Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Mol Cell Oncol ; 5(5): e1409862, 2018.
Article in English | MEDLINE | ID: mdl-30263936

ABSTRACT

Cyclin-dependent kinase 4 (CDK4) is a positive regulator of cell cycle progression, however, there is growing evidence demonstrating that its function exceeds the control of cell division. Here we show that CDK4 is an important regulator of cellular substrate utilization through direct inhibition of the metabolic regulator AMPK (AMP-activated protein kinase).

2.
Cell Mol Life Sci ; 75(6): 975-987, 2018 03.
Article in English | MEDLINE | ID: mdl-28988292

ABSTRACT

In the course of the last decades, metabolism research has demonstrated that adipose tissue is not an inactive tissue. Rather, adipocytes are key actors of whole body energy homeostasis. Numerous novel regulators of adipose tissue differentiation and function have been identified. With the constant increase of obesity and associated disorders, the interest in adipose tissue function alterations in the XXIst century has become of paramount importance. Recent data suggest that adipocyte differentiation, adipose tissue browning and mitochondrial function, lipogenesis and lipolysis are strongly modulated by the cell division machinery. This review will focus on the function of cell cycle regulators in adipocyte differentiation, adipose tissue function and whole body energy homeostasis; with particular attention in mouse studies.


Subject(s)
Adipose Tissue/metabolism , Cell Cycle Proteins/metabolism , Energy Metabolism , Animals , Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/metabolism , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Humans
3.
Oncogene ; 32(42): 5101-10, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-23208508

ABSTRACT

One of the most conserved features of all cancers is a profound reprogramming of cellular metabolism, favoring biosynthetic processes and limiting catalytic processes. With the acquired knowledge of some of these important changes, we have designed a combination therapy in order to force cancer cells to use a particular metabolic pathway that ultimately results in the accumulation of toxic products. This innovative approach consists of blocking lipid synthesis, at the same time that we force the cell, through the inhibition of AMP-activated kinase, to accumulate toxic intermediates, such as malonyl-coenzyme A (malonyl-CoA) or nicotinamide adenine dinucleotide phosphate. This results in excess of oxidative stress and cancer cell death. Our new therapeutic strategy, based on the manipulation of metabolic pathways, will certainly set up the basis for new upcoming studies defining a new paradigm of cancer treatment.


Subject(s)
Molecular Targeted Therapy/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Fatty Acid Synthase, Type I/antagonists & inhibitors , Fatty Acid Synthase, Type I/metabolism , Humans , Male , Malonyl Coenzyme A/metabolism , Mice, Nude , NADP/metabolism , NADPH Oxidases/metabolism , Oxidative Stress , Xenograft Model Antitumor Assays
4.
Cell Death Differ ; 18(1): 109-21, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20596077

ABSTRACT

We have previously demonstrated that the thiazolidinedione ciglitazone inhibited, independently of PPARγ activation, melanoma cell growth. Further investigations now show that ciglitazone effects are mediated through the regulation of secreted factors. Q-PCR screening of several genes involved in melanoma biology reveals that ciglitazone inhibits expression of the CXCL1 chemokine gene. CXCL1 is overexpressed in melanoma and contributes to tumorigenicity. We show that ciglitazone induces a diminution of CXCL1 level in different human melanoma cell lines. This effect is mediated by the downregulation of microphthalmia-associated transcription factor, MITF, the master gene in melanocyte differentiation and involved in melanoma development. Further, recombinant CXCL1 protein is sufficient to abrogate thiazolidinedione effects such as apoptosis induction, whereas extinction of the CXCL1 pathway mimics phenotypic changes observed in response to ciglitazone. Finally, inhibition of human melanoma tumor development in nude mice treated with ciglitazone is associated with a strong decrease in MITF and CXCL1 levels. Our results show that anti-melanoma effects of thiazolidinediones involve an inhibition of the MITF/CXCL1 axis and highlight the key role of this specific pathway in melanoma malignancy.


