1
|
Cao X, Li Y. β 3-Adrenergic receptor regulates hepatic apolipoprotein A-I gene expression. J Clin Lipidol 2017; 11:1168-1176. [PMID: 28802864 DOI: 10.1016/j.jacl.2017.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/19/2017] [Accepted: 07/21/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND β3-adrenergic receptor (β3-AR) was shown to upregulate hepatic apolipoprotein A-I (apoA-I) expression and reverse atherosclerotic plaques in vivo experiments. However, the effect of β3-AR on apoA-I expression in vitro is unknown. The specific mechanism underlying β3-AR prevention of atherosclerosis is unclear. OBJECTIVE The present study was designed to investigate the molecular mechanism of β3-AR-mediated regulation of hepatic apoA-I gene expression. METHODS HepG2 cells were preincubated with/without a selective protein kinase A inhibitor (H-89) and then treated with a selective β3-AR agonist (BRL37344) or antagonist (SR59230A). The hepatic apoA-I expression was detected by reverse transcription real-time quantitative polymerase chain reaction and Western blot analysis. Enzyme-linked immunosorbent assay was used to evaluate the secretion of apoA-I. A recombinant plasmid containing the apoA-I promoter was constructed and transiently transfected into HepG2 cells, and dual-luciferase reporter assays were used to examine the activity of the apoA-I promoter. A chromatin immunoprecipitation polymerase chain reaction assay was used to evaluate binding activities of hepatocyte nuclear factor-4 (HNF-4), HNF-3, and early growth response protein-1. RESULTS β3-AR activation significantly upregulated apoA-I expression, promoted apoA-I secretion, and enhanced the activities of the apoA-I promoter, HNF-4, and HNF-3 in hepatocytes, whereas early growth response protein-1 was not affected. Moreover, protein kinase A inhibition partially suppressed the activation of the apoA-I promoter, HNF-4, and HNF-3 and almost completely blocked the upregulation of apoA-I expression induced by β3-AR. CONCLUSION β3-AR activation increased the activities of the apoA-I promoter, HNF-4, and HNF-3, which might account for the mechanism of β3-AR-mediated upregulation of hepatic apoA-I expression. β3-AR might exert an anti-atherosclerotic effect by upregulating hepatic apoA-I expression and promoting the cholesterol reverse transport process.
Collapse
Affiliation(s)
- Xiaojing Cao
- Emergency Department, Beijing Anzhen Hospital, Capital Medical University, ChaoYang District, Beijing, China
| | - Yanfang Li
- Emergency Department, Beijing Anzhen Hospital, Capital Medical University, ChaoYang District, Beijing, China.
| |
Collapse
|
2
|
Liu J, Hernandez-Ono A, Graham MJ, Galton VA, Ginsberg HN. Type 1 Deiodinase Regulates ApoA-I Gene Expression and ApoA-I Synthesis Independent of Thyroid Hormone Signaling. Arterioscler Thromb Vasc Biol 2016; 36:1356-66. [PMID: 27150392 DOI: 10.1161/atvbaha.116.307330] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/20/2016] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Plasma levels of high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A-I (ApoA-I) are reduced in individuals with defective insulin signaling. Initial studies using liver-specific insulin receptor (InsR) knockout mice identified reduced expression of type 1 deiodinase (Dio1) as a potentially novel link between defective hepatic insulin signaling and reduced expression of the ApoA-I gene. Our objective was to examine the regulation of ApoA-I expression by Dio1. APPROACH AND RESULTS Acute inactivation of InsR by adenoviral delivery of Cre recombinase to InsR floxed mice reduced HDL-C and expression of both ApoA-I and Dio1. Overexpression of Dio1 in InsR knockout mice restored HDL-C and ApoA-I levels and increased the expression of ApoA-I. Dio1 knockout mice had low expression of ApoA-I and reduced serum levels of HDL-C and ApoA-I. Treatment of C57BL/6J mice with antisense to Dio1 reduced ApoA-I mRNA, HDL-C, and serum ApoA-I. Hepatic 3,5,3'-triiodothyronine content was normal or elevated in InsR knockout mice or Dio1 knockout mice. Knockdown of either InsR or Dio1 by siRNA in HepG2 cells decreased the expression of ApoA-I and ApoA-I synthesis and secretion. siRNA knockdown of InsR or Dio1 decreased activity of a region of the ApoA-I promoter lacking thyroid hormone response elements (region B). Electrophoretic mobility shift assay demonstrated that reduced Dio1 expression decreased the binding of nuclear proteins to region B. CONCLUSIONS Reductions in Dio1 expression reduce the expression of ApoA-I in a 3,5,3'-triiodothyronine-/thyroid hormone response element-independent manner.
