Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
J Clin Invest ; 92(1): 514-9, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8326016

ABSTRACT

Chronically culturing HIT-T15 cells in media containing high glucose concentrations leads to decreased insulin mRNA levels, insulin content, and insulin secretion. These changes can be prevented by culturing the cells in media containing lower glucose levels (Robertson, R. P., H.-J. Zhang, K. L. Pyzdrowski, and T. F. Walseth. 1992. J. Clin. Invest. 90:320-325). The mechanism of this seemingly paradoxical phenomenon was examined by transiently transfecting HIT cells with a chloramphenicol acetyl transferase (CAT) reporter gene controlled by the 5'-regulatory domain of the human insulin gene (INSCAT). Early passages of HIT cells readily expressed INSCAT, whereas late passages of cells chronically cultured in 11.1 mM glucose expressed only 28.7 +/- 2.3% (mean +/- SEM) of the CAT activity expressed in early passages. In contrast, late passages of HIT cells chronically cultured in 0.8 mM glucose retained the ability to express the INSCAT reporter gene to 69.6 +/- 10.0% of the CAT activity observed in early passages. The decrease in INSCAT expression in late passages of cells serially cultured in 11.1 mM glucose was associated with the inability to form a specific nuclear protein-DNA complex with the CT motifs of the human insulin promoter. Formation of this specific protein-DNA complex was preserved in late passages of HIT cells when serially cultured in 0.8 mM glucose. Mutations of the CT motifs caused markedly diminished CAT activity in all passages examined. These data indicate that chronic exposure of the beta cell to high glucose concentrations can paradoxically decrease insulin gene transcription, in part, by altering the ability of a regulatory protein (GSTF) to interact with the insulin gene promoter. This provides a potential mechanism for glucotoxic effects on the beta cell at the level of the insulin gene.


Subject(s)
Glucose/administration & dosage , Insulin/genetics , Islets of Langerhans/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , Cells, Cultured , Cricetinae , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , In Vitro Techniques , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Promoter Regions, Genetic , RNA, Messenger/genetics , Transcription, Genetic
2.
J Clin Invest ; 78(4): 1091-6, 1986 Oct.
Article in English | MEDLINE | ID: mdl-3760185

ABSTRACT

We have studied the effect of glucagon on the expression of a triiodothyronine (T3) and carbohydrate-inducible mRNA sequence (mRNA-S14) in rat liver that undergoes a threefold diurnal variation (peak, 2200 h; nadir, 0800 h). Glucagon injection into euthyroid rats (25 micrograms/100 g body wt i.p., three doses at 15-min intervals) during the nocturnal plateau of mRNA-S14 caused a monoexponential disappearance of this sequence (t1/2, 90 min) accompanied by a 90% reduction in the transcriptional rate in a nuclear run-off assay, indicative of a near total reduction of synthesis. This effect was markedly attenuated in rats treated with T3 (200 micrograms/100 g body wt i.p.) 24 h before glucagon injection. When T3 was given 15 min after glucagon, the glucagon-initiated decline in mRNA-S14 was reversed within 90 min, suggesting a rapid interaction between the two hormones in the evening. Curiously, administration of T3 alone at this hour did not affect a significant increase in mRNA-S14. At 0800 h, however, T3 caused the expected brisk induction of this sequence, whereas glucagon was without effect. In essence, glucagon affected mRNA-S14 synthesis only in the evening, while T3 increased levels of this sequence above the baseline only in the morning. T3, however, reversed the effect of prior glucagon injection at night. The observed alterations in hormonal responsivity could underly the diurnal variation of mRNA-S14 expression. Moreover, the data suggest the hypothesis that T3 may act on S14 gene expression by antagonizing factors that inhibit its transcription.


Subject(s)
Circadian Rhythm , Glucagon/pharmacology , Liver/metabolism , RNA, Messenger/metabolism , Triiodothyronine/pharmacology , Animals , Food Deprivation , Gene Expression Regulation/drug effects , Liver/drug effects , Male , Protein Biosynthesis/drug effects , Rats , Rats, Inbred Strains
3.
J Clin Invest ; 67(6): 1739-47, 1981 Jun.
Article in English | MEDLINE | ID: mdl-7240417

