Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 276(50): 46807-14, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11585816

ABSTRACT

Scavenger receptor class B, type I (SR-BI), is a high density lipoprotein receptor that mediates the flux of cholesterol between high density lipoprotein and cells. Recent evidence suggests that SR-BI plays a role in atherosclerosis and that inflammatory mediators down-regulate SR-BI in the macrophage. The purpose of this study was to evaluate the ability of lipopolysaccharide (LPS) to down-regulate the activity of the human SR-BI promoter in the macrophage and to delineate the mechanisms involved. Experiments with cultured cells and in vivo derived macrophages showed that LPS has a powerful suppressive effect on SR-BI expression both in vitro and in vivo. Transient transfection studies demonstrated that LPS represses SR-BI promoter activity in the macrophage cell line RAW 264.7. Cotransfection with either a constitutively active p21-activated protein kinase-1 (PAK1) construct (T423E) or a kinase-deficient PAK1 construct (K299R) resulted in repression of the SR-BI promoter, similar to LPS. These results demonstrate that PAK1-mediated down-regulation of the SR-BI promoter is independent of PAK1 kinase activity and suggest that PAK1 mediates the LPS-induced decrease in promoter activity. Cotransfection with constitutively active Cdc42 or Rac expression constructs also resulted in down-regulation of the promoter; whereas the dominant-negative Cdc42 and Rac constructs elevated basal promoter activity and blunted the LPS response. Cotransfection of PAK1 constructs containing mutations in both the kinase domain and the Cdc42/Rac-binding domain attenuated the PAK1-mediated down-regulation of the promoter, suggesting that Rac and Cdc42 are required for PAK1-mediated decreases in SR-BI promoter activity. 5'-Deletion analysis and gel shift data suggest that LPS inhibits binding of a novel transcription factor to a myeloid zing finger protein-1-like element (-476 to -456) in the human SR-BI promoter. These results demonstrate that the PAK1 pathway down-regulates the SR-BI promoter and suggest that activation of this pathway may play an important role in cholesterol trafficking in the vessel wall.


Subject(s)
CD36 Antigens/genetics , Macrophages/enzymology , Membrane Proteins , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Receptors, Immunologic , Receptors, Lipoprotein , Animals , Base Sequence , Binding, Competitive , Blotting, Western , Cell Nucleus/metabolism , Cells, Cultured , Cholesterol/metabolism , Cytoskeleton/metabolism , Down-Regulation , GTP Phosphohydrolases/metabolism , Genes, Dominant , Humans , Lipopolysaccharides/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , RNA/metabolism , RNA, Messenger/metabolism , Receptors, Scavenger , Scavenger Receptors, Class B , Transfection , cdc42 GTP-Binding Protein/genetics , p21-Activated Kinases , rac GTP-Binding Proteins/genetics
2.
Endocrinology ; 142(5): 1935-49, 2001 May.
Article in English | MEDLINE | ID: mdl-11316759

ABSTRACT

Mechanisms of transcriptional regulation of the human beta(3)-adrenergic receptor were studied using SK-N-MC cells, a human neuroblastoma cell line that expresses beta(3)- and beta(1)-adrenergic receptors endogenously. Deletions spanning different portions of a 7-kb 5'-flanking region of the human beta(3)-adrenergic receptor gene were linked to a luciferase reporter and transfected in SK-N-MC, CV-1, and HeLa cells. Maximal luciferase activity was observed when a 200-bp region located between -6.5 and -6.3 kb from the translation start site was present. This region functioned only in SK-N-MC cells. Electrophoretic mobility shift assays of nuclear extracts from SK-N-MC, CV-1, and HeLa cells using double stranded oligonucleotides spanning different portions of the 200-bp region as probes and transient transfection studies revealed the existence of three cis-acting regulatory elements: A) -6.468 kb-AGGTGGACT--6.458 kb, B) -6.448 kb-GCCTCTCTGGGGAGCAGCTTCTCC-6.428 kb, and C) -6.405 kb-20 repeats of CCTT-6.385 kb. These elements act together to achieve full transcriptional activity. Mutational analysis, antibody supershift, and electrophoretic mobility shift assay competition experiments indicated that element A binds the transcription factor Sp1, element B binds protein(s) present only in nuclear extracts from SK-N-MC cells and brown adipose tissue, and element C binds protein(s) present in both SK-N-MC and HeLa cells. In addition, element C exhibits characteristics of an S1 nuclease-hypersensitive site. These data indicate that cell-specific positive cis-regulatory elements located 6.5 kb upstream from the translation start site may play an important role in transcriptional regulation of the human beta(3)-adrenergic receptor. These data also suggest that brown adipose tissue-specific transcription factor(s) may be involved in the tissue-specific expression of the beta(3)-adrenergic receptor gene.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Receptors, Adrenergic, beta-3/genetics , Adipose Tissue, Brown/metabolism , Base Sequence , Binding Sites , Genes, Regulator , Humans , Molecular Sequence Data , Neuroblastoma/genetics , Promoter Regions, Genetic , Single-Strand Specific DNA and RNA Endonucleases/metabolism , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Tumor Cells, Cultured
3.
Endocrinology ; 141(9): 3403-11, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10965913