Collapse
Affiliation(s)
- Jing Liu
- From the Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY (J.L., A.H.-O., H.N.G.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G.); and Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH (V.A.G.).
| | - Antonio Hernandez-Ono
- From the Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY (J.L., A.H.-O., H.N.G.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G.); and Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH (V.A.G.)
| | - Mark J Graham
- From the Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY (J.L., A.H.-O., H.N.G.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G.); and Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH (V.A.G.)
| | - Valerie Anne Galton
- From the Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY (J.L., A.H.-O., H.N.G.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G.); and Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH (V.A.G.)
| | - Henry N Ginsberg
- From the Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY (J.L., A.H.-O., H.N.G.); Ionis Pharmaceuticals, Inc, Carlsbad, CA (M.J.G.); and Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Lebanon, NH (V.A.G.).
| |
Collapse
|
3
|
Kuang YL, Paulson KE, Lichtenstein AH, Matthan NR, Lamon-Fava S. Docosahexaenoic acid suppresses apolipoprotein A-I gene expression through hepatocyte nuclear factor-3β. Am J Clin Nutr 2011; 94:594-600. [PMID: 21653803 DOI: 10.3945/ajcn.111.012526] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Dietary fish-oil supplementation has been shown in human kinetic studies to lower the production rate of apolipoprotein (apo) A-I, the major protein component of HDL. The underlying mechanism responsible for this effect is not fully understood. OBJECTIVE We investigated the effect and the mechanism of action of the very-long-chain n-3 (omega-3) polyunsaturated fatty acid docosahexaenoic acid (DHA), relative to the saturated fatty acid palmitic acid (PA), on the hepatic expression of apo A-I in HepG2 cells. DESIGN HepG2 cells were treated with different doses of DHA and PA (0-200 μmol/L). mRNA expression levels of apo A-I were assessed by real-time polymerase chain reaction, and apo A-I protein concentrations were measured by immunoassay. DHA dose-dependently suppressed apo A-I mRNA levels and also lowered apo A-I protein concentrations in the media, with maximum effects at 200 μmol/L. This concentration of fatty acids was used in all subsequent experiments. RESULTS To elucidate the mechanism mediating the reduction in apo A-I expression by DHA, transfection experiments were conducted with plasmid constructs containing serial deletions of the apo A-I promoter. The DHA-responsive region was mapped to the -185 to -148 nucleotide region of the apo A-I promoter, which binds the hepatocyte nuclear factor (HNF)-3β. Nuclear extracts from cells treated with DHA or PA had a similar nuclear abundance of HNF-3β. However, electrophoresis mobility shift assays showed less binding of HNF-3β to the -180 to -140 sequence of the apo A-I promoter than did PA-treated cells. As shown by chromatin immunoprecipitation analysis, less HNF-3β was recruited to the apo A-I promoter in DHA-treated cells than in PA-treated cells, which supports the concept of an interference of DHA with the binding of HNF-3β to the apo A-I promoter. CONCLUSION These findings suggest that, in human hepatoma HepG2 cells, DHA inhibits the binding of HNF-3β to the apo A-I promoter, resulting in the repression of apo A-I promoter transactivity and thus a reduction in apo A-I expression.