ABSTRACT

Previous studies from this laboratory have demonstrated an age-related decrease in hepatic malic enzyme (ME) levels and in the response of ME to triiodo-l-thyronine (T(3)). Moreover, we have recently shown a synergistic interaction of T(3) and a high carbohydrate diet in the induction of this enzyme. Studies were therefore undertaken to assess the response of aging rats to a high carbohydrate diet and to test the effect of such dietary manipulations on the responsiveness of ME to T(3). For this purpose, a new radio-immunoassay for ME was developed that, because of a 10-fold higher sensitivity, was particularly suited to the measurement of the low concentrations of hepatic enzyme in older animals. The level of ME per milligram of DNA fell approximately 70% between 1 and 6 mo with only minor further changes demonstrated between 6 and 18 mo. In contrast, the level of ME per milligram DNA in brain was slightly increased in the older animals. Although the absolute increment of hepatic ME resulting from seven daily injections of T(3) (15 mug/100 g body wt) fell with age, the ratio of the ME content per milligram DNA to that observed in control animals maintained on a regular chow diet remained relatively constant with an average value of 11.1. The responsivity of hepatic ME to a high carbohydrate, fat-free diet also decreased with age and could not be attributed exclusively to a reduction in food consumption. The age-related reduction in ME responsivity to dietary stimuli appeared to be due to a reduction in the formation of the specific messenger, (m)RNA for ME as determined in an in vitro translational assay. Our data are consistent with the following hypothesis. There is an age-related decreased hepatic responsivity to a high carbohydrate dietary stimulus. Thyroid hormone administration, as previously postulated by us, interacts with a product or an intermediate of carbohydrate metabolism in a multiplicative fashion. As a consequence, the absolute increment of ME induced by T(3) administration also declines with age.


Subject(s)
Dietary Carbohydrates/administration & dosage , Liver/enzymology , Malate Dehydrogenase/metabolism , Triiodothyronine/pharmacology , Aging , Animals , Brain/enzymology , Diet , Dietary Fats , Enzyme Activation/drug effects , Malate Dehydrogenase/analysis , Male , Radioimmunoassay/methods , Rats , Triiodothyronine/metabolism
4.
J Clin Invest ; 65(5): 1126-34, 1980 May.
Article in English | MEDLINE | ID: mdl-7364940

ABSTRACT

We have investigated the relationship between the administration of triiodothyronine (T3) and a high carbohydrate (CHO) fat-free diet in the induction of lipogenic enzymes in two rat tissues, liver, and fat. Male thyroidectomized rats were treated with graded daily doses of T3 and either supplemented with a high CHO diet or left on a regular diet. Enzymes studied included malic enzyme (ME), fatty acid synthetase, glucose-6-phosphate dehydrogenase, and 6-phosphogluconate dehydrogenase. In the liver, all four lipogenic enzymes showed a synergistic response between T3 administration and high CHO feeding. In fat, ME also responded synergistically. The interaction was reflected in an increased sensitivity to T3. The dose of T3 required to achieve 50% maximal response was reduced three- to seven-fold by the high CHO diet. This phenomenon could not be attributed to a dietary-induced alteration either in T3 metabolism or in number or affinity of the T3-nuclear receptors. Moreover, studies of the relative rate of synthesis of ME suggested a simultaneous time of onset in the induction of ME, within 2 h after the application of either T3 or CHO. Thus, it is unlikely that either stimulus is secondary to the other. Since parallel experiments from this laboratory (Towle, Mariash, and Oppenheimer,1980. Changes in hepatic levels of messenger ribonucleic acid for malic enzyme during induction by thyroid hormone or diet. Biochemistry. 19: 579-585.) show that ME induction both by CHO and T3 is mediated by an increase in specific messenger RNA for ME, the interaction of T3 and the dietary factor occurs at a pretanslational level.


Subject(s)
Dietary Carbohydrates/pharmacology , Lipids/biosynthesis , Liver/enzymology , Triiodothyronine/pharmacology , Adipose Tissue/enzymology , Animals , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Induction/drug effects , Fatty Acid Synthases/biosynthesis , Glucosephosphate Dehydrogenase/biosynthesis , Liver/drug effects , Malate Dehydrogenase/biosynthesis , Male , Phosphogluconate Dehydrogenase/biosynthesis , Rats , Time Factors
5.
J Clin Invest ; 68(6): 1485-90, 1981 Dec.
Article in English | MEDLINE | ID: mdl-6274910

