Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Biol Chem ; 276(32): 29681-7, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11397807

ABSTRACT

Activation of the nuclear hormone peroxisome proliferator-activated receptor gamma (PPARgamma) inhibits cell growth and promotes differentiation in a broad spectrum of epithelial derived tumor cell lines. Here we utilized microarray technology to identify PPARgamma gene targets in intestinal epithelial cells. For each gene, the induction or repression was seen with two structurally distinct PPARgamma agonists, and the change in expression could be blocked by co-treatment with a specific PPARgamma antagonist. A majority of the genes could be regulated independently by a retinoid X receptor specific agonist. Genes implicated in lipid transport or storage (adipophilin and liver fatty acid-binding protein) were also activated by agonists of PPAR subtypes alpha and/or delta. In contrast, PPARgamma-selective targets included genes linked to growth regulatory pathways (regenerating gene IA), colon epithelial cell maturation (GOB-4 and keratin 20), and immune modulation (neutrophil-gelatinase-associated lipocalin). Additionally, three different genes of the carcinoembryonic antigen family were induced by PPARgamma. Cultured cells treated with PPARgamma ligands demonstrated an increase in Ca(2+)-independent, carcinoembryonic antigen-dependent homotypic aggregation, suggesting a potential role for PPARgamma in regulating intercellular adhesion. Collectively, these results will help define the mechanisms by which PPARgamma regulates intestinal epithelial cell biology.


Subject(s)
Colorectal Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Blotting, Northern , Blotting, Western , COS Cells , Cell Adhesion , Cell Division , Colorectal Neoplasms/genetics , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/metabolism , Ligands , Luciferases/metabolism , Mutation , Oligonucleotide Array Sequence Analysis , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Retinoic Acid/agonists , Retinoid X Receptors , Time Factors , Transcription Factors/agonists , Transcription Factors/physiology , Transfection , Tumor Cells, Cultured
2.
Proc Natl Acad Sci U S A ; 97(20): 10990-5, 2000 Sep 26.
Article in English | MEDLINE | ID: mdl-10984506

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARgamma) is a nuclear hormone receptor that plays a key role in the differentiation of adipocytes. Activation of this receptor in liposarcomas and breast and colon cancer cells also induces cell growth inhibition and differentiation. In the present study, we show that PPARgamma is expressed in human prostate adenocarcinomas and cell lines derived from these tumors. Activation of this receptor with specific ligands exerts an inhibitory effect on the growth of prostate cancer cell lines. Further, we show that prostate cancer and cell lines do not have intragenic mutations in the PPARgamma gene, although 40% of the informative tumors have hemizygous deletions of this gene. Based on our preclinical data, we conducted a phase II clinical study in patients with advanced prostate cancer using troglitazone, a PPARgamma ligand used for the treatment of type 2 diabetes. Forty-one men with histologically confirmed prostate cancer and no symptomatic metastatic disease were treated orally with troglitazone. An unexpectedly high incidence of prolonged stabilization of prostate-specific antigen was seen in patients treated with troglitazone. In addition, one patient had a dramatic decrease in serum prostate-specific antigen to nearly undetectable levels. These data suggest that PPARgamma may serve as a biological modifier in human prostate cancer and its therapeutic potential in this disease should be further investigated.


Subject(s)
Antineoplastic Agents/pharmacology , Chromans/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazoles/pharmacology , Thiazolidinediones , Transcription Factors/agonists , Transcription Factors/metabolism , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Cell Division , Chromans/therapeutic use , Humans , Ligands , Male , Middle Aged , Mutation , Prostatic Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Thiazoles/therapeutic use , Transcription Factors/genetics , Troglitazone , Tumor Cells, Cultured
3.
Science ; 289(5483): 1357-60, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10958784

ABSTRACT

Chromosomal translocations that encode fusion oncoproteins have been observed consistently in leukemias/lymphomas and sarcomas but not in carcinomas, the most common human cancers. Here, we report that t(2;3)(q13;p25), a translocation identified in a subset of human thyroid follicular carcinomas, results in fusion of the DNA binding domains of the thyroid transcription factor PAX8 to domains A to F of the peroxisome proliferator-activated receptor (PPAR) gamma1. PAX8-PPARgamma1 mRNA and protein were detected in 5 of 8 thyroid follicular carcinomas but not in 20 follicular adenomas, 10 papillary carcinomas, or 10 multinodular hyperplasias. PAX8-PPARgamma1 inhibited thiazolidinedione-induced transactivation by PPARgamma1 in a dominant negative manner. The experiments demonstrate an oncogenic role for PPARgamma and suggest that PAX8-PPARgamma1 may be useful in the diagnosis and treatment of thyroid carcinoma.