ABSTRACT

Functional interactions or cross-talk between ligand-activated nuclear receptors and the proinflammatory transcription factor nuclear factor-kappaB (NF-kappaB) may play a major role in ligand-mediated modification of diseases processes. In particular, the cardioprotective effects of estrogen replacement therapy are thought to be due in part to the ability of ligand-bound estrogen receptor (ER) to inhibit NF-kappaB function. In the current study 17beta-estradiol-bound ERalpha interfered with cytokine-induced activation of a NF-kappaB reporter in HepG2 cells. The estrogen metabolite, 17alpha-ethinyl estradiol, and the phytoestrogen, genistein, were also effective inhibitors of NF-kappaB activation, whereas tamoxifen, 4-hydroxytamoxifen, and raloxifene were inactive. This inhibition was reciprocal, as NF-kappaB interfered with the trans-activation properties of ERalpha. Ligand-bound ERalpha did not inhibit NF-kappaB binding to DNA, but it did decrease the histone acetyltransferase activity required for NF-kappaB transcriptional activity. Coexpression of the transcription coactivator CREB binding protein (CBP), but not steroid receptor coactivator 1a, reversed the ERalpha-mediated inhibition of NF-kappaB activity. Mammalian two-hybrid experiments also revealed that ligand-bound ERalpha can interact functionally with CBP-NF-kappaB complexes. We suggest that CBP targeting by ERalpha results in the inhibition of NF-kappaB and may occur through formation of transcriptionally inert multimeric complexes that are dependent upon the nature of the ERalpha ligand.


Subject(s)
NF-kappa B/physiology , Nuclear Proteins/physiology , Receptor Cross-Talk/physiology , Receptors, Estrogen/physiology , Trans-Activators/physiology , Adenoviridae/genetics , Anticholesteremic Agents/pharmacology , Blotting, Western , CREB-Binding Protein , Cell Line , Electrophoresis , Estrogens/pharmacology , Genetic Vectors , Histone Deacetylase Inhibitors , Humans , Interleukin-1/antagonists & inhibitors , Interleukin-1/pharmacology , Luciferases/genetics , Plasmids/genetics , Receptor Cross-Talk/drug effects , Receptors, Estrogen/drug effects , Transfection/genetics
4.
Biochem Biophys Res Commun ; 269(1): 46-53, 2000 Mar 05.
Article in English | MEDLINE | ID: mdl-10694475

ABSTRACT

Since human hepatocytes are available only in limited number, the development of a serum-free culture system for long-term cultivation of differentiated and functional hepatocytes is of great importance. Here we describe the culture of human hepatocytes in a chemically defined serum-free medium for up to 5 weeks. Cell morphology was assayed by light and electron microscopy and revealed a well-preserved cellular morphology. Marker proteins for epithelial and bile duct cells, cytokeratin (CK) 18 and 19, and liver-specific proteins, like phosphoenolpyruvate carboxykinase-2 (PCK2) and serum proteins, were expressed. Liver-enriched transcription factors CCAAT/enhancer binding protein alpha (C/EBPalpha) and hepatocyte nuclear factor-4 (HNF-4), cytokine and mitogen activated factors (nuclear factor kappa B) NFkappaB, and activator protein-1 (AP-1) were maintained and active for several weeks in our cultures. In summary, our serum-free culture system allows the culture of differentiated human hepatocytes for several weeks. It may serve as a model system for metabolic, pharmacologic-toxicologic studies, and studies on human pathogens under defined chemical conditions.


Subject(s)
Liver/cytology , Liver/physiology , Transcription Factors/metabolism , Blood Proteins/genetics , Blood Proteins/metabolism , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Culture Media, Serum-Free , Gene Expression , Humans , Liver/metabolism , Microscopy, Electron , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Transcription Factors/genetics
5.
Nucleic Acids Res ; 26(7): 1761-8, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9512550

ABSTRACT

The apolipoprotein AI (apoAI) promoter/enhancer contains multiple cis -acting elements on which a variety of hepatocyte-enriched and ubiquitous transcription factors function synergistically to regulate liver-specific transcription. Adenovirus E1A proteins repress tissue-specific gene expression and disrupt the differentiated state in a variety of cell types. In this study expression of E1A 12Sor 13S in hepatoblastoma HepG2 cells repressed apoAI enhancer activity 8-fold. Deletion mapping analysis showed that inhibition by E1A was mediated by the apoAI promoter site B. E1A selectively inhibited the ability of HNF3beta and HNF3alpha to transactivate reporter genes controlled by the apoAI site B and the HNF3 binding site from the transthyretin promoter. The E1A-mediated repression of HNF3 activity was not reversed by overexpression of HNF3beta nor did E1A alter nuclear HNF3beta protein levels or inhibit HNF3 binding to DNA in mobility shift assays. Overexpression of two cofactors known to interact with E1A, pRb and CBP failed to overcome inhibition of HNF3 activity. Similarly, mutations in E1A that disrupt its interaction with pRb or CBP did not compromise its ability to repress HNF3beta transcriptional activity. These data suggest that E1A inhibits HNF3 activity by inactivating a limiting cofactor(s) distinct from pRb or CBP.