ABSTRACT

We have stimulated in a cultured hepatocyte system the synergistic interaction between triiodothyronine (T3) and dietary carbohydrate in the induction of malic enzyme (ME). Kinetic studies revealed that isolated hepatocytes equilibrate with media T3 within 5 min; nuclei equilibrate with media T3 by 45 min; and the half-time of T3 metabolism was 10 h in 10% serum. We demonstrated nuclear T3 receptors in isolated hepatocytes and the induction of ME by T3 in physiological concentrations. However, in the complete absence of T3 glucose could still induce ME. At all concentrations of glucose (100-1,000 mg/dl), T3 (0.3 nM free T3) resulted in a relatively constant (1.4- to 1.7-fold) increase in ME response. The augmentation in ME activity was paralleled by an enhanced rate of enzyme synthesis as determined by [3H]leucine incorporation into immunoprecipitable ME. Cells cultured in serum free media also demonstrated a glucose-dependent increase in ME. Insulin greatly stimulated the glucose induction of ME, whereas dexamethasone had very little influence on ME induction. These studies demonstrate the usefulness of an adult hepatocyte tissue culture model for the study of the effects of T3 on gene expression in cells that are not derived from tumor. They clearly demonstrate that well established effects of T3 can be simulated in such a system at levels of free hormone that approximate those in extracellular body fluids. Our results indicate that an increased concentration of glucose per se can induce the formation of ME in the absence of alterations in extrahepatic hormones or factors. Moreover, our findings confirm inferences from in vivo studies that T3 acts as a multiplier of a glucose-induced signal.


Subject(s)
Glucose/pharmacology , Liver/enzymology , Malate Dehydrogenase/biosynthesis , Triiodothyronine/pharmacology , Animals , Cell Nucleus/metabolism , Cells, Cultured , Enzyme Induction , Glucose/metabolism , Insulin/pharmacology , Kinetics , Liver/drug effects , Male , Rats , Rats, Inbred Strains , Receptors, Cell Surface/metabolism , Triiodothyronine/metabolism
6.
Mol Cell Biol ; 5(10): 2642-6, 1985 Oct.
Article in English | MEDLINE | ID: mdl-3837180

ABSTRACT

The regulation of a thyroid hormone-responsive gene in rats, designated spot 14, was explored. The expression of this gene in liver is rapidly (less than 10 min) and markedly (greater than 10-fold) altered by the administration of 3,5,3'-triiodo-L-thyronine (T3) to hypothyroid rats (P. Narayan, C. W. Liaw, and H. C. Towle, Proc. Natl. Acad. Sci. USA 81:4687-4691, 1984). To investigate the cellular site at which T3 acts to induce this hepatic mRNA, we made parallel measurements of the relative levels of spot 14 mRNA and nuclear precursor RNA and of the rate of gene transcription after treatments designed to alter the thyroid status of rats. The relative levels of both the mRNA and nuclear precursor were elevated roughly 5- to 6-fold in euthyroid animals and 9- to 12-fold in hyperthyroid animals over those in hypothyroid controls. However, only a small difference of approximately 1.5-fold was detected in the rate of spot 14 gene transcription. After a single injection of T3 into hypothyroid animals, a small and transient rise in the transcription rate was detected at 30 min. However, the levels of spot 14 mRNA and nuclear precursor RNA increased much more dramatically throughout the first 4 h of treatment. In both cases, changes in the rate of gene transcription were not capable of accounting for the alterations observed in mRNA levels. Thus, the major site of spot 14 gene regulation by T3 is at a posttranscriptional level. The proportional changes observed in the nuclear precursor and mRNA levels suggest that the site of control is at the level of stability of the nuclear precursor RNA for spot 14 mRNA.


Subject(s)
Liver/physiology , RNA, Messenger/genetics , Triiodothyronine/physiology , Animals , Cell Nucleus/physiology , Gene Expression Regulation , Isoelectric Point , Male , Molecular Weight , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Rats , Time Factors , Transcription, Genetic
7.
Mol Cell Biol ; 15(3): 1499-512, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7862143