Subject(s)
Adenocarcinoma, Follicular/genetics , DNA-Binding Proteins/physiology , Nuclear Proteins , Oncogene Proteins, Fusion/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Thiazolidinediones , Thyroid Neoplasms/genetics , Trans-Activators/physiology , Transcription Factors/physiology , Adenocarcinoma, Follicular/metabolism , Adenoma/genetics , Adenoma/metabolism , Adult , Aged , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Cell Line , Cell Nucleus/metabolism , Child , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/pharmacology , Humans , Middle Aged , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , PAX8 Transcription Factor , Paired Box Transcription Factors , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Response Elements , Thiazoles/pharmacology , Thyroid Neoplasms/metabolism , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/pharmacology , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/pharmacology , Transcription, Genetic , Transcriptional Activation , Translocation, Genetic
4.
J Med Genet ; 37(6): 410-4, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10851250

ABSTRACT

PPARgamma, the gamma isoform of a family of peroxisome proliferator activated receptors, plays a key role in adipocyte differentiation. Recently, its broad expression in multiple tissues and several epithelial cancers has been shown. Further, somatic loss of function mutations in PPARgamma have been found in primary colorectal carcinomas. We sought to determine if somatic high penetrance mutations in this gene might also play a role in glioblastoma multiforme (GBM). We also examined this gene to determine if common low penetrance polymorphic alleles might lend low level susceptibility to GBM in the general population. No somatic high penetrance mutations were detected in 96 sporadic GBMs. However, polymorphic alleles at codons 12 and 449 were significantly over-represented among the 27 unrelated American patients with sporadic GBM compared to 80 race matched controls. While nine (33%) were heterozygous for the P12A variant, c.34C/G (cytosine to guanine change at nucleotide 34), 12 (15%) controls were heterozygous for P12A (p<0.05). Similarly, 13 of 26 (50%) glioblastoma patients compared to 10 of 80 (12%) normal controls were found to have the heterozygous H449H polymorphism (p<0.001). The over-representation of H449H in glioblastoma patients was confirmed with a second validation set of American patients. When both American series were combined, polymorphic H449H was over-represented among cases versus controls (p<0.001) and there was a similar trend (p=0.07) for P12A. The precise mechanism for this association is unknown but these PPARgamma polymorphisms may be acting in a low penetrance predisposing manner. However, these associations were not found in a German population, possibly arguing that if these variants are in linkage disequilibrium with a third locus, then this effect is relatively new, after the settlement of the American colonies.


Subject(s)
Brain Neoplasms/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Glioblastoma/genetics , Penetrance , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Alleles , Chi-Square Distribution , Codon/genetics , DNA Mutational Analysis , Gene Frequency/genetics , Germ-Line Mutation/genetics , Germany , Heterozygote , Humans , Linkage Disequilibrium/genetics , Matched-Pair Analysis , Odds Ratio , Polymorphism, Genetic/genetics , Protein Isoforms/genetics , United States
5.
J Biol Chem ; 275(24): 18527-33, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10748014

ABSTRACT

The nuclear hormone receptor peroxisome proliferator-activated receptor (PPAR) gamma is a ligand-activated transcription factor that regulates several crucial biological processes such as adipogenesis, glucose homeostasis, and cell growth. It is also the functional receptor for a new class of insulin-sensitizing drugs, the thiazolidinediones, now widely used in the treatment of type 2 diabetes mellitus. Here we report that PPARgamma protein levels are significantly reduced in adipose cells and fibroblasts in response to specific ligands such as thiazolidinediones. Studies with several doses of different ligands illustrate that degradation of PPARgamma correlates well with the ability of ligands to activate this receptor. However, analyses of PPARgamma mutants show that, although degradation does not strictly depend on the transcriptional activity of the receptor, it is dependent upon the ligand-gated activation function 2 (AF2) domain. Proteasome inhibitors inhibited the down-regulation of PPARgamma and ligand activation enhanced the ubiquitination of this receptor. These data indicate that, although ligand binding and activation of the AF2 domain increase the transcriptional function of PPARgamma, these same processes also induce ubiquitination and subsequent degradation of this receptor by the proteasome.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adipocytes/metabolism , Animals , Cysteine Endopeptidases/metabolism , Electrophoresis, Polyacrylamide Gel , Ligands , Mice , Multienzyme Complexes/metabolism , Proteasome Endopeptidase Complex , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Transcriptional Activation , Transfection
6.
Mol Cell ; 4(4): 611-7, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10549292