Subject(s)
Adenovirus E1A Proteins/metabolism , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/genetics , Enhancer Elements, Genetic , Liver/metabolism , Promoter Regions, Genetic , Binding Sites , Carcinoma, Hepatocellular , Chloramphenicol O-Acetyltransferase/biosynthesis , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Genes, Reporter , Hepatocyte Nuclear Factor 3-alpha , Hepatocyte Nuclear Factor 3-beta , Humans , Liver Neoplasms , Luciferases/biosynthesis , Nuclear Proteins/metabolism , Organ Specificity , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/biosynthesis , TATA Box , Transcription Factors , Transcriptional Activation , Transfection , Tumor Cells, Cultured
6.
J Biol Chem ; 273(15): 9270-8, 1998 Apr 10.
Article in English | MEDLINE | ID: mdl-9535920

ABSTRACT

Estrogen replacement therapy increases plasma concentrations of high density lipoprotein and its major protein constituent, apolipoprotein AI (apoAI). Studies with animal model systems, however, suggest opposite effects. In HepG2 cells stably expressing estrogen receptor alpha (ERalpha), 17beta-estradiol (E2) potently inhibited apoAI mRNA steady state levels. ApoAI promoter deletion mapping experiments indicated that ERalpha plus E2 inhibited apoAI activity through the liver-specific enhancer. Although the ERalpha DNA binding domain was essential but not sufficient for apoAI enhancer inhibition, ERalpha binding to the apoAI enhancer could not be detected by electrophoretic mobility shift assays. Western blotting and cotransfection assays showed that ERalpha plus E2 did not influence the abundance or the activity of the hepatocyte-enriched factors HNF-3beta and HNF-4, two transcription factors essential for apoAI enhancer function. Expression of the ERalpha coactivator RIP140 dramatically repressed apoAI enhancer function in cotransfection experiments, suggesting that RIP140 may also function as a coactivator on the apoAI enhancer. Moreover, estrogen regulation of apoAI enhancer activity was dependent upon the balance between ERalpha and RIP140 levels. At low ratios of RIP140 to ERalpha, E2 repressed apoAI enhancer activity, whereas at high ratios this repression was reversed. Regulation of the apoAI gene by estrogen may thus vary in direction and magnitude depending not only on the presence of ERalpha and E2 but also upon the intracellular balance of ERalpha and coactivators utilized by ERalpha and the apoAI enhancer.


Subject(s)
Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/genetics , DNA-Binding Proteins/metabolism , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Receptors, Estrogen/metabolism , Transcription, Genetic/drug effects , Adaptor Proteins, Signal Transducing , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Binding Sites , Carcinoma, Hepatocellular , Enhancer Elements, Genetic , Hepatocyte Nuclear Factor 3-beta , Hepatocyte Nuclear Factor 4 , Humans , Kinetics , Liver Neoplasms , Luciferases/biosynthesis , Nuclear Receptor Interacting Protein 1 , Phosphoproteins/metabolism , Promoter Regions, Genetic/drug effects , RNA, Messenger/biosynthesis , Receptors, Estrogen/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured
7.
J Steroid Biochem Mol Biol ; 67(5-6): 421-9, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10030691

ABSTRACT

Phytoestrogens are defined as plant substances that are structurally or functionally similar to estrogen. They are present in many foods and their higher consumption in certain populations has been correlated with protection against many diseases including coronary heart disease, breast cancer and endometrial and ovarian cancer. In this report, ten phytoestrogens with diverse chemical structures were studied for their binding to the human estrogen receptor and their transcription activation properties in yeast and mammalian cells. Our results showed that some of these compounds bind with relatively high affinity to the estrogen receptor and activate the receptor in the yeast and mammalian cell system. In addition, none of these compounds showed anti-estrogenic activity. We conclude that the yeast system accurately predicts the estrogenic activity of compounds with diverse chemical structures in mammalian cells. In addition, our data with phytoestrogens that do not show transcription activation properties raise the possibility that these compounds may exert their biological effects through pathways different from the classical estrogen signalling mechanism.


Subject(s)
Estrogens, Non-Steroidal/pharmacology , Isoflavones , Receptors, Estrogen/metabolism , Binding, Competitive , Carcinoma, Hepatocellular , Cloning, Molecular , Estradiol/metabolism , Humans , Kinetics , Liver Neoplasms , Phytoestrogens , Plant Preparations , Radioligand Assay , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Transcriptional Activation , Transfection , Tumor Cells, Cultured , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
8.
J Biol Chem ; 272(17): 11384-91, 1997 Apr 25.
Article in English | MEDLINE | ID: mdl-9111047