ABSTRACT

The thyroid hormone receptors (TR) are members of the nuclear receptor family of ligand-mediated transcription factors. The large region of TR that lies C-terminal to its DNA-binding domain subserves functions of ligand binding, dimerization, and transactivation. Little is known regarding the structural or functional determinants of these processes. We have utilized genetic screening in the yeast Saccharomyces cerevisiae to identify residues involved in these functions. Random mutations of the rat TR beta 1 isoform between amino acid residues 179 and 456 were screened, and mutants with reduced hormone-dependent activation of reporter gene activity were isolated. In this paper we describe the characterization of a class of mutants that exhibit a dissociation between hormone binding and transcriptional activation. These mutants retained hormone binding (> 15% of the wild-type level) yet failed to transactivate a reporter gene. A number of these mutations occurred within the D region, which links the DNA-binding and ligand-binding domains of the receptor. One subset of these mutations abrogated DNA binding, supporting a role of the D region in this process. The remainder retain DNA binding and thus highlight residues critical for receptor activation. In addition, an unexpected group of "superactivator" mutations that led to enhanced hormone-dependent activation in S. cerevisiae were found. These mutations localized to the carboxy-terminal portion of the receptor in a region which contains elements conserved across the superfamily of nuclear receptors. The hormone-dependent phenotype of these superactivator mutations suggests an important role of this segment in ligand-mediated transcriptional activation.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Triiodothyronine/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Chlorocebus aethiops , Cloning, Molecular , DNA-Binding Proteins/isolation & purification , Macromolecular Substances , Molecular Sequence Data , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Rats , Receptors, Thyroid Hormone/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Transcription, Genetic , Transcriptional Activation , Transfection , beta-Galactosidase/metabolism
8.
Mol Cell Biol ; 7(4): 1352-7, 1987 Apr.
Article in English | MEDLINE | ID: mdl-3648478

ABSTRACT

The regulation of a gene, designated spot 14, which is rapidly induced in rat liver in response to 3,5,3'-triiodo-L-thyronine (T3) was studied as a model for exploring the molecular basis of thyroid hormone action. The time course of induction of the nuclear precursor to spot 14 mRNA after intramuscular injection of T3 displayed a very short lag period of between 10 and 20 min. The rapidity of this effect suggests that the induction in gene expression occurs as a primary response to the hormone-receptor interaction. The protein synthesis inhibitor cycloheximide injected 15 min before T3 completely blocked the accumulation of nuclear precursor RNA 30 min after T3 treatment. Emetine, an inhibitor of protein synthesis which acts by a different mechanism than cycloheximide, also blocked the induction of the spot 14 nuclear precursor RNA. The increased rate of spot 14 gene transcription observed after T3 treatment, as measured by nuclear run-on assay, was similarly completely abolished in the presence of cycloheximide. In addition, ongoing protein synthesis was required for maintaining spot 14 nuclear precursor RNA at induced levels in animals previously treated with T3. On the other hand, cycloheximide had no effect on T3 uptake or binding to the nuclear receptor during the 45-min time frame studied. The paradox of the rapid kinetics of induction and the requirement of ongoing protein synthesis may be explained by a protein with an extremely short half-life which is necessary for T3 induction of the spot 14 gene.


Subject(s)
Genes, Regulator/drug effects , Genes/drug effects , Liver/metabolism , Nucleic Acid Precursors/genetics , Protein Biosynthesis , RNA, Messenger/genetics , Triiodothyronine/pharmacology , Animals , Cell Nucleus/metabolism , Cycloheximide/pharmacology , Hypothyroidism/metabolism , Kinetics , Liver/drug effects , Male , Protein Biosynthesis/drug effects , RNA Precursors , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Rats , Rats, Inbred Strains , Receptors, Thyroid Hormone/metabolism , Transcription, Genetic/drug effects , Triiodothyronine/metabolism
9.
Mol Cell Biol ; 15(5): 2582-8, 1995 May.
Article in English | MEDLINE | ID: mdl-7739539

ABSTRACT

Adipocyte determination- and differentiation-dependent factor 1 (ADD1), a member of the basic helix-loop-helix (bHLH) family of transcription factors, has been associated with both adipocyte differentiation and cholesterol homeostasis (in which case it has been termed SREBP1). Using PCR-amplified binding analysis, we demonstrate that ADD1/SREBP1 has dual DNA sequence specificity, binding to both an E-box motif (ATCACGTGA) and a non-E-box sequence previously shown to be important in cholesterol metabolism, sterol regulatory element 1 (SRE-1; ATCACCCCAC). The ADD1/SREBP1 consensus E-box site is similar to a regulatory sequence designated the carbohydrate response element, defined by its ability to regulate transcription in response to carbohydrate in genes involved in fatty acid and triglyceride metabolism in liver and fat. When expressed in fibroblasts, ADD1/SREBP1 activates transcription through both the carbohydrate response E-box element and SRE-1. Substitution of an atypical tyrosine in the basic region of ADD1/SREBP1 to an arginine found in most bHLH protein causes a restriction to only E-box binding. Conversely, substitution of a tyrosine for the equivalent arginine in another bHLH protein, upstream stimulatory factor, allows this factor to acquire a dual binding specificity similar to that of ADD1/SREBP1. Promoter activation by ADD1/SREBP1 through the carbohydrate response element E box is not sensitive to the tyrosine-to-arginine mutation, while activation through SRE-1 is completely suppressed. These data illustrate that ADD1/SREBP1 has dual DNA sequence specificity controlled by a single amino acid residue; this dual specificity may provide a novel mechanism to coordinate different pathways of lipid metabolism.