Subject(s)
Antineoplastic Agents/therapeutic use , Chemokine CXCL1/metabolism , Melanoma/drug therapy , Microphthalmia-Associated Transcription Factor/metabolism , Thiazolidinediones/therapeutic use , Animals , Apoptosis , Cell Differentiation , Cell Line, Tumor , Chemokine CXCL1/genetics , Chemokine CXCL1/pharmacology , Down-Regulation , Humans , Melanoma/metabolism , Mice , Mice, Nude , Microphthalmia-Associated Transcription Factor/antagonists & inhibitors , Microphthalmia-Associated Transcription Factor/physiology , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , Transplantation, Heterologous
5.
Oncogene ; 29(31): 4369-77, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20514019

ABSTRACT

Cancer development involves major alterations in cells' metabolism. Enhanced glycolysis and de novo fatty acids synthesis are indeed characteristic features of cancer. Cell proliferation and metabolism are tightly linked cellular processes. Others and we have previously shown a close relationship between metabolic responses and proliferative stimuli. In addition to trigger proliferative and survival signaling pathways, most oncoproteins also trigger metabolic changes to transform the cell. We present herein the view that participation of cell-cycle regulators and oncogenic proteins to cancer development extend beyond the control of cell proliferation, and discuss how these new functions may be implicated in metabolic alterations concomitant to the pathogenesis of human cancers.


Subject(s)
Cell Proliferation , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction/physiology , Animals , Cell Cycle/physiology , Humans , Lipid Metabolism/physiology , Models, Biological , Neoplasms/physiopathology , Signal Transduction/genetics , Transcription Factors/metabolism , Transcription Factors/physiology
6.
Int J Obes (Lond) ; 29 Suppl 1: S10-2, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15711575

ABSTRACT

Regulation of peroxisome proliferator-activated receptor gamma (PPARgamma) activity is the result of several events. The first control level is the regulation of the expression of PPARgamma. Examples of this regulation, during adipogenesis, is the transactivation of the PPARgamma promoter by transcription factors of the classical pathway, such as C/EBPs or ADD1/SREBP1, but also newly identified factors, such as E2Fs. When preadipocytes re-enter the cell cycle, PPARgamma expression is induced coincident with an increase in DNA synthesis, suggesting the involvement of the E2F family of cell cycle regulators. E2F1 induces PPARgamma transcription during clonal expansion, whereas E2F4 represses PPARgamma expression during terminal adipocyte differentiation. Hence, E2Fs represent the link between proliferative signaling pathways, triggering clonal expansion, and terminal adipocyte differentiation through regulation of PPARgamma expression. A second regulatory level of PPARgamma action is interaction with cofactors. We will focus our attention on the atypical PPARgamma modulators. We have described an interaction between PPARgamma and the retinoblastoma protein, RB, which is both dependent upon ligand binding by PPARgamma and upon the phosphorylation status of RB. The interaction between PPARgamma and RB decreases the transcriptional activity of PPARgamma through recruitment of the histone deacetylase HDAC3. Inhibition of HDAC activity consequently results in a strong activation of PPARgamma.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , Obesity/metabolism , PPAR gamma/genetics , Signal Transduction/physiology , Adipocytes/cytology , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , E2F4 Transcription Factor , Histone Deacetylases/metabolism , Humans , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism
7.
J Mol Endocrinol ; 27(1): 1-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11463572

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors, initially described as molecular targets for synthetic compounds inducing peroxisome proliferation. PPAR-gamma, the best characterized of the PPARs, plays a crucial role in adipogenesis and insulin sensitization. Furthermore, PPAR-gamma has been reported to affect cell proliferation/differentiation pathways in various malignancies. We discuss in the present review recent advances in the understanding of the function of PPAR-gamma in both cell proliferation and adipocyte differentiation.


