Vascular endothelial growth factor D has recently been linked to the control of lymphangiogenesis and lymphatic metastasis. The molecular determinants regulating vegf-D gene transcription, however, have not yet been identified. After isolation of 2 kb of 5′-flanking DNA of the human vegf-D gene, we identified a novel, atypical direct repeat (DR) element consisting of a consensus half-site (AGGTCA) at −125/−119 and a degenerated DR half-site (ATGTTA) at −99/−94 as sufficient and necessary for vegf-D transcription. The vegf-D DR element is bound and activated by the orphan receptors hepatocyte nuclear factor 4α (HNF-4α) and chicken ovalbumin upstream promoter transcription factor (COUP-TF)-1/COUP-TF2. Additionally, chromatin immunoprecipitation assays identified transcriptional coactivators cyclic AMP–responsive element binding protein–binding protein and glucocorticoid receptor interacting protein 1 at the vegf-D DR element and functional assays confirmed their stimulatory effect on the vegf-D promoter. Histone deacetylase inhibition by trichostatin A led to accumulation of acetylated histones H3/H4 at the vegf-D promoter, up-regulation of vegf-D mRNA levels, and transactivation of vegf-D promoter reporter gene constructs in cancer cell lines. This study for the first time describes the molecular determinants in cis and trans controlling vegf-D gene transcription and identifies interaction of HNF-4α and COUP-TF1/COUP-TF2 with a proximal, atypical DR element as indispensable for vegf-D transcription. Moreover, our findings suggest that epigenetic control of histone acetylation represents an important determinant of vegf-D gene expression in cancer cells. These results provide novel insights into the molecular machinery controlling vegf-D gene expression and may add to a better understanding of the regulation of lymphangiogenesis in vascular development and cancer. [Cancer Res 2008;68(2):457–66]

Angiogenesis, the formation of new blood vessels from endothelial precursors, is a prerequisite for growth and dissemination of solid malignancies (1), and the vascular endothelial growth factor (VEGF) superfamily of endothelial growth factors has been identified to critically influence tumor-related angiogenesis (1). Recently, it was shown that basic mechanisms of hemangiogenesis also apply to the lymphatic system and that VEGF-C and its close homologue VEGF-D are intimately involved in the regulation of lymphangiogenesis and lymphatic tumor spread (2). The lymphangiogenic effects of VEGF-C and VEGF-D are essentially mediated via activation of VEGF receptor (VEGFR)-3, which is primarily expressed by lymphatic vessels but can also be found on angiogenic endothelial cells of tumor blood vessels (2). In line with their proposed role in cancer pathobiology, the expression of VEGF-C, VEGF-D, and/or VEGFR-3 has been shown to correlate with lymphatic spread, tissue invasion, and/or poor prognosis in solid malignancies (3) including gastric cancer (4). In other studies, however, conflicting observations were made (5, 6) indicating that the role of the VEGF-C/VEGF-D/VEGFR-3 system in cancer is complex and may differ between malignancies.

Despite the accumulating evidence supporting the importance of VEGF-C and VEGF-D for lymphangiogenesis and cancer spread, the current knowledge on molecular determinants and regulating pathways is very limited. In vitro studies showed that proinflammatory cytokines, including interleukin-1β and tumor necrosis factor (TNFα), as well as phorbol esters and growth factors can stimulate vegf-C expression (710), whereas these stimulators had no effect on vegf-D (710). Although resistant to various extracellular stimulators, vegf-D mRNA levels were found to be stimulated by a cell-cell contact–triggered mechanism involving cadherin-11 (8). In another report, it was shown that a β-catenin–dependent mechanism can down-regulate vegf-D expression by influencing mRNA stability (11). Because the human vegf-D cDNA was initially isolated using an activator protein (AP-1) overexpression strategy (12), it was suggested that AP-1 may be directly involved in regulation of vegf-D gene expression. Because the transcription factors and regulatory promoter elements controlling vegf-D gene expression have not yet been identified, the role of AP-1 in regulation of VEGF-D, however, remains hypothetical.

Orphan receptor hepatocyte nuclear factor 4α (HNF-4α) represents a key regulator of liver specific gene expression and influences important metabolic pathways regulating glucose and lipid homeostasis (13, 14). Additionally, hepatocyte-specific deletion of hnf-4α in mice leads to severely altered hepatocyte differentiation as well as distorted organization of sinusoidal endothelium (15). Recent studies showed that at some gene promoters, HNF-4α functionally cooperates with orphan receptors chicken ovalbumin upstream promoter transcription factor (COUP-TF)-1 and COUP-TF2 (16, 17). Both COUP-TFs display high (96–98%) homology on the protein level and influence key biological processes (18, 19): homozygous disruption of the coup-tf 1 allele in mice leads to severe defects in the central and peripheral nervous system, whereas loss of coup-tf2 results in disturbed angiogenesis and defective development of the murine heart leading to embryonic death (18, 19). Similar to HNF-4α, target genes executing the effects of COUP-TFs in context with endothelial morphogenesis and angiogenesis have not yet been described.

To characterize the molecular determinants of vegf-D gene transcription, we cloned and sequenced the human vegf-D gene promoter and found that a novel, atypical direct repeat (DR) element located in the proximal human vegf-D gene promoter is essential for vegf-D gene expression. This element is bound and activated by a complex comprising the orphan receptors HNF-4α and COUP-TF1/COUP-TF2 as well as transcriptional coactivators glucocorticoid receptor interacting protein 1 (GRIP-1) and cyclic AMP–responsive element binding protein–binding protein (CBP) to drive vegf-D gene transcription. Moreover, our results strongly suggest that the vegf-D gene is under control of epigenetic mechanisms. These results for the first time uncover the molecular components controlling vegf-D transcription and thereby add to our current understanding of mechanisms regulating the process of lymphangiogenesis.