Subject(s)
CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/metabolism , DNA/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adipose Tissue/cytology , Adipose Tissue/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Differentiation , Cholesterol/metabolism , DNA/genetics , DNA Primers/genetics , DNA-Binding Proteins/genetics , Helix-Loop-Helix Motifs/genetics , Homeostasis , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Sequence Homology, Amino Acid , Sterol Regulatory Element Binding Protein 1 , Transcription Factors/genetics , Transcriptional Activation , Tyrosine/metabolism
10.
Diabetes ; 43(4): 546-51, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7907998

ABSTRACT

We observed in the HIT cell, a clonal insulin-secreting cell line, that epinephrine and somatostatin lower insulin mRNA levels and intracellular insulin content in addition to the well-recognized effect of these hormones to inhibit insulin secretion. To determine whether these inhibitory hormones might regulate insulin synthesis at the level of insulin gene transcription, we studied HIT cell expression of a human insulin-chloramphenicol acetyl transferase (CAT) reporter gene in the presence of glucose, epinephrine, and somatostatin. HIT cell expression of this human insulin-CAT reporter gene was responsive to glucose in a concentration-dependent manner, increasing threefold as the glucose concentration increased from 0.4 to 11 mM. Epinephrine significantly inhibited insulin-CAT reporter gene expression (61 +/- 5% of control), an effect mediated specifically by the human insulin gene promoter/enhancer sequence. Somatostatin significantly inhibited expression of the human insulin-CAT reporter gene (65 +/- 4% of control) and, to a lesser extent, expression of a control reporter gene, pRSVCAT (78 +/- 4% of control). Thus, somatostatin may inhibit insulin gene transcription by insulin gene-specific effects as well as more general effects on gene expression. Both epinephrine and somatostatin inhibited expression of the human insulin-CAT reporter gene in a concentration-dependent manner that paralleled inhibition of insulin secretion. These studies indicate that epinephrine and somatostatin lower HIT cell insulin mRNA levels in part by inhibiting insulin gene transcription. Thus, hormonal inhibition of insulin secretion may be coupled with inhibition of insulin synthesis, thereby allowing the beta-cell to match insulin supply to secretory demand.


Subject(s)
Epinephrine/pharmacology , Gene Expression Regulation/drug effects , Glucose/pharmacology , Insulin/biosynthesis , Somatostatin/pharmacology , Transcription, Genetic/drug effects , Animals , Base Sequence , Cell Line , Chloramphenicol O-Acetyltransferase/biosynthesis , Cricetinae , Gene Expression/drug effects , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans , Kinetics , Mesocricetus , Molecular Sequence Data , Oligodeoxyribonucleotides , Transfection
11.
Mol Endocrinol ; 8(8): 1021-37, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7997231

ABSTRACT

Hepatic expression of the rat S14 gene is markedly and rapidly induced in response to T3. Previously, three contiguous restriction fragments of the S14 gene with thyroid hormone response activity were mapped to a region 2.5-3.0 kilobases upstream from the start of transcription [Far Upstream Regulatory region (FUR)]. To further investigate the molecular basis of the thyroid hormonal control of S14 gene expression, we have mapped the functional TRE sequences in the FUR region of the S14 gene. In vitro translated thyroid hormone receptor (TR) and retinoid X receptor were used in the gel retardation assays to map receptor binding sites in the S14 gene. Three TR-binding sequences were identified in the FUR region of the S14 gene and designated: FUR10 (from -2718 to -2694), FUR11 (from -2632 to -2595), and FUR12 (from -2582 to -2558). Each binding site contains two or more elements related to the consensus monomer binding motif 5'-Pu-GGTCA. In FUR10 and FUR12, these motifs were arranged as direct repeats with 4 base pair spacing, while in FUR11 a more complex arrangement occurred. From mutagenesis experiments, all three TR-binding sequences in the S14 gene were found to play a role and synergize with each other in the responsiveness to T3. The importance of this functional synergy is also shown by the observations that at least two TR-binding sites are required for T3 induction in hepatocytes. In addition, synergy occurs between TR and additional regulatory sequences present in the FUR region and provides the maximal T3 response of the S14 gene.