ABSTRACT

The process of adipogenesis is known to involve the interplay of several transcription factors. Activation of one of these factors, the nuclear hormone receptor PPAR gamma, is known to promote fat cell differentiation in vitro. Whether PPAR gamma is required for this process in vivo has remained an open question because a viable loss-of-function model for PPAR gamma has been lacking. We demonstrate here that mice chimeric for wild-type and PPAR gamma null cells show little or no contribution of null cells to adipose tissue, whereas most other organs examined do not require PPAR gamma for proper development. In vitro, the differentiation of ES cells into fat is shown to be dependent on PPAR gamma gene dosage. These data provide direct evidence that PPAR gamma is essential for the formation of fat.


Subject(s)
Adipose Tissue/growth & development , Cell Differentiation/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Animals , Chimera , Female , Gene Dosage , Gene Targeting/methods , Glucose/metabolism , In Situ Hybridization , Male , Mice , Mice, Knockout , RNA, Antisense , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sebaceous Glands/cytology , Skin/cytology , Stem Cells/metabolism , Transcription Factors/metabolism
7.
Mol Cell ; 3(6): 799-804, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10394368

ABSTRACT

The gamma isoform of the peroxisome proliferator-activated receptor, PPAR gamma, regulates adipocyte differentiation and has recently been shown to be expressed in neoplasia of the colon and other tissues. We have found four somatic PPAR gamma mutations among 55 sporadic colon cancers: one nonsense, one frameshift, and two missense mutations. Each greatly impaired the function of the protein. c.472delA results in deletion of the entire ligand binding domain. Q286P and K319X retain a total or partial ligand binding domain but lose the ability to activate transcription through a failure to bind to ligands. R288H showed a normal response to synthetic ligands but greatly decreased transcription and binding when exposed to natural ligands. These data indicate that colon cancer in humans is associated with loss-of-function mutations in PPAR gamma.


Subject(s)
Colorectal Neoplasms/genetics , DNA-Binding Proteins/metabolism , Linoleic Acids, Conjugated , Mutation , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazolidinediones , Transcription Factors/metabolism , Amino Acid Substitution , Binding Sites , Chromans/metabolism , Chromans/pharmacology , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/genetics , Dimerization , Exons/genetics , Genes, Tumor Suppressor/genetics , Genes, Tumor Suppressor/physiology , Humans , Hydroxyeicosatetraenoic Acids/metabolism , Ligands , Linoleic Acids/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Prostaglandin D2/pharmacology , Protein Binding , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Retinoic Acid/metabolism , Response Elements/genetics , Retinoid X Receptors , Rosiglitazone , Thiazoles/metabolism , Thiazoles/pharmacology , Transcription Factors/genetics , Transcriptional Activation/drug effects , Troglitazone
8.
Proc Natl Acad Sci U S A ; 96(7): 3951-6, 1999 Mar 30.
Article in English | MEDLINE | ID: mdl-10097144

ABSTRACT

Agonist ligands for the nuclear receptor peroxisome proliferator-activated receptor-gamma have been shown to induce terminal differentiation of normal preadipocytes and human liposarcoma cells in vitro. Because the differentiation status of liposarcoma is predictive of clinical outcomes, modulation of the differentiation status of a tumor may favorably impact clinical behavior. We have conducted a clinical trial for treatment of patients with advanced liposarcoma by using the peroxisome proliferator-activated receptor-gamma ligand troglitazone, in which extensive correlative laboratory studies of tumor differentiation were performed. We report here the results of three patients with intermediate to high-grade liposarcomas in whom troglitazone administration induced histologic and biochemical differentiation in vivo. Biopsies of tumors from each of these patients while on troglitazone demonstrated histologic evidence of extensive lipid accumulation by tumor cells and substantial increases in NMR-detectable tumor triglycerides compared with pretreatment biopsies. In addition, expression of several mRNA transcripts characteristic of differentiation in the adipocyte lineage was induced. There was also a marked reduction in immunohistochemical expression of Ki-67, a marker of cell proliferation. Together, these data indicate that terminal adipocytic differentiation was induced in these malignant tumors by troglitazone. These results indicate that lineage-appropriate differentiation can be induced pharmacologically in a human solid tumor.