ABSTRACT

The estrogen receptor (ER) belongs to a superfamily of ligand-inducible transcription factors. Functions of these proteins (dimerization, DNA binding, and interaction with other transcription factors) are modulated by binding of their corresponding ligands. It is, however, controversial whether various ER ligands affect the receptor's ability to bind its specific DNA element (ERE). By using real time interaction analysis we have investigated the kinetics of human (h)ER binding to DNA in the absence and presence of 17beta-estradiol, 17alpha-ethynyl estradiol, analogs of tamoxifen, raloxifene, and ICI-182,780. We show that ligand binding dramatically influences the kinetics of hER interaction with specific DNA. We have found that binding of estradiol induces the rapid formation of a relatively unstable ER.ERE complex, and binding of ICI-182,780 leads to slow formation (ka is approximately 10 times lower) of a stable receptor-DNA complex (kd is almost 2 orders of magnitude lower). Therefore, binding of estradiol accelerates the frequency of receptor-DNA complex formation more than 50-fold, compared with unliganded ER, and more than 1000-fold compared with ER liganded with ICI-182,780. We hypothesize that a correlation exists between the rate of gene transcription and the frequency of receptor-DNA complex formation. We further show that a good correlation exists between the kinetics of hER-ERE interaction induced by a ligand and its biological effect.


Subject(s)
DNA-Binding Proteins/metabolism , Receptors, Estrogen/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Ethinyl Estradiol/pharmacology , Fulvestrant , Humans , Ligands , Piperidines/pharmacology , Protein Binding/drug effects , Raloxifene Hydrochloride , Tamoxifen/analogs & derivatives
9.
J Biol Chem ; 271(23): 13621-8, 1996 Jun 07.
Article in English | MEDLINE | ID: mdl-8662915

ABSTRACT

Apolipoprotein AI (apoAI) gene expression in liver depends on synergistic interactions between transcription factors bound to three distinct sites (A, B, and C) within a hepatocyte-specific enhancer in the 5'-flanking region of the gene. In this study, we showed that a segment spanning sites A and B retains substantial levels of enhancer activity in hepatoblastoma HepG2 cells and that sites A and B are occupied by the liver-enriched hepatocyte nuclear factors (HNFs) 4 and 3, respectively, in these cells. In non-hepatic CV-1 cells, HNF-4 and HNF-3beta activated this minimal enhancer synergistically. This synergy was dependent upon simultaneous binding of these factors to their cognate sites, but it was not due to cooperativity in DNA binding. Separation of these sites by varying helical turns of DNA did not affect simultaneous binding of HNF-3beta and HNF-4 nor did it influence their functional synergy. The synergy was, however, dependent upon the cell type used for functional analysis. In addition, this synergy was further potentiated by estrogen treatment of cells cotransfected with the estrogen receptor. These data indicate that a cell type-restricted intermediary factor jointly recruited by HNF-4 and HNF-3 participates in activation of the apoAI enhancer in liver cells and suggest that the activity of this factor is regulated by estrogen.


Subject(s)
Apolipoprotein A-I/genetics , DNA-Binding Proteins/administration & dosage , Gene Expression Regulation/drug effects , Nuclear Proteins/administration & dosage , Phosphoproteins/administration & dosage , Transcription Factors/administration & dosage , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Binding Sites/genetics , Cell Line , DNA-Binding Proteins/metabolism , Drug Synergism , Enhancer Elements, Genetic , Estradiol/pharmacology , HeLa Cells , Hepatocyte Nuclear Factor 3-beta , Hepatocyte Nuclear Factor 4 , Humans , L Cells , Liver/drug effects , Liver/metabolism , Mice , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Transcription Factors/metabolism
10.
Mol Cell Biol ; 16(4): 1824-31, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8657158

ABSTRACT

The orphan nuclear receptor hepatocyte nuclear factor 4 (HNF-4) is required for development and maintenance of the liver phenotype. HNF-4 activates several hepatocyte-specific genes, including the gene encoding apolipoprotein AI (apoAI), the major protein component of plasma high-density lipoprotein. The apoAI gene is activated by HNF-4 through a nuclear receptor binding element (site A) located in its liver-specific enhancer. To decipher the mechanism whereby HNF-4 enhances apoAI gene transcription, we have reconstituted its activity in a cell-free system. Functional HNF-4 was purified to homogeneity from a bacterial expression system. In in vitro transcription assays employing nuclear extract from HeLa cells, which do not contain HNF-4, recombinant HNF-4 stimulated transcription from basal promoters linked to site A. Activation by HNF-4 did not exhibit a ligand requirement, but phosphorylation of HNF-4 in the in vitro transcription system was observed. The activation function of HNF-4 was localized to a domain displaying strong homology to the conserved AF-2 region of nuclear receptors. Dissection of the transcription cycle revealed that HNF-4 activated transcription by facilitating assembly of a preinitiation complex intermediate consisting of TBP, the TATA box-binding protein component of TFIID and TFIID, via direct physical interactions with TFIIB. However, recruitment of TFIIB by HNF-4 was not sufficient for activation, since HNF-4 deletion derivatives lacking AF-2 bound TFIIB. On the basis of these results, HNF-4 appears to activate transcription at two distinct levels. The first step involves AF-2-independent recruitment of TFIIB to the promoter complex; the second step is AF-2 dependent and entails entry of preinitiation complex components acting downstream of TFIIB.


Subject(s)
DNA-Binding Proteins/genetics , Liver/metabolism , Phosphoproteins/genetics , Transcription Factors/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , Apolipoprotein A-I/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , DNA-Binding Proteins/metabolism , HeLa Cells , Hepatocyte Nuclear Factor 4 , Humans , Ligands , Liver/cytology , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Rats , Transcription Factor TFIIB , Transcription Factors/metabolism
11.
J Clin Invest ; 96(1): 528-38, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7615825

ABSTRACT

We have used apolipoprotein genes to investigate the signal transduction mechanisms involved in the control of intestinal specific gene expression. The human apoAI, apoCIII, and apoAIV genes are tandemly organized within a 15-kb DNA segment and are expressed predominantly in the liver and intestine. Transient transfection of various human apoAI gene plasmid constructs into human hepatoma (HepG2) and colon carcinoma (Caco-2) cells showed that apoAI gene transcription is under the control of two separate and distinct cell-specific promoters. The region between nucleotides -192 and -41 is essential for expression in HepG2 cells, whereas the region from -595 to -192 is essential for expression in Caco-2 cells. A third 0.6 kb DNA fragment in the apoCIII gene promoter region, approximately 5 kb down-stream from the human apoAI gene, enhances transcription mediated by either of these two tissue-specific apoAI promoters. In Caco-2 cells, expression of the apoAI gene and activation by the distal enhancer required the presence of a nuclear hormone receptor response element (NHRRE) located in the -214 to -192 apoAI promoter region. Overexpression of the orphan receptor hepatocyte nuclear factor 4 (HNF-4), which binds to the NHRRE, dramatically stimulates apoAI gene expression in Caco-2 cells but not in HepG2 cells. Maximal stimulation of transcription by HNF-4 in Caco-2 cells required the presence of both the intestinal specific promoter, the NHRRE, and distal enhancer elements. Transactivation by HNF-4 thus appears to result from functional synergy between the NHRRE binding HNF-4 and distal DNA elements containing intestinal-specific DNA binding activities. The apoAI gene provides a model system to define the mechanism(s) governing intestinal cell specific gene regulation and the role of nuclear hormone receptors in the establishment and regulation of enterocytic gene transcription.


Subject(s)
Apolipoprotein A-I/genetics , DNA-Binding Proteins , DNA/metabolism , Gene Expression Regulation , Phosphoproteins , Transcription Factors/physiology , Apolipoprotein C-III , Apolipoproteins A/genetics , Apolipoproteins C/genetics , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Binding Sites , Cells, Cultured , Hepatocyte Nuclear Factor 4 , Humans , Intestinal Mucosa/metabolism , Liver/metabolism , Molecular Sequence Data , Organ Specificity , Promoter Regions, Genetic , Transcription, Genetic
12.
Nucleic Acids Res ; 23(9): 1536-43, 1995 May 11.
Article in English | MEDLINE | ID: mdl-7784207

ABSTRACT

ARP-1 is a ubiquitous orphan nuclear receptor that binds to a site (site A) in the apolipoprotein AI (apoAI) liver-specific enhancer and represses its transcriptional activity in hepatoblastoma HepG2 cells. Electrophoretic mobility shift analysis of HepG2 cell nuclear extracts showed that in addition to ARP-1, site A also binds the orphan nuclear receptors Ear-2 and HNF-4. In in vitro transcription assays, Hela cell nuclear extracts which contain ARP-1 had no effect on transcription from a basal promoter linked to multiple copies of site A. However, supplementation of these extracts with excess amounts of recombinant ARP-1 resulted in significant stimulation. Supplementation of the extracts with purified polypeptides representing fusions between the ARP-1 N- or C-terminal domains and the yeast activator GAL4 DNA binding domain also stimulated transcription from a basal promoter linked to multiple GAL4 DNA binding sites. Co-immunoprecipitation assays using ARP-1-selective antibodies revealed specific physical interactions between ARP-1 and the basal transcription factor TFIIB. We conclude that ARP-1 possesses intrinsic transcription activation potential which is modulated, at least in part, by the intracellular balance of other nuclear receptors that also bind to its cognate DNA binding site.


Subject(s)
Apolipoproteins A/metabolism , DNA-Binding Proteins/metabolism , Receptors, Steroid/metabolism , Transcriptional Activation , COUP Transcription Factor II , COUP Transcription Factors , Cell-Free System , Cells, Cultured , DNA-Binding Proteins/genetics , Escherichia coli/genetics , HeLa Cells , Humans , Receptors, Steroid/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factor TFIIB , Transcription Factors/metabolism
13.
J Biol Chem ; 270(12): 7004-10, 1995 Mar 24.
Article in English | MEDLINE | ID: mdl-7896852

ABSTRACT

Liver-specific expression of the apolipoprotein AI (apoAI) gene is mediated by transcription factors bound to three sites (A, B, and C) in the apoAI enhancer. Sites A and C bind various members of the nuclear receptor superfamily, including the orphan nuclear receptor apolipoprotein regulatory protein-1 (ARP-1); site B binds the liver-enriched factor hepatic nuclear factor-3. The immediate early growth response factor (Egr-1), which is transiently expressed in various pathophysiologic states of the liver, activates the apoAI enhancer and overcomes ARP-1-mediated repression of the enhancer in hepatoblastoma HepG2 cells. Deletion mapping analysis revealed two Egr-1 binding sites, E1 and E2, flanking site A. Erg-1 bound efficiently to both E1 and E2. Sp1 in HepG2 nuclear extracts bound to E2 but not E1. In HepG2 cells, E1 functioned as an Egr-1 response element, whereas E2 had high basal activity and was not further induced by Egr-1. Mutations that prevent Egr-1 binding to the apoAI enhancer abolished its responsiveness to Erg-1, while they had only minor effects on its constitutive activity. These mutations also diminished the ability of Egr-1 to overcome ARP-1-mediated repression. Elimination of transcription factor binding to sites A, B, or C reduced enhancer activity without affecting Egr-1-dependent activation. We argue that Egr-1 is recruited to the apoAI enhancer complex under unusual circumstances, such as those prevailing during liver regeneration, to maintain apoAI transcription levels by overriding prior transcriptional controls.


Subject(s)
Apolipoprotein A-I/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation , Immediate-Early Proteins , Transcription Factors/physiology , Base Sequence , Early Growth Response Protein 1 , Enhancer Elements, Genetic , Humans , Molecular Sequence Data , Sp1 Transcription Factor/physiology , Transcription, Genetic
14.
Gene Expr ; 4(6): 319-36, 1995.
Article in English | MEDLINE | ID: mdl-7549464

ABSTRACT

Many traditional drugs target cell surface receptors. Medicinal chemists and pharmacologists have not ventured into the field of transcription regulation due to the fear that drugs that interfere with transcription regulation may not be selective or efficacious. The past 5 years have seen some exciting developments in the field of signal transduction in general, and transcription regulation in particular. Our understanding of mechanisms of regulated and basal transcription is advanced to a degree that it should be possible to selectively modulate a target gene directly. In this review we have argued that sufficient diversity exists in the combinatorial interplay of the transcription factors to offer opportunities for selective therapeutic intervention. We have focused our attention on transcriptional factors that play a role in three different therapeutic areas: osteoporosis, immune modulation, and cardiovascular diseases. Human estrogen receptor is considered as a model transcription factor. The role of estrogen in bone remodeling is discussed. Opportunities for tissue-specific modulation of estrogen receptors are described. For selective immune modulation, we have discussed the role of NF-AT (nuclear factors for activated T cells) transcription factors in interleukin-2 gene regulation. The last section focuses on the transcriptional mechanisms conferring tissue specificity in regulated expression of the apoAI gene, a major component of HDL, in liver. We have highlighted opportunities for rational development of transcription-based drugs useful for raising HDL plasma levels and atherosclerosis prevention.


Subject(s)
Cardiovascular Diseases/drug therapy , Drug Design , Immunity/drug effects , Osteoporosis/drug therapy , Transcription Factors/drug effects , Eukaryotic Cells , Humans , Lipoproteins, HDL/blood , Receptors, Adrenergic/drug effects , Transcription, Genetic/drug effects
15.
Nucleic Acids Res ; 22(25): 5665-71, 1994 Dec 25.
Article in English | MEDLINE | ID: mdl-7838720

ABSTRACT

Hepatocyte Nuclear Factor 4 (HNF-4), a liver-enriched orphan receptor of the nuclear receptor superfamily, is required for the expression of a wide variety of liver-specific genes including apoAI. To explore the possibility that site A of the apoAI gene enhancer might also be the target for HNF-4 without the interference of endogenous mammalian cell proteins that also bind to site A, we tested the ability of HNF-4 to activate transcription from site A in yeast cells. Electrophoretic mobility shift assays (EMSA) and Scatchard plot analysis demonstrated that yeast produced HNF-4 binds to site A with an affinity two times higher than that of yeast produced RXR alpha. Mapping analysis indicated that the 5' portion of site A containing two imperfect direct repeats (TGAACCCTTGACC) and the sequence of the trinucleotide spacer (CCT) between these imperfect repeats are critical determinants for selective binding and transactivation by HNF-4. Similar observations were obtained when these mutated versions of site A were evaluated by transient cotransfection assays in CV1 cells. We conclude that the unique structural determinants of site A in conjunction with the differential binding affinity of HNF-4 for site A may play a fundamental role in apoAI gene regulation.


Subject(s)
Apolipoprotein A-I/genetics , Phosphoproteins , Transcription Factors/physiology , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Hepatocyte Nuclear Factor 4 , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors , Saccharomyces cerevisiae , Transcription Factors/metabolism , Transfection
16.
J Biol Chem ; 269(45): 28220-6, 1994 Nov 11.
Article in English | MEDLINE | ID: mdl-7961760