Subject(s)
Gene Expression Regulation/drug effects , Liver/drug effects , Protein Biosynthesis , Triiodothyronine/pharmacology , Animals , Base Sequence , Binding Sites , Cells, Cultured , Chlorocebus aethiops , Consensus Sequence , Fibroblasts , Genes , Liver/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins , Polymerase Chain Reaction , Promoter Regions, Genetic , Proteins/genetics , Rats , Receptors, Retinoic Acid/physiology , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Retinoid X Receptors , Transcription Factors/physiology
12.
Mol Endocrinol ; 3(9): 1434-42, 1989 Sep.
Article in English | MEDLINE | ID: mdl-2608066

ABSTRACT

Using a gel shift assay, we analyzed the binding of in vitro translated alpha- and beta-thyroid hormone (T3) receptors to a T3-response element (TRE) derived from the rat GH gene. No receptor-TRE complexes were observed when translated receptor alone was incubated with the TRE. However, addition of a nuclear extract from liver to the translational products resulted in the formation of two receptor-DNA complexes for both the alpha- and beta-receptors. These complexes were shown to contain translated receptor by comigration of 32P-labeled TRE and 35S-labeled receptor in the gel shift assay. A competition experiment demonstrated that formation of the complexes was sequence specific. Preincubation of the liver nuclear extract at 60 C abolished formation of both complexes indicating that receptor-TRE binding required a heat-labile nuclear factor. Phosphocellulose chromatography of the nuclear extract resulted in separation of the activities required for formation of the two complexes. Analysis of nuclear extracts from different tissues revealed that one complex formed in the presence of all extracts, whereas the second complex appeared predominantly with a nuclear extract from liver. Addition of T3 to the binding reaction had no effect on receptor-TRE complex formation. We suggest that nuclear factors interact with the T3 receptor to enhance hormone-independent binding to a TRE.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Thyroid Hormone/physiology , Thyroid Hormones , Triiodothyronine/metabolism , Binding, Competitive , Chromatography , Liver/metabolism , Nuclear Proteins/physiology , Protein Biosynthesis , Transcription, Genetic , Thyroid Hormone-Binding Proteins
13.
Mol Endocrinol ; 8(9): 1147-53, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7838147

ABSTRACT

L-type pyruvate kinase (L-PK) is a key glycolytic enzyme regulating the flux of metabolites through the pyruvate-phosphoenolpyruvate cycle (1). The regulation of L-PK is complex involving both hormones and nutrients. We have found that feeding rats diets containing polyunsaturated fatty acids (PUFA) significantly inhibits hepatic pyruvate kinase enzyme activity (> 60%) and suppresses mRNAPK abundance (> 70%). Studies with primary hepatocytes indicate that PUFA act directly on hepatocytes. Specifically, arachidonic (20:4, omega 6) and eicosapentaenoic (20:5, omega 3) acid suppressed both mRNAPK llevels and the activity of a transfected PKCAT (-4300/+12) fusion gene by > 70%. This is due to an inhibition of the insulin/glucose-mediated transactivation of L-PKCAT. Deletion analysis localized PUFA-regulated cis-acting elements to a region within the L-PK proximal promoter, i.e. between -197 and -96 base pairs. This region binds two transcription factors involved in the hormone/nutrient regulation of L-PK gene transcription, i.e. a major late transcription factor-like factor and HNF-4. Linker scanning mutation analysis localized the PUFA-regulated cis-acting elements to the vicinity of the HNF-4 binding site. Thus, PUFA-regulated factors abrogate the insulin/glucose activation of L-PK gene transcription by targeting the HNF-4 elements. These studies suggest that PUFA may have significant effects on hepatic carbohydrate metabolism by inhibiting the L-PK side of the pyruvate-phosphoenolpyruvate cycle.


Subject(s)
DNA-Binding Proteins , Dietary Fats/pharmacology , Fatty Acids, Unsaturated/pharmacology , Fish Oils/pharmacology , Glucose/pharmacology , Insulin/pharmacology , Phosphoproteins , Pyruvate Kinase/biosynthesis , Transcription, Genetic/drug effects , Animals , Arachidonic Acid/pharmacology , Base Sequence , Cells, Cultured , Depression, Chemical , Eicosapentaenoic Acid/pharmacology , Enzyme Induction/drug effects , Glycolysis/drug effects , Hepatocyte Nuclear Factor 4 , Liver/drug effects , Liver/enzymology , Male , Molecular Sequence Data , Mutagenesis, Site-Directed , Oleic Acid , Oleic Acids/pharmacology , Pyruvate Kinase/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/biosynthesis , Transcription Factors/metabolism
14.
Mol Endocrinol ; 8(12): 1714-9, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7535894