Subject(s)
Chromans/therapeutic use , Liposarcoma/drug therapy , Thiazoles/therapeutic use , Thiazolidinediones , Adult , Biopsy , Cell Differentiation , Chromans/adverse effects , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Liposarcoma/metabolism , Liposarcoma/pathology , Male , Middle Aged , Phosphatidylcholines/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazoles/adverse effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Triglycerides/metabolism , Troglitazone
9.
Mol Cell Biol ; 19(3): 2330-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022919

ABSTRACT

The Src homology 3 (SH3) motif is found in numerous signal transduction proteins involved in cellular growth and differentiation. We have purified and cloned a novel protein, DEF-1 (differentiation-enhancing factor), from bovine brain by using a Src SH3 affinity column. Ectopic expression of DEF-1 in fibroblasts resulted in the differentiation of a significant fraction of the culture into adipocytes. This phenotype appears to be related to the induction of the transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma), since DEF-1 NIH 3T3 cells demonstrated augmented levels of PPARgamma mRNA and, when treated with activating PPARgamma ligands, efficient induction of differentiation. Further evidence for a role for DEF-1 in adipogenesis was provided by heightened expression of DEF-1 mRNA in adipose tissue isolated from obese and diabetes mice compared to that in tissue isolated from wild-type mice. However, DEF-1 mRNA was detected in multiple tissues, suggesting that the signal transduction pathway(s) in which DEF-1 is involved is not limited to adipogenesis. These results suggest that DEF-1 is an important component of a signal transduction process that is involved in the differentiation of fibroblasts and possibly of other types of cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Adipocytes/cytology , Carrier Proteins/physiology , Cell Differentiation , src Homology Domains , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/genetics , Cattle , Cloning, Molecular , DNA, Complementary , Disease Models, Animal , Fibroblasts/cytology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Obesity , Rabbits , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Transcription Factors/metabolism
10.
Proc Natl Acad Sci U S A ; 95(21): 12602-7, 1998 Oct 13.
Article in English | MEDLINE | ID: mdl-9770532

ABSTRACT

The rapid loss of muscle mass that accompanies many disease states, such as cancer or sepsis, is primarily a result of increased protein breakdown in muscle, and several observations have suggested an activation of the ubiquitin-proteasome system. Accordingly, in extracts of atrophying muscles from tumor-bearing or septic rats, rates of 125I-ubiquitin conjugation to endogenous proteins were found to be higher than in control extracts. On the other hand, in extracts of muscles from hypothyroid rats, where overall proteolysis is reduced below normal, the conjugation of 125I-ubiquitin to soluble proteins decreased by 50%, and treatment with triiodothyronine (T3) restored ubiquitination to control levels. Surprisingly, the N-end rule pathway, which selectively degrades proteins with basic or large hydrophobic N-terminal residues, was found to be responsible for most of these changes in ubiquitin conjugation. Competitive inhibitors of this pathway that specifically block the ubiquitin ligase, E3alpha, suppressed most of the increased ubiquitin conjugation in the muscle extracts from tumor-bearing and septic rats. These inhibitors also suppressed ubiquitination in normal extracts toward levels in hypothyroid extracts, which showed little E3alpha-dependent ubiquitination. Thus, the inhibitors eliminated most of the differences in ubiquitination under these different pathological conditions. Moreover, 125I-lysozyme, a model N-end rule substrate, was ubiquitinated more rapidly in extracts from tumor-bearing and septic rats, and more slowly in those from hypothyroid rats, than in controls. Thus, the rate of ubiquitin conjugation increases in atrophying muscles, and these hormone- and cytokine-dependent responses are in large part due to activation of the N-end rule pathway.