Subject(s)
Adipose Tissue/cytology , Cell Transformation, Neoplastic , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Cell Differentiation/physiology , Cell Division/physiology , Humans
8.
Nutr Metab Cardiovasc Dis ; 11(1): 64-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11383325

ABSTRACT

The peroxisome proliferator-activated receptor gamma is a nuclear hormone receptor playing a crucial role in adipogenesis and insulin sensitization. Prostaglandin J2 derivatives and the antidiabetic thiazolidinediones are its respective natural and synthetic ligands. The RXR/PPAR gamma heterodimer has also been reported to have important immunomodulatory activities and its pleiotropic functions suggest wide-ranging medical implications.


Subject(s)
Adipose Tissue/metabolism , Insulin/physiology , Prostaglandin D2/analogs & derivatives , Receptors, Cytoplasmic and Nuclear/physiology , Thiazolidinediones , Transcription Factors/physiology , Animals , Arteriosclerosis/etiology , Gene Expression , Humans , Inflammation/etiology , Ligands , Prostaglandin D2/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazoles , Transcription Factors/genetics , Transcription Factors/metabolism
9.
J Mol Med (Berl) ; 79(1): 30-47, 2001.
Article in English | MEDLINE | ID: mdl-11327101

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors, initially described as molecular targets for synthetic compounds that induce peroxisome proliferation. PPARgamma is the best characterized of the PPARs. The heterodimer of PPARgamma with the retinoid X receptor (RXR) plays a crucial role in adipogenesis and insulin sensitization. The RXR/PPARgamma heterodimer furthermore has been reported to have important immunomodulatory activities and to affect cell proliferation/differentiation pathways in various malignancies. PPARgamma is activated by a number of naturally occurring fatty acid derivatives and by several synthetic compounds, including the thiazolidinediones and L-tyrosine-based insulin sensitizers. This review gives an overview of the pleiotropic functions of PPARgamma and discusses the wide-ranging medical implications that modulation of PPARgamma activity might have for various diseases, ranging from obesity and type 2 diabetes to cancer and inflammation.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Adipose Tissue/metabolism , Animals , Arteriosclerosis/etiology , Colon/metabolism , Diabetes Mellitus, Type 2/etiology , Humans , Inflammation/etiology , Insulin/pharmacology , Mice , Neoplasms/metabolism , Obesity/etiology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors
10.
Mol Cell Biol ; 21(8): 2956-66, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283272

ABSTRACT

E4F is a ubiquitously expressed GLI-Krüppel-related transcription factor which has been identified for its capacity to regulate transcription of the adenovirus E4 gene in response to E1A. However, cellular genes regulated by E4F are still unknown. Some of these genes are likely to be involved in cell cycle progression since ectopic p120E4F expression induces cell cycle arrest in G1. Although p21WAF1 stabilization was proposed to mediate E4F-dependent cell cycle arrest, we found that p120E4F can induce a G1 block in p21(-/-) cells, suggesting that other proteins are essential for the p120E4F-dependent block in G1. We show here that cyclin A promoter activity can be repressed by p120E4F and that this repression correlates with p120E4F binding to the cyclic AMP-responsive element site of the cyclin A promoter. In addition, enforced expression of cyclin A releases p120E4F-arrested cells from the G1 block. These data identify the cyclin A gene as a cellular target for p120E4F and suggest a mechanism for p120E4F-dependent cell cycle regulation.


Subject(s)
Cyclin A/metabolism , G1 Phase/physiology , Transcription Factors/metabolism , 3T3 Cells , Animals , Base Sequence , Binding Sites/genetics , Cell Line , Cricetinae , Cyclin A/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , DNA/genetics , DNA/metabolism , DNA Primers/genetics , GA-Binding Protein Transcription Factor , Gene Expression , Mice , Mice, Knockout , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Transcription Factors/genetics
11.
Proc Natl Acad Sci U S A ; 97(14): 7738-43, 2000 Jul 05.
Article in English | MEDLINE | ID: mdl-10869426