ABSTRACT

The nuclear mechanism by which GH acts to induce gene expression after binding to its receptor on the cell surface is not defined. We have characterized an element in the 5'-flanking region of the rat GH-responsive serine protease inhibitor (Spi) 2.1 gene responsible for its induction by GH. This element binds a hepatic nuclear protein(s) in a GH state-specific manner. Activation of binding by GH does not require de novo protein synthesis, suggesting that a reversible posttranslational process is required for binding to the element. To define the mechanism of this process, hepatic nuclear extracts were analyzed by electrophoretic mobility shift assays using a DNA fragment (-147 to -103) of the Spi 2.1 gene. Treatment of extracts with phosphatases resulted in a marked reduction of GH state-specific binding. Addition of phosphatase inhibitors antagonized the reduction in binding after phosphatase treatment. The specific nature of the phosphorylation event involved in binding was explored using phosphotyrosine antibodies and a protein tyrosine phosphatase. Treatment of nuclear extracts with either of these reagents ablated binding to the response element. Because the tyrosine-phosphorylated transcription factor protein p91 has recently been implicated in cytokine signal transduction mediated by JAK2, we sought evidence that p91 was part of the GH-responsive binding complex. Analysis of an enriched preparation of GH-inducible binding complexes by Western blots using anti-p91 demonstrated no immunoreactivity. We conclude that tyrosine phosphorylation of a nuclear factor is required for GH state-specific binding to this GH response element in vivo, but that p91 is not present in the binding complex.


Subject(s)
Growth Hormone/pharmacology , Nuclear Proteins/metabolism , Tyrosine/analogs & derivatives , Animals , Antibodies/pharmacology , Blotting, Western , Cell Nucleus/chemistry , DNA/metabolism , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Hypophysectomy , Liver/ultrastructure , Male , Nuclear Proteins/genetics , Nuclear Proteins/isolation & purification , Phosphotyrosine , Protein Tyrosine Phosphatases/antagonists & inhibitors , Rats , STAT1 Transcription Factor , Signal Transduction , Trans-Activators/immunology , Trans-Activators/metabolism , Tyrosine/immunology , Tyrosine/metabolism
15.
Endocrinology ; 118(5): 1892-6, 1986 May.
Article in English | MEDLINE | ID: mdl-3754508

ABSTRACT

The messenger RNA (mRNA) coding for the rat hepatic protein spot 14(S14) (mol wt 17,000; pI 4.9) is rapidly responsive to T3 and carbohydrate administration in the adult animal. In 15-day-old suckling rats, the level of expression is 200-fold less than in 60-day-old chow-fed animals. However, the relative transcriptional activity of the S14 gene is only 30% higher in the 60-day-old rat than in the 15-day-old animal. Although T3 induces a 100-fold increase in mRNAS14 in 15-day-old animals, there was only a 17% increase in the S14 gene transcriptional activity. These results indicate that the hormonal and developmental regulation of the S14 gene in liver appears to be predominantly directed at the posttranscriptional level.


Subject(s)
Liver/growth & development , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Triiodothyronine/pharmacology , Animals , Animals, Suckling , Cell Nucleus/metabolism , DNA/genetics , Liver/drug effects , Liver/metabolism , Male , Nuclear Proteins , Nucleic Acid Hybridization , Proteins , Rats , Rats, Inbred Strains , Transcription Factors
16.
Endocrinology ; 110(2): 671-3, 1982 Feb.
Article in English | MEDLINE | ID: mdl-6276146

ABSTRACT

Triiodothyronine (T3) rapidly induces the accumulation of two hepatic mRNA sequences (spot 14 and spot CyT) in thyroidectomized rats as revealed by two-dimensional gel electrophoresis of in vitro translated products of isolated poly(A) containing RNA. T3 acting alone induced a 29-fold increase of spot 14 within 4 h, an increase which was completely inhibited by the concomitant administration of cycloheximide. On the other hand, CyT could be detected only after the administration of cycloheximide, and the combined action of cycloheximide and T3 resulted in a 4-fold increase in CyT 4 h after administration of both agents. These observations suggested that the early cellular action of T3 is contingent on the participation of rapidly turning over protein and mRNA.