Subject(s)
Muscles/metabolism , Ubiquitins/metabolism , Animals , Atrophy , Hydrolysis , Male , Muscle Proteins/metabolism , Muscles/pathology , Rats
11.
Nat Med ; 4(9): 1046-52, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9734398

ABSTRACT

PPARgamma is a nuclear receptor that has a dominant regulatory role in differentiation of cells of the adipose lineage, and has recently been shown to be expressed in the colon. We show here that PPARgamma is expressed at high levels in both well- and poorly-differentiated adenocarcinomas, in normal colonic mucosa and in human colon cancer cell lines. Ligand activation of this receptor in colon cancer cells causes a considerable reduction in linear and clonogenic growth, increased expression of carcinoembryonic antigen and the reversal of many gene expression events specifically associated with colon cancer. Transplantable tumors derived from human colon cancer cells show a significant reduction of growth when mice are treated with troglitazone, a PPARgamma ligand. These results indicate that the growth and differentiation of colon cancer cells can be modulated through PPARgamma.


Subject(s)
Adenocarcinoma/physiopathology , Colonic Neoplasms/physiopathology , Receptors, Cytoplasmic and Nuclear/physiology , Thiazolidinediones , Transcription Factors/physiology , Animals , Cell Differentiation , Cell Division , Chromans/pharmacology , Gene Expression , Humans , Ligands , Mice , Mice, Nude , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Thiazoles/pharmacology , Transcription Factors/agonists , Transcription Factors/genetics , Troglitazone , Tumor Cells, Cultured
12.
Atherosclerosis ; 140(1): 121-34, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9733223

ABSTRACT

In order to examine the association between arterial fluid dynamics and the distribution of subendothelial macrophages in the normal rabbit aorta, steady and pulsatile particle flow visualization was performed in a geometrically realistic model of the rabbit aorto-celiac junction region. Over a range of aorto-celiac steady flow ratios, particle pathlines along the upstream lateral aortic walls curved to enter the celiac orifice, while two asymmetric regions of reversing spiral secondary flow originated along the downstream lateral portions of the orifice flow divider. These regions increased in size as either the Reynolds number or flow into the celiac artery increased. In pulsatile flow studies, particles along the lateral aortic walls near the celiac orifice began to spiral into the branch during peak systole. During systolic deceleration, the size of this spiral flow region increased as particles reversed direction to enter the celiac orifice. This contrasted with flow patterns directly upstream and downstream of the orifice, which remained unidirectional throughout this period even along the distal lip of the orifice. The highest frequency of subendothelial white blood cells in the normal rabbit aorta was associated with regions where secondary flow patterns occurred, and where the orientation of endothelial cell nuclei deviated from the major direction of aortic flow. Secondary flow patterns may aid the accumulation of monocytes and macrophages about the lateral regions of the celiac artery flow divider by transporting monocytes to the walls, allowing them time to attach to the endothelial cells, or by stimulating the endothelial cells to express leukocyte adhesion molecules. These same regions are associated with increased endothelial permeability to low density lipoprotein and, under hypercholesterolemic conditions, lesion origination.


Subject(s)
Aorta/physiology , Celiac Artery/physiology , Hemorheology , Macrophages/physiology , Models, Cardiovascular , Animals , Endothelium, Vascular/physiology , Rabbits , Stress, Mechanical
13.
Mol Cell ; 1(3): 465-70, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9660931

ABSTRACT

We have previously demonstrated that PPAR gamma stimulates the terminal differentiation of adipocyte precursors when activated by synthetic ligands, such as the antidiabetic thiazolidinedione (TZD) drugs. We show here that PPAR gamma is expressed at significant levels in human primary and metastatic breast adenocarcinomas. Ligand activation of this receptor in cultured breast cancer cells causes extensive lipid accumulation, changes in breast epithelial gene expression associated with a more differentiated, less malignant state, and a reduction in growth rate and clonogenic capacity of the cells. Inhibition of MAP kinase, shown previously to be a powerful negative regulator of PPAR gamma, improves the TZD ligand sensitivity of nonresponsive cells. These data suggest that the PPAR gamma transcriptional pathway can induce terminal differentiation of malignant breast epithelial cells and thus may provide a novel, nontoxic therapy for human breast cancer.