ABSTRACT

The retinoblastoma protein pRB is involved in the transcriptional control of genes essential for cell cycle progression and differentiation. pRB interacts with different transcription factors and thereby modulates their activity by sequestration, corepression, or activation. We report that pRB, but not p107 and p130, binds to and facilitates repression by p120(E4F), a ubiquitously expressed GLI-Kruppel-related protein identified as a cellular target of E1A. The interaction involves two distinct regions of p120(E4F) and the C-terminal part of pRB. In vivo pRB-p120(E4F) complexes can only be detected in growth-arrested cells, and accordingly contain the hypophosphorylated form of pRB. Repression of an E4F-responsive promoter is strongly increased by combined expression of p120(E4F) and pRB, which correlates with pRB-dependent enhancement of p120(E4F) binding activity. Elevated levels of p120(E4F) have been shown to block growth of mouse fibroblasts in G(1). We find this requires pRB, because RB(-/-) fibroblasts are significantly less sensitive to excess p120(E4F).


Subject(s)
Adenovirus E1A Proteins/metabolism , Adenovirus E4 Proteins/metabolism , Repressor Proteins/metabolism , Retinoblastoma Protein/metabolism , Adenovirus E4 Proteins/genetics , Animals , Binding Sites , Cell Division , Growth Inhibitors , Mice , Mutation , Protein Binding , Repressor Proteins/genetics , Zinc Fingers
12.
Curr Biol ; 10(9): 543-6, 2000 May 04.
Article in English | MEDLINE | ID: mdl-10801445

ABSTRACT

During skeletal muscle differentiation, a subset of myoblasts remains quiescent and undifferentiated but retains the capacity to self-renew and give rise to differentiating myoblasts [1] [2] [3]: this sub-population of muscle cells was recently termed 'reserve cells' [3]. In order to characterise genes that can regulate the ratio between reserve cells and differentiating myoblasts, we examined members of the retinoblastoma tumor suppressor family - Rb, p107 and p130 - an important family of negative regulators of E2F transcription factors and cell cycle progression [4]. Although pRb and p107 positively regulate muscle cell differentiation [5] [6] [7], the role of p130 in muscle cells remains unknown. We show here that p130 (protein and mRNA), but neither pRb nor p107, preferentially accumulates during muscle differentiation in reserve cells. Also, p130 is the major Rb-family protein present in E2F complexes in this sub-population of cells. Although forced expression of either p130 or pRb in mouse C2 myoblasts efficiently blocked cell cycle progression, only p130 inhibited the differentiation program. Furthermore, muscle cells overexpressing p130 had reduced levels of the muscle-promoting factor MyoD. In addition, p130 repressed the transactivation capacity of MyoD, an effect abolished by co-transfection of pRb. Thus, we propose that p130, by blocking cell cycle progression and differentiation, could be part of a specific pathway that defines a pool of reserve cells during terminal differentiation.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Muscle, Skeletal/cytology , Phosphoproteins/metabolism , Proteins , Animals , Cell Cycle , Cell Differentiation , Cell Line , E2F Transcription Factors , Gene Expression Regulation , Mice , MyoD Protein/genetics , Phosphoproteins/genetics , Retinoblastoma-Binding Protein 1 , Retinoblastoma-Like Protein p130 , Stem Cells/cytology , Transcription Factor DP1 , Transcription Factors/metabolism , Transcriptional Activation
13.
FEBS Lett ; 471(1): 29-33, 2000 Apr 07.
Article in English | MEDLINE | ID: mdl-10760507

ABSTRACT

The bipartite repressor elements, termed cell cycle-dependent element (CDE)/cell cycle regulatory element (CCRE)-cell cycle homology region (CHR) control the growth-dependent transcription of the cyclin A, cdc25C, cdc2 genes. Here, we have identified a functional element displaying the signature of the CDE-CHR in the promoter of the mouse RB2 (p130) gene, encoding the retinoblastoma protein family (pRB)-related protein p130. This element locates close to the major transcription start site where it makes major groove contacts with proteins that can be detected in a cellular context using in vivo genomic footprinting techniques. Inactivation of either the CDE or CHR sequence strongly up-regulates the p130 promoter activity in exponentially growing cells, a situation where endogenous p130 gene expression is almost undetectable. Electrophoretic mobility shift assays suggest that two different protein complexes bind independently to the p130 CDE and CHR elements, and that the protein(s) bound to the CDE might be related to those bound on cyclin A and cdc2 promoters.


Subject(s)
Gene Expression Regulation , Phosphoproteins/genetics , Promoter Regions, Genetic , Proteins , Animals , Base Sequence , Cloning, Molecular , DNA , DNA-Binding Proteins/metabolism , Genes, cdc , Humans , Mice , Molecular Sequence Data , Mutation , Retinoblastoma-Like Protein p130 , Sequence Homology, Nucleic Acid , Transcription, Genetic , Up-Regulation
14.
Genes Chromosomes Cancer ; 28(1): 126-30, 2000 May.
Article in English | MEDLINE | ID: mdl-10738311

ABSTRACT

E2F transcription factors (E2F1 to 6) are central players in the control of animal cell proliferation as regulators of genes involved in cell cycle progression and in transformation. In this report, we have investigated the potential involvement of the E2F5 gene in tumorigenesis. We show that E2F5 can promote the formation of morphologically transformed foci in primary baby rat kidney cells (BRK) when it is overexpressed in the presence of its heterodimeric partner DP1 and activated RAS. This suggests that E2F5 behaves like a MYC-type cooperating oncogene in functional assays, prompting us to monitor potential amplifications of the E2F5 gene in primary human tumors. We mapped the human E2F5 gene to 8q21.1-21.3 equidistant from the MOS (8q12) and MYC (8q24) oncogenes. Since the long arm of chromosome 8 is frequently the site of increased gene copy number (ICN) in breast cancer, we screened 442 breast tumor DNAs for gains of E2F5, MOS, and MYC genes. The three genes showed ICN, albeit at variable incidence and levels of amplification, with the ICN of E2F5 occurring concomitantly with those of MOS and/or MYC in almost half of the cases. Moreover, a marked increase of the 2. 5-kb E2F5 transcript was also detected in some tumors and tumor cell lines. In conclusion, the evidence that sustained unregulated expression of E2F5 can cooperate with other oncogenes to promote cell transformation in functional assays, together with the detection of chromosomal amplifications and overexpressions of the E2F5 gene in breast tumors, provides the first indications that E2F5 deregulation may have a role in human tumor development.


Subject(s)
Breast Neoplasms/genetics , Gene Amplification/genetics , Oncogenes/genetics , Transcription Factors/genetics , Animals , E2F5 Transcription Factor , Gene Dosage , Humans , Rats , Rats, Sprague-Dawley , Transcription Factors/biosynthesis , Tumor Cells, Cultured
15.
Int J Obes Relat Metab Disord ; 24(2): 195-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10702770

ABSTRACT

OBJECTIVE: The Pro12Ala polymorphism of the Peroxisome Proliferator Activated Receptor gamma2 (PPARgamma2) gene has been inconsistently associated with body mass index variations and non-insulin-dependent diabetes mellitus (NIDDM). We investigated the impact of this polymorphism on obesity markers, lipid and glucose variables in a sample of French subjects and evaluated its possible role in the onset of NIDDM. DESIGN AND SUBJECTS: Within the framework of the WHO-MONICA project, a population study composed of 1195 subjects aged 35-64 y was randomly sampled from the electoral rolls of the urban community of Lille, in northern France. Subjects receiving medical treatment for hypercholesterolemia, hypertension or diabetes mellitus were excluded for the analyses, to avoid any interferences between medical treatment and biological variables. This resulted in a sample size of 839 subjects (421 men/418 women, age=49.4+/-8.1 y, body mass index (BMI)=25.7+/-4.4 kg/m2). To evaluate the role of the Pro12Ala polymorphism in the onset of NIDDM, we evaluated its distribution in 170 Caucasian NIDDM subjects from a clinical series (117 men/53 women, age=62.3+/-9.0 y, BMI=30.1+/-3.6 kg/m2). MEASUREMENTS: The PPARgamma2 Pro12Ala polymorphism genotyping was carried out with allele specific oligonucleotides hybridisation. Data were statistically analysed for association with various obesity markers (body weight (BW), BMI, waist-to-hip ratio (WHR), plasma leptin concentrations, lipid and glucose variables. RESULTS: In the WHO-MONICA population, the Ala allele frequency was 0.11. The presence of the Ala allele was significantly associated with higher body weight (P=0.002), BMI (P=0.02), height (P=0.02) and waist circumference (P=0.04). Increased plasma concentrations of total cholesterol (P=0.01), LDL-cholesterol (P=0.004) and apolipoprotein B (P=0.01) were also detected in Ala allele bearers. The distribution of the Pro12Ala polymorphism was similar in NIDDM subjects (Ala allele frequency: 0.10) and in the WHO-MONICA population subjects. CONCLUSION: Our results suggest that genetic variability of PPARgamma2 affects body weight control and lipid homeostasis in humans and do not support a significant role for the PPARgamma2 Pro12Ala polymorphism in the aetiology of NIDDM. International Journal of Obesity (2000) 24, 195-199


Subject(s)
Diabetes Mellitus, Type 2/genetics , Hyperlipidemias/genetics , Obesity/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , White People/genetics , Adult , Alleles , Body Mass Index , DNA Primers , Female , France , Gene Frequency , Humans , Male , Middle Aged , Polymerase Chain Reaction , Polymorphism, Genetic
16.
J Endocrinol ; 162(3): 331-40, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10467224

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARgamma), a fatty acid-activated nuclear receptor, is implicated in adipocyte differentiation and insulin sensitisation. In view of the association of dietary fat intake and bowel disease, the expression of PPARgamma in rodent and human intestine was studied. Expression of PPARgamma mRNA was examined by Northern blot hybridisation, RNase protection, and/or competitive RT-PCR assays, whereas PPARgamma protein levels were evaluated by immunoblotting and immunohistochemistry. PPARgamma mRNA and protein were abundantly expressed in colon relative to the small intestine both in rodents and in man. Interestingly, expression of PPARgamma was primarily localised in the more differentiated epithelial cells in the colon. The level of expression of PPARgamma in colon was similar to the levels seen in adipose tissue. Expression of PPARgamma increased from proximal to distal segments of the colon in man. In Caco-2 and HT-29 human adenocarcinoma cells, PPARgamma expression increased upon differentiation, consistent with PPARgamma being associated with a differentiated epithelial phenotype. High-level expression of PPARgamma was observed in the colon, but not in the small intestine, suggesting a potential role of this nuclear receptor in the colon.


Subject(s)
Colon/chemistry , Receptors, Cytoplasmic and Nuclear/analysis , Transcription Factors/analysis , Aged , Animals , Blotting, Northern , Caco-2 Cells , Cell Differentiation , Cells, Cultured , Epithelium/chemistry , HT29 Cells , Humans , Immunoblotting , Immunohistochemistry , Intestine, Small/chemistry , Male , Middle Aged , RNA, Messenger/analysis , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
17.
Mol Cell Biol ; 19(8): 5495-503, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10409739

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARgamma) is a nuclear receptor implicated in adipocyte differentiation and insulin sensitivity. We investigated whether PPARgamma expression is dependent on the activity of adipocyte differentiation and determination factor 1/sterol regulatory element binding protein 1 (ADD-1/SREBP-1), another transcription factor associated with both adipocyte differentiation and cholesterol homeostasis. Ectopic expression of ADD-1/SREBP-1 in 3T3-L1 and HepG2 cells induced endogenous PPARgamma mRNA levels. The related transcription factor SREBP-2 likewise induced PPARgamma expression. In addition, cholesterol depletion, a condition known to result in proteolytic activation of transcription factors of the SREBP family, induced PPARgamma expression and improved PPRE-driven transcription. The effect of the SREBPs on PPARgamma expression was mediated through the PPARgamma1 and -3 promoters. Both promoters contain a consensus E-box motif that mediates the regulation of the PPARgamma gene by ADD-1/SREBP-1 and SREBP-2. These results suggest that PPARgamma expression can be controlled by the SREBP family of transcription factors and demonstrate new interactions between transcription factors that can regulate different pathways of lipid metabolism.