Subject(s)
Cycloheximide/pharmacology , Liver/metabolism , RNA, Messenger/metabolism , Triiodothyronine/pharmacology , Animals , Base Sequence , Male , Protein Biosynthesis , Rats , Receptors, Cell Surface/drug effects , Receptors, Thyroid Hormone
17.
Mol Cell Endocrinol ; 150(1-2): 151-9, 1999 Apr 25.
Article in English | MEDLINE | ID: mdl-10411309

ABSTRACT

Rat liver contains a growth hormone inducible nuclear factor complex, GHINF, that binds to the growth hormone response element (GHRE) of the serine protease inhibitor (Spi) 2.1 gene. GHINF contains Stat5 and binds to paired gamma-activated sites (GAS) within the GHRE, but poorly to either one alone. By analysis of the sequence of various GAS sites that bind the GHINF complex (based on the GHRE 3' GAS motif), we demonstrate that a 13 nucleotide high affinity DNA recognition sequence (haGHRE) for GHINF complex binding is (ANTTC)C/T(N)A/G(GAA)A/T(A)/T. One copy of the haGHRE will replace the requirement for two GAS elements present in the wild type promoter in supporting a GH response in primary hepatocyte culture. Mutation of the native Spi 2.1 from a paired GAS site to a single haGHRE does not appreciably change its affinity for binding to the GHINF complex, nor does it alter its sensitivity to GH concentration.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Growth Hormone/metabolism , Liver/metabolism , Milk Proteins , Nuclear Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Animals , DNA/genetics , DNA-Binding Proteins/genetics , Growth Hormone/genetics , Mutation , Nuclear Proteins/genetics , Rats , STAT5 Transcription Factor , Second Messenger Systems , Trans-Activators/genetics , Transcriptional Activation
18.
Arch Surg ; 123(3): 340-4, 1988 Mar.
Article in English | MEDLINE | ID: mdl-3341913

ABSTRACT

The synthesis of acute-phase proteins by the liver during sepsis has been thought to be induced primarily by monokines released from activated macrophages, although glucocorticoid hormones may also stimulate this process to a lesser degree. According to this concept, synthesis of these proteins following administration of bacterial endotoxins would be an indirect effect and would not reflect a direct interaction of the endotoxin molecule with the hepatic parenchymal cell. We observed, however, that the synthesis of a 23-kilodalton protein was stimulated directly by the addition of lipopolysaccharide to cultures of primary mouse hepatocytes. The synthesis of this protein was also stimulated by glucocorticoids and interleukin 1. These findings demonstrate that certain hepatic proteins are subject to complex regulation by several factors thought to be important mediators of sepsis; in addition, they suggest that hepatic parenchymal cells may have the intrinsic capacity to respond directly to bacterial endotoxins.


Subject(s)
Endotoxins/pharmacology , Liver/drug effects , Protein Biosynthesis , Animals , Cells, Cultured , Female , Liver/metabolism , Mice , Mice, Inbred BALB C
20.
Chem Phys Lipids ; 38(3): 223-37, 1985 Sep.
Article in English | MEDLINE | ID: mdl-3910285

ABSTRACT

This report summarizes our recent studies on the protein known as sterol carrier protein (SCP) or fatty acid binding protein (FABP). SCP is a highly abundant, ubiquitous protein with multifunctional roles in the regulation of lipid metabolism and transport. SCP in vitro activates membrane-bound enzymes catalyzing cholesterol synthesis and metabolism, as well as those catalyzing long chain fatty acid metabolism. SCP also binds cholesterol and fatty acids with high affinity and rapidly penetrates cholesterol containing model membranes. Studies in vivo showed SCP undergoes a remarkable diurnal cycle in level and synthesis, induced by hormones and regulated in liver by translational events. SCP rapidly responds in vivo to physiological events and manipulations affecting lipid metabolism by changes in level. Thus SCP appears to be an important regulator of lipid metabolism. Preliminary evidence is presented that SCP is secreted by liver and intestine into blood and then taken up by tissues requiring SCP but incapable of adequate SCP synthesis.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Fatty Acids/metabolism , Neoplasm Proteins , Nerve Tissue Proteins , Tumor Suppressor Proteins , Animals , Carrier Proteins/genetics , Circadian Rhythm , Dexamethasone/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diet , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Female , Humans , Insulin/pharmacology , Kinetics , Liver/drug effects , Liver/metabolism , Male , Protein Biosynthesis , RNA, Messenger/genetics , Rats , Subcellular Fractions/metabolism , Tissue Distribution , Triiodothyronine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL