alpha Subunits , Adaptor Proteins, Vesicular Transport , Adipocytes/cytology , Animals , CCAAT-Enhancer-Binding Proteins , Cell Differentiation , Complement Factor D , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Dexamethasone/pharmacology , Gene Expression , Glucose Transporter Type 4 , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Ligands , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Monosaccharide Transport Proteins/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Thiazoles/pharmacology , Transcription Factors/biosynthesis , Transcription Factors/genetics
17.
Mol Cell Biol ; 16(11): 5964-73, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8887626

ABSTRACT

It is now well-recognized that the mitogen-activated protein (MAP) kinase cascade facilitates signaling from an activated tyrosine kinase receptor to the nucleus. In fact, an increasing number of extracellular effectors have been reported to activate the MAP kinase cascade, with a significant number of cellular responses attributed to this activation. We set out to explore how two extracellular effectors, basic fibroblast growth factor (bFGF) and insulin-like growth factor 1 (IGF-1), which have both been reported to activate MAP kinase, generate quite distinct cellular responses in C2C12 myoblasts. We demonstrate here that bFGF, which is both a potent mitogen and inhibitor of myogenic differentiation, is a strong MAP kinase agonist. By contrast, IGF-1, which is equally mitogenic for C2C12 cells but ultimately enhances the differentiated phenotype, is a weak activator of the MAP kinase cascade. We further demonstrate that IGF-1 is a potent activator of both insulin receptor substrate IRS-1 tyrosyl phosphorylation and association of IRS-1 with activated phosphatidylinositol 3-kinase (PI 3-kinase). Finally, use of the specific MAP kinase kinase inhibitor, PD098059, and wortmannin, a PI 3-kinase inhibitor, suggests the existence of an IGF-1-induced, MAP kinase-independent signaling event which contributes to the mitogenic response of this factor, whereas bFGF-induced mitogenesis appears to strongly correlate with activation of the MAP kinase cascade.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division/physiology , Fibroblast Growth Factor 2/pharmacology , Insulin-Like Growth Factor I/pharmacology , Signal Transduction , Androstadienes/pharmacology , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , DNA/biosynthesis , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Insulin Receptor Substrate Proteins , Kinetics , Mice , Mitogen-Activated Protein Kinase Kinases , Muscle, Skeletal , Phenotype , Phosphatidylinositol 3-Kinases , Phosphoproteins/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotyrosine/analysis , Protein Kinase Inhibitors , Recombinant Proteins , Signal Transduction/drug effects , Wortmannin
18.
Mol Cell Biol ; 16(8): 4128-36, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8754811

ABSTRACT

The differentiation of 3T3 preadipocytes into adipocytes is accompanied by a transient induction of C/EBPbeta and C/EBPdelta expression in response to treatment of the cells with methylisobutylxanthine (MIX) and dexamethasone (DEX), respectively. In this report, we demonstrate that peroxisome proliferator-activated receptor gamma (PPARgamma) expression in 3T3-L1 preadipocytes is induced by MIX and DEX, suggesting that C/EBPbeta and C/EBPdelta may be involved in this process. Using a tetracycline-responsive expression system, we have recently shown that the conditional ectopic expression of C/EBPbeta in NIH 3T3 fibroblasts (beta2 cells) in the presence of DEX activates the synthesis of peroxisome PPARgamma mRNA. Subsequent exposure of these cells to PPAR activators stimulates their conversion into adipocytes; however, neither the expression of C/EBPbeta nor exposure to DEX alone is capable of inducing PPARgamma expression in the beta2 cell line. We find that unlike the case for 3T3 preadipocytes, C/EBPdelta is not induced by DEX in these 3T3 fibroblasts and therefore is not relaying the effect of this glucocorticoid to the PPARgamma gene. To define the role of glucocorticoids in regulating PPARgamma expression and the possible involvement of C/EBPdelta, we have established an additional set of NIH 3T3 cell lines expressing either C/EBPdelta alone (delta23 cells) or C/EBPdelta and C/EBPbeta together (beta/delta39 cells), using the tetracycline-responsive system. Culture of these cells in tetracycline-deficient medium containing DEX, MIX, insulin, and fetal bovine serum shows that the beta/delta39 cells express PPARgamma and aP2 mRNAs at levels that are almost equivalent to those observed in fully differentiated 3T3-L1 adipocytes. These levels are approximately threefold higher than their levels of expression in the beta2 cells. Despite the fact that these beta/delta39 cells produce abundant amounts of C/EBPbeta and C/EBPdelta (in the absence of tetracycline), they still require glucocorticoids to attain maximum expression of PPARgamma mRNA. Furthermore, the induction of PPARgamma mRNA by exposure of these cells to DEX occurs in the absence of ongoing protein synthesis. The delta23 cells, on the other hand, are not capable of activating PPARgamma gene expression when exposed to the same adipogenic inducers. Finally, attenuation of ectopic C/EBPbeta production at various stages during the differentiation process results in a concomitant inhibition of PPARgamma and the adipogenic program. These data strongly suggest that the induction of PPARgamma gene expression in multipotential mesenchymal stem cells (NIH 3T3 fibroblasts) is dependent on elevated levels of C/EBPbeta throughout the differentiation process, as well as an initial exposure to glucocorticoids. C/EBPdelta may function by synergizing with C/EBPbeta to enhance the level of PPARgamma expression.