Subject(s)
Adipocytes/metabolism , CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Nuclear Proteins/physiology , Receptors, Cytoplasmic and Nuclear/biosynthesis , Transcription Factors/biosynthesis , Adipocytes/cytology , Cell Differentiation/genetics , Cholesterol/metabolism , Cholesterol/pharmacology , Consensus Sequence , Fatty Acids/pharmacology , Gene Expression Regulation, Developmental/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipid Metabolism , Multigene Family , Peroxisome Proliferators/pharmacology , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/genetics , Simvastatin/pharmacology , Sterol Regulatory Element Binding Protein 1 , Transcription Factors/genetics , Transcription Factors/physiology
19.
J Biol Chem ; 274(12): 7681-8, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10075656

ABSTRACT

The nuclear peroxisome proliferator-activated receptor gamma (PPARgamma) activates the transcription of multiple genes involved in intra- and extracellular lipid metabolism. Several cofactors are crucial for the stimulation or the silencing of nuclear receptor transcriptional activities. The two homologous cofactors p300 and CREB-binding protein (CBP) have been shown to co-activate the ligand-dependent transcriptional activities of several nuclear receptors as well as the ligand-independent transcriptional activity of the androgen receptor. We show here that the interaction between p300/CBP and PPARgamma is complex and involves multiple domains in each protein. p300/CBP not only bind in a ligand-dependent manner to the DEF region of PPARgamma but also bind directly in a ligand-independent manner to a region in the AB domain localized between residue 31 to 99. In transfection experiments, p300/CBP could thereby enhance the transcriptional activities of both the activating function (AF)-1 and AF-2 domains. p300/CBP displays itself at least two docking sites for PPARgamma located in its N terminus (between residues 1 and 113 for CBP) and in the middle of the protein (between residues 1099 and 1460).


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazolidinediones , Trans-Activators/metabolism , Transcription Factors/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , HeLa Cells , Humans , Ligands , Rosiglitazone , Thiazoles/pharmacology , Zinc Fingers
20.
Cell Growth Differ ; 10(1): 43-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9950217

ABSTRACT

Adipocyte differentiation is driven by the expression and activation of three transcription factor families: the differentially expressed CAAT/enhancer binding proteins (C/EBPs) alpha, beta, and delta; the helix-loop-helix adipocyte differentiation and determination factor-1; and peroxisome proliferator activated receptor gamma (PPARgamma), expressed as two isoforms, PPARgamma1 and the adipocyte-specific PPARgamma2. Overexpression of PPARgamma can induce adipocyte differentiation; therefore, we analyzed the expression of the two PPARgamma isoforms during early stages of differentiation to determine whether one was preferentially induced as an early determining event. Surprisingly, in the first 24 h, a 3-6-fold increase of PPARgamma2 mRNA was observed, whereas PPARgamma1 mRNA remained unchanged. PPARgamma1 was induced 1 day later. Overexpression of C/EBPbeta has also been shown to induce adipocyte differentiation. A C/EBP site was identified only in the human PPARgamma2 promoter. Its deletion blunted the response of PPARgamma2 promoter to cotransfected C/EBPbeta or methylisobutylxanthine treatment. We hypothesize that PPARgamma2 initiates adipocyte differentiation.


Subject(s)
Adipocytes/cytology , Gene Expression Regulation , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , 3T3 Cells , Animals , CCAAT-Enhancer-Binding Proteins , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Isoforms , RNA, Messenger , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...