Collapse
Affiliation(s)
- Yu-Lin Kuang
- Lipid Metabolism Laboratory, Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | | | | | | | | |
Collapse
|
4
|
Haas MJ, Horani MH, Wong NCW, Mooradian AD. Induction of the apolipoprotein AI promoter by Sp1 is repressed by saturated fatty acids. Metabolism 2004; 53:1342-8. [PMID: 15375792 DOI: 10.1016/j.metabol.2004.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Insulin induces transcription of the hepatic apolipoprotein AI (apo AI) gene by increasing Sp1 binding to the promoter. To determine the effect of fatty acids on this process, HepG2 cells cotransfected with the plasmid pAI.474.CAT containing the full-length apo AI promoter and the Sp1-expressing plasmid, pCMV-Sp1, were studied. Chloramphenicol acetyl transferase (CAT) activity (% acetylation) increased 1.98-fold in cells receiving the Sp1 expression construct relative to control cells (46.4% +/- 0.6% v 23.4% +/- 1.3%, P < .05). Treatment of cells with 3 saturated fatty acids, stearic, myristic, and palmitic acid, repressed the ability of exogenous Sp1 to induce apo AI reporter gene expression (15.2% +/- 1.7%, 22.5% +/- 0.3%, 22.9% +/- 0.1%, 23.5% +/- 0.8%, respectively, P < .05). Unsaturated fatty acids, oleic, linoleic, or linolenic acid had no effect on Sp1-mediated induction of the apo AI promoter. In the presence of the trans fatty acids, CAT activity in the Sp1-transfected cells was similar to control cells (16.7% +/- 3.3%, 19.3% +/- 0.5%, and 21.0% +/- 2.1% acetylation in cells exposed to elaidic acid, linolelaidic, or linolenelaidic acid, respectively). In cells treated with an equimolar mixture of oleic acid and stearic acid, apo AI promoter activity was suppressed in a manner similar to that observed in stearic acid-treated cells. Insulin (100 microU/mL) induced apo AI promoter activity 2.9-fold (22.4% +/- 1.7% v 7.8% +/- 2.4%, P < .05). However, in the presence of stearic acid, insulin was unable to induce apo AI promoter (6.3% +/- 1.6%). Stearic acid treatment did not alter Sp1-DNA binding as measured by gel shift analysis. Therefore, saturated fatty acids blunt Sp1 induction of apo AI promoter probably at a step beyond DNA binding.
Collapse
Affiliation(s)
- Michael J Haas
- Department of Internal Medicine, St Louis University School of Medicine, MO, USA
| | | | | | | |
Collapse
|
5
|
Abstract
The hepatitis C virus (HCV) protease genes (NS2/3 and NS3) were expressed in yeast with their natural substrates fused to a ligand-dependent transcriptional activator, the retinoic acid receptor (RARbeta). RARbeta can activate transcription in yeast cells in response to retinoic acids. We hypothesized that cis-cleavage at the NS2-3 or NS3-4A junctions by the appropriate HCV proteases would release RARbeta, thereby activating transcription of a reporter gene. Our results from Western blot analyses and reporter gene activation indicate that the wild-type NS2/3 and NS3 enzymes are catalytically active in yeast cells, whereas mutations in the catalytic domain of NS2(C993V) and NS3(S1165A) lead to inactive enzymes. We conclude that HCV NS2/3 and NS3 protease activities can be reconstituted in yeast.
Collapse
Affiliation(s)
- P Mak
- Molecular Biology and Virology Section, Wyeth-Ayerst Research, 401 N. Middletown Road, Pearl River, NY 10965, USA.
| | | | | | | |
Collapse
|
6
|
Sladek FM, Ruse MD, Nepomuceno L, Huang SM, Stallcup MR. Modulation of transcriptional activation and coactivator interaction by a splicing variation in the F domain of nuclear receptor hepatocyte nuclear factor 4alpha1. Mol Cell Biol 1999; 19:6509-22. [PMID: 10490591 PMCID: PMC84621 DOI: 10.1128/mcb.19.10.6509] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/1998] [Accepted: 06/25/1999] [Indexed: 12/14/2022] Open
Abstract
Transcription factors, such as nuclear receptors, often exist in various forms that are generated by highly conserved splicing events. Whereas the functional significance of these splicing variants is often not known, it is known that nuclear receptors activate transcription through interaction with coactivators. The parameters, other than ligands, that might modulate those interactions, however, are not well characterized, nor is the role of splicing variants. In this study, transient transfection, yeast two-hybrid, and GST pulldown assays are used to show not only that nuclear receptor hepatocyte nuclear factor 4 alpha1 (HNF4alpha1, NR2A1) interacts with GRIP1, and other coactivators, in the absence of ligand but also that the uncommonly large F domain in the C terminus of the receptor inhibits that interaction. In vitro, the F domain was found to obscure an AF-2-independent binding site for GRIP1 that did not map to nuclear receptor boxes II or III. The results also show that a natural splicing variant containing a 10-amino-acid insert in the middle of the F domain (HNF4alpha2) abrogates that inhibition in vivo and in vitro. A series of protease digestion assays indicates that there may be structural differences between HNF4alpha1 and HNF4alpha2 in the F domain as well as in the ligand binding domain (LBD). The data also suggest that there is a direct physical contact between the F domain and the LBD of HNF4alpha1 and -alpha2 and that that contact is different in the HNF4alpha1 and HNF4alpha2 isoforms. Finally, we propose a model in which the F domain of HNF4alpha1 acts as a negative regulatory region for transactivation and in which the alpha2 insert ameliorates the negative effect of the F domain. A conserved repressor sequence in the F domains of HNF4alpha1 and -alpha2 suggests that this model may be relevant to other nuclear receptors as well.