Subject(s)
Adipocytes/cytology , DNA-Binding Proteins/physiology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Nuclear Proteins/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , 3T3 Cells/physiology , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins , Cell Differentiation , DNA-Binding Proteins/metabolism , Estradiol/pharmacology , Gene Expression Regulation, Developmental , Insulin/pharmacology , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/genetics , Tetracycline/pharmacology , Transcription Factor AP-2 , Transcription Factors/metabolism , Triamcinolone Acetonide/pharmacology , Up-Regulation
19.
In Vitro Cell Dev Biol Anim ; 32(2): 90-9, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8907122

ABSTRACT

Our previous studies have demonstrated that expression of growth-associated genes is regulated by the adhesive state of the cell. To understand the role of cell adhesion in regulating the switch from growth to differentiation, we are studying the differentiation of mouse myoblasts into multinucleated contractile myotubes. In this report, we describe a novel means of culturing C2C12 myoblasts that permits an analysis of the role of cell adhesion in regulating the sequential induction of muscle-specific genes that control myogenesis. Suspension of an asynchronous, proliferating population of myoblasts in a viscous gel of methylcellulose dissolved in medium containing 20% serum induces growth arrest in G0 phase of the cell cycle without a concomitant induction of muscle-specific genes. Reattachment to a solid substratum in 20% serum, 0.5 nM bFGF, or 10 nM IGF-1 rapidly activates entry of the quiescent cells into G1 followed by a synchronous progression of the cell population through into S phase. bFGF or IGF-1 added separately facilitate only one passage through the cell cycle, whereas 20% serum or the two growth factors added together support multiple cell divisions. Adhesion of suspended cells in DMEM alone or with 3 nM IGF-1 induces myogenesis as evidenced by the synthesis of myogenin and myosin heavy chain (MHC) proteins followed by fusion into myotubes. bFGF completely inhibits this differentiation process even in the presence of myogenic doses of IGF-1. Addition of 3 nM IGF-1 to quiescent myoblasts maintained in suspension culture in serum-free conditions does not induce myogenin or MHC expression. Thus, adhesion is a requirement for the induction of muscle gene expression in mouse myoblasts. The development of a muscle cell culture environment in which proliferating myoblasts can be growth arrested in G0 without activating muscle-specific gene expression provides a means of analyzing the synchronous activation of either the myogenic or growth programs and how adhesion affects each process, respectively.


Subject(s)
Cell Adhesion , Muscle Fibers, Skeletal/metabolism , MyoD Protein/metabolism , Myogenin/metabolism , Signal Transduction/physiology , Animals , Cell Cycle/physiology , Cell Differentiation , Cell Division , Culture Media , Fibroblast Growth Factor 2/pharmacology , Gene Expression , Insulin-Like Growth Factor I/pharmacology , Mice , Mitogens/pharmacology , Muscle Fibers, Skeletal/cytology , Resting Phase, Cell Cycle
20.
Genes Dev ; 9(19): 2350-63, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7557387

ABSTRACT

Activation of adipogenesis in 3T3 preadipocytes by exposure to the adipogenic inducers dexamethasone, methylisobutylxanthine, insulin, and fetal bovine serum is accompanied by a transient burst of C/EBP beta protein expression that precedes the induction of the fat gene program. In this study we have investigated the role of C/EBP beta in initiating the adipogenic program by overexpressing C/EBP beta in multipotential NIH-3T3 fibroblasts. Conditional ectopic expression of C/EBP beta was accomplished by using an artificial transcriptional regulatory system based on the Escherichia coli tetracycline repressor to generate a stable cell line, beta 2, that expresses C/EBP beta mRNA and protein in a tightly controlled tetracycline dose-dependent manner. Induction of C/EBP beta DNA-binding activity in NIH-3T3 beta 2 cells exposed to dexamethasone in the presence of insulin and fetal bovine serum activates the expression of an adipocyte-specific nuclear hormone receptor, PPAR gamma, that stimulates the conversion of these fibroblasts into committed preadipocytes. Either ectopic expression of C/EBP beta or treatment with dexamethasone alone is incapable of inducing PPAR gamma expression, but when present together, they have a synergistic effect on the adipogenic program. Exposure of these stimulated cells to a PPAR activator 5,8,11,14-eicosatetraynoic acid (ETYA) results in the accumulation of fat droplets and expression of the adipocyte-enriched genes aP2 and glycerol phosphate dehydrogenase (GPD). The number of beta 2 cells that can differentiate into adipocytes is related to the concentration of tetracycline and, therefore, the amount of the exogenous C/EBP beta protein expressed. C/EBP beta can induce PPAR gamma mRNA in the absence of ETYA; however, expression of aP2 mRNA and maximum fat deposition is dependent on the PPAR activator. Our results suggest that enhanced expression of C/EBP beta converts multipotential mesenchymal precursor cells into preadipocytes that respond to adipogenic inducers, including dexamethasone and PPAR activators to differentiate into adipocytes.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , 1-Methyl-3-isobutylxanthine/pharmacology , 3T3 Cells , 5,8,11,14-Eicosatetraynoic Acid/pharmacology , Adipocytes/cytology , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins , Cell Differentiation/genetics , Cell Fractionation , DNA/metabolism , DNA Probes , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Gene Expression Regulation, Developmental , Insulin/pharmacology , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , Tetracycline/pharmacology , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...