Subject(s)
Breast Neoplasms , Gene Expression Regulation, Neoplastic/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation/physiology , Cell Division/physiology , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Humans , Lipid Metabolism , Mitogen-Activated Protein Kinase Kinases , Protein Kinase Inhibitors , RNA, Messenger/metabolism , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/enzymology
14.
J Clin Invest ; 101(1): 1-9, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9421459

ABSTRACT

The ability to regulate specific genes of energy metabolism in response to fasting and feeding is an important adaptation allowing survival of intermittent food supplies. However, little is known about transcription factors involved in such responses in higher organisms. We show here that gene expression in adipose tissue for adipocyte determination differentiation dependent factor (ADD) 1/sterol regulatory element binding protein (SREBP) 1, a basic-helix-loop-helix protein that has a dual DNA-binding specificity, is reduced dramatically upon fasting and elevated upon refeeding; this parallels closely the regulation of two adipose cell genes that are crucial in energy homeostasis, fatty acid synthetase (FAS) and leptin. This elevation of ADD1/SREBP1, leptin, and FAS that is induced by feeding in vivo is mimicked by exposure of cultured adipocytes to insulin, the classic hormone of the fed state. We also show that the promoters for both leptin and FAS are transactivated by ADD1/SREBP1. A mutation in the basic domain of ADD1/SREBP1 that allows E-box binding but destroys sterol regulatory element-1 binding prevents leptin gene transactivation but has no effect on the increase in FAS promoter function. Molecular dissection of the FAS promoter shows that most if not all of this action of ADD1/SREBP1 is through an E-box motif at -64 to -59, contained with a sequence identified previously as the major insulin response element of this gene. These results indicate that ADD1/SREBP1 is a key transcription factor linking changes in nutritional status and insulin levels to the expression of certain genes that regulate systemic energy metabolism.


Subject(s)
CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/metabolism , Fatty Acid Synthases/genetics , Helix-Loop-Helix Motifs , Nuclear Proteins/metabolism , Proteins/genetics , Transcription Factors , 3T3 Cells , Animals , Cell Line , DNA-Binding Proteins/genetics , Eating , Fatty Acid Synthases/metabolism , Female , Gene Expression Regulation/drug effects , Insulin/pharmacology , Leptin , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Proteins/metabolism , Rats , Sterol Regulatory Element Binding Protein 1 , Transcriptional Activation
15.
J Biol Chem ; 272(8): 5283-90, 1997 Feb 21.
Article in English | MEDLINE | ID: mdl-9030601

ABSTRACT

The ob gene product, leptin, is a major hormonal regulator of appetite and fat cell mass. Recent work has suggested that the antidiabetic agents, the thiazolidinediones (TZ), which are also high affinity ligands of peroxisome proliferator-activated receptor-gamma (PPARgamma), inhibit leptin expression in rodents. To examine the effects of this class of drug on the leptin gene in adipocytes we performed Northern analysis on primary rat adipocytes cultured in the presence or absence of TZ. TZ reduced leptin mRNA levels by 75%. To determine whether this effect was mediated at the transcriptional level, we isolated 6510 base pairs of 5'-flanking sequence of the leptin promoter and studied reporter constructs in primary rat adipocytes and CV-1 cells. Sequence analysis demonstrated the presence of a consensus direct repeat with a 1-base-pair gap site between -3951 and -3939 as well as a consensus CCAAT/enhancer binding protein (C/EBP) site between -55 and -47. Our functional analysis in transfected primary rat adipocytes demonstrates that, despite the presence of a canonical direct repeat with a 1-base-pair gap site, TZ alone decreases reporter gene expression of leptin promoter constructs ranging from -6510 to +9 to -65 to +9. In CV-1 cells, which contain endogenous PPARgamma, TZ treatment alone had little effect on these constructs. However, TZ treatment did inhibit C/EBPalpha-mediated transactivation of the leptin promoter. This down-regulation of leptin reporter constructs mapped to a -65 to +9 promoter fragment which binds C/EBPalpha in gel-mobility shift assays but does not bind PPARgamma2 alone or as a heterodimer with 9-cis-retinoic acid receptor. Conversely, the promoter (-5400 to +24 base pairs) of the aP2 gene, another adipocyte-specific gene, was induced 7.3-fold by TZ. Co-transfection with C/EBPalpha minimally stimulated the aP2 promoter from basal levels but notably blocked activation by TZ. These data indicate that PPARgamma and C/EBPalpha can functionally antagonize each other on at least two separate promoters and that this mechanism may explain the down-regulation of leptin expression by thiazolidinediones.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Proteins/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins , Cells, Cultured , DNA-Binding Proteins/genetics , Leptin , Male , Molecular Sequence Data , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics
16.
Proc Natl Acad Sci U S A ; 94(1): 237-41, 1997 Jan 07.
Article in English | MEDLINE | ID: mdl-8990192