Collapse
Affiliation(s)
- F M Sladek
- Environmental Toxicology, University of California, Riverside, California 92521, USA.
| | | | | | | | | |
Collapse
|
7
|
Fukai K, Takada S, Yokosuka O, Saisho H, Omata M, Koike K. Characterization of a specific region in the hepatitis B virus enhancer I for the efficient expression of X gene in the hepatic cell. Virology 1997; 236:279-87. [PMID: 9325235 DOI: 10.1006/viro.1997.8750] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Hepatitis B virus (HBV) enhancer I has been shown to consist of several cis-acting sequences for the HBV gene expression efficiently in certain types of cells. Transcriptional regulation of HBV X gene mediated by enhancer I might be one of the mechanisms by which HBV obtains hepatotropism. By mutagenesis analysis of enhancer I function in the enhancer I/X gene promoter complex, we characterized a specific transcriptional regulatory region (designated as a LSR element, nt 989-1030) of enhancer I for the X gene promoter by means of the transient transfection technique using hepatic and nonhepatic cells. Based on the analysis of protein factors interacting with the LSR element, liver-enriched transcriptional factors, HNF3 and HNF4 or retinoid X receptor alpha (RXR alpha), are probably implicated in the activity of enhancer I for the efficient expression of X gene through their interaction with the LSR element in the hepatic cell. Furthermore, the isolated LSR element was demonstrated to function alone as a specific cis-acting element and to be able to activate transcription from the X gene promoter efficiently in the hepatic cell in an orientation-independent manner.
Collapse
Affiliation(s)
- K Fukai
- Department of Gene Research, Cancer Institute, JFCR, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
8
|
Abstract
Transcriptional activation of the Drosophila ecdysone receptor (EcR) was studied in yeast cells, which carry a reporter plasmid containing the ecdysone response element in the absence or presence of its heterodimeric partners, ultraspiracle protein (USP) or human retinoid X receptor (RXRalpha). High constitutive transcriptional activation was detected in the yeast strain expressing EcR, but not USP or RXRalpha in the absence of ponasterone or muristerone A. Incubation of these ligands with yeast cells coexpressing EcR and USP or RXRalpha did not enhance the constitutive transcriptional activity. However, specific ligand binding using [3H]ponasterone A as a radioactive ligand was detected only in yeast extracts prepared from the yeast strain coexpressing EcR and USP, but not from yeast strains expressing only EcR or USP. The ligand binding characteristics of the EcR/USP complexes were similar to those reported in an insect cell line with a Kd value of 1.8 nM for [3H]ponasterone A. These data are in contrast to mammalian cell transfection studies, and indicate that the EcR is the only member of the nuclear receptor superfamily of ligand-activated transcription factors which functions as a constitutive transcriptional activator in yeast, although the EcR/USP complexes exhibit normal ligand binding properties.