ABSTRACT

Liver-specific expression of the apolipoprotein AI (apoA-I) gene is controlled by the coordinate action of transcription factors bound to three sites (A, B, and C) located within a powerful liver-specific enhancer which spans the -222 to -110 region upstream of the apoA-I gene transcription start site (+1). Sites A and C bind various members of the nuclear receptor superfamily including the liver-enriched factor HNF-4. In the current report, enhancer derivatives with mutagenized protein-binding sites were tested for their ability to stimulate the apoA-I basal promoter in hepatoblastoma HepG2 cells. The results revealed that occupation of both sites A and B, but not C is essential for high level expression. Electrophoretic mobility shift assays showed that in HepG2 cells site B is occupied by the liver-enriched factor HNF-3 beta. Binding of HNF-3 beta to site B transactivates the apoA-I basal promoter in hepatic and nonhepatic cells. HNF-3 beta binding and transactivation were dependent upon the close proximity of two HNF-3 beta binding motifs within site B. Furthermore, HNF-3 beta and HNF-4, bound to their cognate sites within the apoA-I enhancer exhibited strong synergy in transactivation of the apoA-I basal promoter in nonhepatic cells, highlighting the central role of HNF-3 beta in liver-specific transcription of the apoA-I gene. It is concluded that cooperative binding of HNF-3 beta to site B and synergistic interactions between HNF-4 and HNF-3 beta bound to their cognate sites in the apoA-I enhancer may play a fundamental role in apoA-I gene expression in liver.


Subject(s)
Apolipoprotein A-I/biosynthesis , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Liver/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , CHO Cells , Carcinoma, Hepatocellular , Cell Line , Chlorocebus aethiops , Cricetinae , DNA/metabolism , HeLa Cells , Hepatocyte Nuclear Factor 3-beta , Humans , Kidney , Liver Neoplasms , Luciferases/biosynthesis , Molecular Sequence Data , Oligodeoxyribonucleotides , Organ Specificity , Promoter Regions, Genetic , Transcription, Genetic , Transfection
17.
Gene ; 145(1): 129-33, 1994 Jul 22.
Article in English | MEDLINE | ID: mdl-8045412

ABSTRACT

The possibility that different retinoids activate transcription from a specific retinoic acid (RA)-responsive element known as site A via different homo and heterodimeric versions of RA receptors cannot be evaluated in mammalian cells because they contain endogenous RA receptors (RAR). However, this limitation can be overcome by using yeast cells, which do not contain endogenous RAR, to study retinoid signaling pathways. Here, we describe heterologous expression of the human retinoid X receptor (RXR alpha) in yeast and hormone-dependent activation of a reporter construct containing site A upstream from a yeast promoter fused to the lacZ gene of Escherichia coli. Western blot analysis of yeast extracts containing RXR alpha revealed a distinct immunoreactive polypeptide co-migrating with the mammalian-produced RXR alpha. Electrophoretic mobility shift assays demonstrated that RXR alpha produced in yeast binds efficiently to site A in the absence of 9-cis-RA. However, transcription activation experiments showed that RXR alpha transactivates a yeast basal promoter linked to site A only in the presence of 9-cis-RA. We conclude that RXR alpha homodimers bind to site A in the absence of 9-cis-RA, but function as ligand-dependent transactivators in yeast cells. This retinoid-responsive transcription unit created in yeast cells provides a powerful genetic tool for the systemic unraveling of the synergistic interactions between RXR alpha and its heterodimeric partners.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid , Receptors, Steroid , Saccharomyces cerevisiae/metabolism , Trans-Activators/metabolism , Transcription Factors , Base Sequence , Blotting, Western , COUP Transcription Factors , Cloning, Molecular , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Humans , Molecular Sequence Data , Oligodeoxyribonucleotides , Promoter Regions, Genetic , Retinoid X Receptors , Saccharomyces cerevisiae/genetics , Signal Transduction
18.
J Biol Chem ; 269(18): 13185-92, 1994 May 06.
Article in English | MEDLINE | ID: mdl-8175747

ABSTRACT

Expression of the apolipoprotein AI (apoAI) gene in the liver is controlled by a liver-specific enhancer. The function of this enhancer depends on synergistic interactions between transcription factors bound to at least three sites (designated A, B, and C) located within this enhancer. We have previously shown that an apoAI gene reporter construct containing the entire enhancer is expressed efficiently in a hepatoma cell line and that its activity is repressed by the orphan receptor ARP-1. Moreover, repression by ARP-1 is overcome by the retinoid X receptor RXR alpha in the presence of retinoic acid. In this study, we show that ARP-1 represses the apoAI promoter by binding to site A of the apoAI liver-specific enhancer, the repression being a promoter context-specific event. Mapping analysis of ARP-1 indicated that its DNA binding domain is essential but not sufficient for repression. Two separate repression domains located at the amino- and carboxyl-terminal halves of ARP-1 were found to individually complement the DNA binding domain for efficient repression. We also demonstrate the reversibility of ARP-1 repression by transcription factors C/EBP and Egr-1, which might also be involved in apoAI gene expression. Significantly, repression by ARP-1 was found to be a prerequisite for C/EBP-mediated transactivation. We interpret our results in terms of a model in which ARP-1 repression via its interaction with site A is an obligatory intermediate step in switching from one activated state of the apoAI gene to another.