ABSTRACT

Induction of terminal differentiation represents a promising therapeutic approach to certain human malignancies. The peroxisome proliferator-activated receptor gamma (PPAR gamma) and the retinoid X receptor alpha (RXR alpha) form a heterodimeric complex that functions as a central regulator of adipocyte differentiation. Natural and synthetic ligands for both receptors have been identified. We demonstrate here that PPAR gamma is expressed at high levels in each of the major histologic types of human liposarcoma. Moreover, primary human liposarcoma cells can be induced to undergo terminal differentiation by treatment with the PPAR gamma ligand pioglitazone, suggesting that the differentiation block in these cells can be overcome by maximal activation of the PPAR pathway. We further demonstrate that RXR-specific ligands are also potent adipogenic agents in cells expressing the PPAR gamma/RXR alpha heterodimer, and that simultaneous treatment of liposarcoma cells with both PPAR gamma- and RXR-specific ligands results in an additive stimulation of differentiation. Liposarcoma cell differentiation is characterized by accumulation of intracellular lipid, induction of adipocyte-specific genes, and withdrawal from the cell cycle. These results suggest that PPAR gamma ligands such as thiazolidinediones and RXR-specific retinoids may be useful therapeutic agents for the treatment of liposarcoma.


Subject(s)
Liposarcoma/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/metabolism , Retinoids/pharmacology , Thiazoles/pharmacology , Thiazolidinediones , Transcription Factors/metabolism , Adipocytes/metabolism , Antigens, Differentiation/analysis , Cell Differentiation , Fibroblasts/drug effects , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans , Ligands , Lipid Metabolism , Liposarcoma/classification , Liposarcoma/pathology , Pioglitazone , Retinoid X Receptors , Tumor Cells, Cultured
17.
J Exp Med ; 185(1): 171-5, 1997 Jan 06.
Article in English | MEDLINE | ID: mdl-8996253

ABSTRACT

Several inflammatory cytokines, most notably tumor necrosis factor (TNF) and IL-1, induce anorexia and loss of lean body mass, common manifestations of acute and chronic inflammatory conditions. In C57BL/6 female mice, the administration of TNF, IL-1, and, to a lesser extent, leukemia inhibitory factor (LIF), produced a prompt and dose-dependent increase in serum leptin levels and leptin mRNA expression in fat. IL-10, IL-4, ciliary neurotrophic factor, and IL-2, cytokines not known to induce anorexia or decrease food intake, had no effect on leptin gene expression or serum leptin levels. After administration of Escherichia coli lipopolysaccharide (LPS), leptin gene expression and leptin levels were increased. These findings suggest that leptin levels may be one mechanism by which anorexia is induced during acute inflammatory conditions.


Subject(s)
Adipose Tissue/metabolism , Anorexia , Cytokines/pharmacology , Inflammation , Interleukin-6 , Protein Biosynthesis , Transcription, Genetic/drug effects , Adipose Tissue/drug effects , Adipose Tissue/immunology , Animals , Ciliary Neurotrophic Factor , Escherichia coli , Female , Growth Inhibitors/pharmacology , Humans , Interleukin-10/pharmacology , Interleukin-2/pharmacology , Interleukin-4/pharmacology , Interleukins/pharmacology , Kinetics , Leptin , Leukemia Inhibitory Factor , Lipopolysaccharides/pharmacology , Lymphokines/pharmacology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/pharmacology , Proteins/analysis , RNA, Messenger/biosynthesis , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
18.
Science ; 274(5295): 2100-3, 1996 Dec 20.
Article in English | MEDLINE | ID: mdl-8953045

ABSTRACT

Adipocyte differentiation is an important component of obesity and other metabolic diseases. This process is strongly inhibited by many mitogens and oncogenes. Several growth factors that inhibit fat cell differentiation caused mitogen-activated protein (MAP) kinase-mediated phosphorylation of the dominant adipogenic transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma) and reduction of its transcriptional activity. Expression of PPARgamma with a nonphosphorylatable mutation at this site (serine-112) yielded cells with increased sensitivity to ligand-induced adipogenesis and resistance to inhibition of differentiation by mitogens. These results indicate that covalent modification of PPARgamma by serum and growth factors is a major regulator of the balance between cell growth and differentiation in the adipose cell lineage.