Collapse
Affiliation(s)
- F Dela Cruz
- American Cyanamid Company, Agricultural Research Division, Princeton, NJ 08540, USA
| | | |
Collapse
|
9
|
Fraser JD, Keller D, Martinez V, Santiso-Mere D, Straney R, Briggs MR. Utilization of recombinant adenovirus and dominant negative mutants to characterize hepatocyte nuclear factor 4-regulated apolipoprotein AI and CIII expression. J Biol Chem 1997; 272:13892-8. [PMID: 9153249 DOI: 10.1074/jbc.272.21.13892] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Using recombinant adenoviral vectors and a dominant negative mutant of HNF-4, we have examined the contribution of hepatocyte nuclear factor 4 (HNF-4) to endogenous apolipoprotein AI and CIII mRNA expression. Overexpression of HNF-4 leads to a 7.4-fold increase in apolipoprotein CIII expression, while infection with the dominant negative mutant of HNF-4 reduces the level of apolipoprotein CIII mRNA by 80%, demonstrating that endogenous HNF-4 is necessary for apolipoprotein CIII expression. Experiments using the hepatoma cell lines, HepG2 and Hep3B, indicate that HNF-4 is also involved in the regulation of apolipoprotein AI expression in these lines. However, the effect of HNF-4 on apolipoprotein AI expression is much more dramatic in cell lines derived from intestinal epithelium. Infection of the intestinal-derived cell line IEC-6 with the HNF-4 adenovirus resulted in a greater than 20-fold increase in the level of apolipoprotein AI mRNA. These results indicate that HNF-4 does regulate apolipoprotein AI and CIII mRNA expression and suggest that HNF-4 is critical for intestinal apolipoprotein AI expression.
Collapse
MESH Headings
- Adenoviridae
- Apolipoprotein A-I/genetics
- Apolipoprotein A-I/metabolism
- Apolipoprotein C-III
- Apolipoproteins C/genetics
- Apolipoproteins C/metabolism
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors
- DNA, Complementary/chemistry
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Genetic Vectors
- Hepatocyte Nuclear Factor 4
- Humans
- In Situ Hybridization
- Mutagenesis, Site-Directed
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- Promoter Regions, Genetic
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic/drug effects
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- J D Fraser
- Ligand Pharmaceuticals Inc., San Diego, California 92121, USA.
| | | | | | | | | | | |
Collapse
|
10
|
Giffin W, Kwast-Welfeld J, Rodda DJ, Préfontaine GG, Traykova-Andonova M, Zhang Y, Weigel NL, Lefebvre YA, Haché RJ. Sequence-specific DNA binding and transcription factor phosphorylation by Ku Autoantigen/DNA-dependent protein kinase. Phosphorylation of Ser-527 of the rat glucocorticoid receptor. J Biol Chem 1997; 272:5647-58. [PMID: 9038175 DOI: 10.1074/jbc.272.9.5647] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
NRE1 is a DNA sequence element through which Ku antigen/DNA-dependent protein kinase (DNA-PK) catalytic subunit represses the induction of mouse mammary tumor virus transcription by glucocorticoids. Although Ku is an avid binder of DNA ends and has the ability to translocate along DNA, we report that direct sequence-specific Ku binding occurs with higher affinity (Kd = 0.84 +/- 0.24 nM) than DNA end binding. Comparison of Ku binding to several sequences over which Ku can accumulate revealed two classes of sequence. Sequences with similarity to NRE1 competed efficiently for NRE1 binding. Conversely, sequences lacking similarity to NRE1 competed poorly for Ku and were not recognized in the absence of DNA ends. Phosphorylation of glucocorticoid receptor (GR) fusion proteins by DNA-PK reflected Ku DNA-binding preferences and demonstrated that co-localization of GR with DNA-PK on DNA in cis was critical for efficient phosphorylation. Phosphorylation of the GR fusion protein by DNA-PK mapped to a single site, Ser-527. This site occurs adjacent the GR nuclear localization sequence between the DNA and ligand binding domains of GR, and thus its phosphorylation, if confirmed, has the potential to affect receptor function in vivo.