Subject(s)
Apolipoprotein A-I/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation , Immediate-Early Proteins , Receptors, Steroid/physiology , Transcription, Genetic , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins , COUP Transcription Factor II , COUP Transcription Factors , Cells, Cultured , Chickens , DNA/metabolism , DNA-Binding Proteins/metabolism , Early Growth Response Protein 1 , Enhancer Elements, Genetic , Haplorhini , Humans , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured
19.
Mol Cell Biol ; 12(8): 3380-9, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1321332

ABSTRACT

The gene coding for apolipoprotein AI (apoAI), a lipid binding protein involved in the transport of cholesterol and other lipids in the plasma, is expressed in mammals predominantly in the liver and the intestine. Liver-specific expression is controlled by synergistic interactions between transcription factors bound to three separate sites, sites A (-214 to -192), B (-169 to -146), and C (-134 to -119), within a powerful liver-specific enhancer located between nucleotides -222 and -110 upstream of the apoAI gene transcription start site (+1). Previous studies in our laboratory have shown that ARP-1, a member of the nuclear receptor superfamily whose ligand is unknown (orphan receptor), binds to site A and represses transcription of the apoAI gene in liver cells. In a more recent series of experiments, we found that site A is a retinoic acid (RA) response element that responds preferentially to the recently identified RA-responsive receptor RXR alpha over the previously characterized RA receptors RAR alpha and RAR beta. In this study we investigated the combined effects of ARP-1 and RXR alpha on apoAI gene expression in liver cells. Transient transfection assays showed that site A is necessary and sufficient for RXR alpha-mediated transactivation of the apoAI gene basal promoter in human hepatoma HepG2 cells in the presence of RA and that this transactivation is abolished by increasing amounts of cotransfected ARP-1. Electrophoretic mobility shift assays and subsequent Scatchard analysis of the data revealed that ARP-1 and RXR alpha bind to site A with similar affinities. These assays also revealed that ARP-1 and RXR alpha bind to site A as heterodimers with an affinity approximately 10 times greater than that of either ARP-1 or RXR alpha alone. Further transfection assays in HepG2 cells, using as a reporter a construct containing the apoAI gene basal promoter and its upstream regulatory elements (including site A) in their natural context, revealed that RXR alpha has very little effect on the levels of expression regardless of the presence or absence of RA. However, while ARP-1 alone or ARP-1 and RXR alpha together dramatically repress expression in the absence of RA, the repression by ARP-1 and RXR alpha together, but not ARP-1 alone, is almost completely alleviated in the presence of RA. These results indicate that transcriptional repression by ARP-1 sensitizes apoAI gene responsiveness to RXR alpha and RA and suggest that the magnitude of this responsiveness is regulated by the intracellular ratio of ARP-1 to RXR alpha. These observations raise the possibility that transcriptional repression is a general mechanism for switching gene transcription between alternative transcription activation pathways.


Subject(s)
Apolipoprotein A-I/genetics , DNA-Binding Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Retinoic Acid , Receptors, Steroid/metabolism , Transcription Factors , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Binding Sites , COUP Transcription Factor II , COUP Transcription Factors , Carcinoma, Hepatocellular , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Cloning, Molecular , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , Humans , Kinetics , Liver Neoplasms , Macromolecular Substances , Plasmids , Promoter Regions, Genetic , Receptors, Cell Surface/genetics , Receptors, Cell Surface/isolation & purification , Retinoid X Receptors , Transcriptional Activation , Transfection
20.
J Lipid Res ; 33(6): 831-42, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1512510

ABSTRACT

In mammals, the apolipoprotein (apo) A-I gene is expressed predominantly in liver and intestine, while in avian species it is expressed in all tissues. Although liver and intestine are the major sites of chicken apoA-I mRNA synthesis, there are appreciable amounts of apoA-I mRNA in kidney, ovary/testes, brain, lung, skeletal, and heart muscle. In this study, the nucleotide sequences of the chicken apoA-I gene and its 5' flanking region, as well as the sequences involved in the expression of this gene, have been determined. The gene spans 1.5 kilobases and contains 4 exons and 3 introns, closely resembling the mammalian apoA-I gene. To determine the sequences involved in the expression of the chicken apoA-I gene, plasmid constructs containing serial deletions of the 5' flanking region of the chicken apoA-I gene fused to the bacterial chloramphenicol acetyltransferase (CAT) gene were transfected in human hepatoma (HepG2), colon carcinoma (Caco2), epithelial (Hela), mouse embryonal fibroblast (NIH3T3) cells, and quail myoblasts (QMLA29). The shortest deletion construct, containing 60 bp of the 5' upstream region, was sufficient for maximal transcriptional activity in all cell lines tested. This region contains a short sequence (nucleotides -60 to -54) that is highly conserved in birds and mammals, and an Sp1 binding site. Although the sequence between nucleotides -232 and -101 of the 5' region of the chicken apoA-I gene is partially homologous to the hepatic cell-specific enhancer of the mammalian apoA-I gene (located between nucleotides -222 and -110 upstream of the human apoA-I gene transcription start site), this chicken sequence is transcriptionally inactive in HepG2 cells. These results suggest that differences in the cis-acting regulatory elements of the apoA-I gene play a fundamental role in determining the differences in the tissue-specific expression of this gene in avian and mammalian species.


Subject(s)
Apolipoprotein A-I/genetics , Genes , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Amino Acid Sequence , Animals , Base Sequence , Biological Evolution , Chickens , Cloning, Molecular , Female , Gene Expression Regulation , Humans , Male , Mice , Molecular Sequence Data , Quail , RNA, Messenger/analysis , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...