Subject(s)
Adipocytes/cytology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adipocytes/metabolism , Animals , Blood , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Differentiation , Cell Line , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Flavonoids/pharmacology , Insulin/pharmacology , Ligands , Mice , Mitogens/pharmacology , Mutation , Phosphorylation , Rats , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transfection
19.
Am J Surg ; 171(6): 576-80, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8678203

ABSTRACT

BACKGROUND: The authors examined the efficacy of human growth hormone (HGH) in patients dependent on mechanical ventilation who were being weaned from the respirator. METHODS: A total of 53 patients were chosen by the primary surgical team in consultation with the critical care service to undergo HGH therapy. These patients had been receiving standard ICU support and had failed standard ventilator weaning protocols. As such, they were treated with HGH in an attempt to increase respiratory muscle strength and facilitate weaning from mechanical ventilation. RESULTS: General demographic information was recorded. Patients suffered from a high incidence of co-morbid conditions and infectious complications. The average duration of HGH therapy was 38 days, and 81% of the previously unweanable patients were eventually weaned from mechanical ventilation with an overall survival of 76%. Mortality as predicted by APACHE II Scores was significantly less than actual mortality (24% actual mortality vs. 42% predicted mortality, P < 0.05). CONCLUSIONS: This phase I study presents clinical evidence supporting the safety and efficacy of HGH in promoting respiratory independence in a selected group of surgical ICU patients. Randomized, blinded, controlled trials now seem warranted.


Subject(s)
Growth Hormone/therapeutic use , Postoperative Complications/drug therapy , Respiratory Insufficiency/drug therapy , Respiratory Insufficiency/etiology , Aged , Critical Illness , Humans , Middle Aged , Respiration, Artificial , Retrospective Studies , Treatment Outcome , Ventilator Weaning
20.
J Trauma ; 39(3): 526-30; discussion 530-2, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7473919

ABSTRACT

BACKGROUND: Advances in the management of patients with major thermal injury have resulted in a progressive increase in survival rates. We report preliminary data evaluating the safety and potential efficacy of human growth hormone (HGH) administration in a high-risk population of burned patients. METHODS: From 1989 to 1993, 69 patients sustaining major burns (defined as patient age plus percentage of body surface area with deep second- and third-degree burns > or = 90) were evaluated. Patients routinely received anti-inflammatory pharmacotherapy including antioxidants, an endotoxin binder, and cyclooxygenase blockade. Half of the 54 patients who survived more than 7 days received HGH to enhance wound healing. Injury severity, morbidity, and mortality for patients receiving HGH was compared to the 27 patients not receiving HGH. RESULTS: For the entire population (n = 69), average age was 56 +/- 23 years, body surface area burned was 58% +/- 24%, and 30% sustained smoke inhalation. Actual mortality was 41%, significantly less than the more than 70% mortality rate predicted from reported outcome data. Patients receiving HGH were well matched with the group not receiving HGH with respect to extent of injury, burn management, pharmacotherapy, and in-hospital morbidity. Mortality of the patients receiving HGH was 11%, significantly less than the 37% mortality rate of the patients without HGH (p = 0.027). CONCLUSION: Compared to standard predictors of burn mortality our small patient group appears to have an improved survival rate, suggesting that the use of anti-inflammatory agents appears safe and potentially beneficial. Patients receiving HGH exhibited minimal drug-related complications and mortality rates were improved when this population was compared with both predicted mortality rates and a well-matched control population of concurrently treated patients. Prospective blinded trials are now necessary to confirm these findings in a larger patient group.


Subject(s)
Burns/drug therapy , Burns/mortality , Growth Hormone/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Humans , Middle Aged , Survival Rate , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...