Collapse
Affiliation(s)
- W Giffin
- Department of Medicine, University of Ottawa, Loeb Medical Research Institute, Ottawa Civic Hospital, Ottawa, Ontario, Canada K1Y 4E9
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Taylor DG, Haubenwallner S, Leff T. Characterization of a dominant negative mutant form of the HNF-4 orphan receptor. Nucleic Acids Res 1996; 24:2930-5. [PMID: 8760876 PMCID: PMC146029 DOI: 10.1093/nar/24.15.2930] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The HNF-4 orphan receptor is a member of the nuclear receptor family of transcription factors and a major regulator of genes involved in carbohydrate and lipid metabolism. As an initial step in characterizing the role of HNF-4 in the regulation of metabolism, we have generated a dominant negative form of HNF-4 (DN-HNF-4) that contains a defective DNA-binding domain. In gel mobility shift assays, DN-HNF-4 did not bind an oligonucleotide probe representing an essential HNF-4 binding site, C3P contained in the human apo CIII promoter, but did prevent the binding of two recombinant isoforms, HNF-4alpha1 and HNF-4alpha2, as well as naturally-occurring HNF-4. DN-HNF-4 had no effect on the binding of PPARgamma-RXRalpha heterodimers to a PPAR response element. In transfected HepG2 cells, DN-HNF-4 dramatically reduced constitutive transcriptional activity of the human apo CIII promoter and abolished the positive transcriptional activity caused by plasmids expressing either isoform of HNF-4. These results indicate that DN-HNF-4 is a selective dominant negative mutant which forms defective heterodimers with wild-type HNF-4, thereby preventing DNA binding and subsequent transcriptional activation by HNF-4.
Collapse
Affiliation(s)
- D G Taylor
- Department of Biotechnology, Parke-Davis Pharmaceutical Research, Ann Arbor, MI 48105, USA
| | | | | |
Collapse
|
12
|
Harnish DC, Malik S, Kilbourne E, Costa R, Karathanasis SK. Control of apolipoprotein AI gene expression through synergistic interactions between hepatocyte nuclear factors 3 and 4. J Biol Chem 1996; 271:13621-8. [PMID: 8662915 DOI: 10.1074/jbc.271.23.13621] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
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.
Collapse
Affiliation(s)
- D C Harnish
- Department of Cardiovascular Molecular Biology, Lederle Laboratories, Pearl River, New York 10965, USA
| | | | | | | | | |
Collapse
|
13
|
Malik S, Karathanasis SK. TFIIB-directed transcriptional activation by the orphan nuclear receptor hepatocyte nuclear factor 4. Mol Cell Biol 1996; 16:1824-31. [PMID: 8657158 PMCID: PMC231169 DOI: 10.1128/mcb.16.4.1824] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
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.
Collapse
Affiliation(s)
- S Malik
- Department of Cardiovascular Molecular Biology, Lederle Laboratories, Pearl River, New York 10965, USA
| | | |
Collapse
|
14
|
Salerno AJ, He Z, Goos-Nilsson A, Ahola H, Mak P. Differential transcriptional regulation of the apoAI gene by retinoic acid receptor homo- and heterodimers in yeast. Nucleic Acids Res 1996; 24:566-72. [PMID: 8604295 PMCID: PMC145681 DOI: 10.1093/nar/24.4.566] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Several members of the nuclear receptor superfamily including RXR (retinoid X receptor) bind to a specific retinoic acid response element (site A) of the apoAI promoter. However, transcriptional activation of the apoAI gene by different homo- and heterodimeric forms of RXR or RAR (retinoic acid receptors) cannot be evaluated in mammalian cells, which contain endogenous RXR or RAR. In order to circumvent this limitation, we assessed the DNA-binding activities and transcriptional activation of different homo- and heterodimers of these receptors in yeast. Electrophoretic mobility shift assays (EMSA) demonstrated that yeast expressed RARalpha does not bind to site A of the apoAl promoter, whereas binding of RARbeta to site A is ligand-dependent. Both RARalpha and RARbeta form heterodimers with RXRalpha and bind to site A with high affinity. These DNA-binding studies correlate with the transcriptional data, which indicated that RARbeta but not RARalpha activates transcription from site A in response equally well to 9-cis and all-trans retinoic acids. 9-cis RA is a more potent ligand than all-trans RA to activate transcription via RXR/RAR heterodimers. We conclude that this yeast expression system is a useful tool to elucidate the 'transactivation code' for apoAl site A via specific combinations of different homo and heterodimeric versions of RXR and RAR.
Collapse
Affiliation(s)
- A J Salerno
- Wyeth-Ayerst Research, Lederle Laboratories, Pearl River, NY 10965, USA
| | | | | | | | | |
Collapse
|