Cell culture and transfections. Cell lines were grown in DMEM (Invitrogen) supplemented with 2 mmol/L glutamine, 100 units/mL penicillin, 100 μg/mL streptomycin, and 10% bovine calf serum in a humified atmosphere (Sanyo). Transient transfections were carried out as previously described (20) and luciferase activities were detected using the Dual-Luciferase-Reporter Assay System (Promega). In a set of experiments, cells transfected with expression constructs encoding peroxisome proliferator–activated receptor (PPAR)-γ, retinoic acid receptor (RAR)-γ, and/or retinoid X receptor (RXR)-α, together with reporter gene construct vegf-D(−135/−81), were treated with prostaglandin J2, 9-cis retinoic acid, or all-trans retinoic acid at indicated concentrations for 24 or 48 h. The same treatment was applied to transfectants receiving empty expression constructs along with vegf-D(−135/−81) (negative controls). The functionality of nuclear hormone receptor agonists and expression constructs was confirmed in a series of transfections using the reporter construct 5× HRE-Luc as a readout (data not shown). Incubations were done in triplicates and results were normalized for transfection efficiency and calculated as mean ± SE. Values were expressed as fold increases of control value. Statistical significances were calculated using Student's t test (*, P < 0.05; **, P < 0.01; ***, P < 0.001).

Immunofluorescence in cell lines. Immunofluorescence in cell lines was done as previously described (4). Briefly, cells were grown on chamber slides, fixed, and permeabilized with saponin solution (Sigma). Following incubation with polyclonal VEGF-D antibody (C-18, Santa Cruz Biotechnology), stainings were visualized with an antigoat secondary antibody labeled with Texas red (Vector Laboratories).

Quantitative real-time reverse transcription-PCR. Quantitative reverse transcription-PCR (RT-PCR) analysis was done as previously described (4). Briefly, total RNA was isolated from cell lines or tissues using the TRIzol reagent (Invitrogen) and reversely transcribed using oligo-dT primers and SuperScript II polymerase (Invitrogen). cDNAs generated from 50 ng of total RNA were used for quantification using the TaqMan-Universal or SYBR Green PCR Master Mix (Applied Biosystems) on an ABI PRISM 7700 Sequence Detection System. Primers and probes were applied as listed in Supplementary Table S1.

DNA constructs and reporter plasmids. Using the Genome Walker Kit (Clontech) and two gene-specific primers (VEGF-D/GSP1, 5′-gcagttgacatcaggcagctagag-3′, and VEGF-D/GSP2, 5′-caaaaggcattctccagaaggaaag-3′) located within the first exon of the vegf-D gene (NM_004469), a genomic fragment comprising 2,011 nucleotides of the 5′ flanking region of the human vegf-D gene as well as 110 nucleotides of noncoding exon 1 was isolated (GenBank accession no. AY874421). Subcloning of this DNA fragment into the promoterless luciferase reporter gene vector pGL3 (Promega) yielded the promoter-reporter gene construct vegf-D(−2011/+110). Using vegf-D(−2011/+110) as a template, 5′-deletion constructs were generated using a common 3′ primer and different 5′ primers (Supplementary Table S2) as previously described (20). To study vegf-D promoter elements in a heterologous promoter system, vector pT81-Luc, in which the firefly luciferase gene is under control of the enhancerless herpes simplex thymidine kinase viral promoter, was used as described earlier (20). Site-directed mutagenesis was done with the QuikChange kit (Stratagene). To increase efficiency and reliability of mutagenesis, construct vegf-D(−264/+110) was used as a template instead of a plasmid containing a longer fragment of vegf-D 5′ sequence. Primers for mutagenesis were applied as listed in Supplementary Table S2. Constructs encoding COUP-TF1, COUP-TF2, HNF-4α, its mutant HNF-4α-DBD (DNA binding domain deletion Δ49–115), or RAR isoforms α, β, and γ, as well as RXRα, have been described before (2123). Similarly, the construct encoding wild-type PPARγ was previously used (24).

Electrophoretic mobility shift assays. Electrophoretic mobility shift assays (EMSA) were carried out as previously described (20). In brief, nuclear protein extracts (5 μg) were incubated with [γ-32P]ATP–radiolabeled double-stranded oligonucleotides (Supplementary Table S3). For competition experiments, nuclear extracts were incubated with 100-fold molar excess of double-stranded, unlabeled competitor oligonucleotides (Supplementary Table S3). For supershift experiments, nuclear extracts were incubated with antibodies recognizing PPARγ, RARγ, RXRα (Santa Cruz Biotechnology), HNF-4α (25), or COUP-TF (26).

Chromatin immunoprecipitation. Chromatin immunoprecipitation assays were carried out as previously described (27) with 5 μg of anti–acetylated H3 or H4 antibodies (Upstate Biotechnologies) and rabbit pre-immunosera. Quantitative amplification using the qPCR SYBR Green Core Kit (Eurogentec) was done according to the manufacturer's conditions and results were normalized to input controls.

Trichostatin A and 5-aza-2-deoxycytidine treatment of cells. Exposure of cells to indicated concentrations of trichostatin A (TSA) and/or 5 μmol/L 5-aza-2-deoxycytidine (Sigma-Aldrich) was done for 24 and/or 72 h. Control incubations received the appropriate solvent for identical time periods. In experiments examining the influence of TSA on vegf-D promoter-reporter gene constructs, transfected cells were exposed to 100 ng/mL TSA or vehicle for 24 h. To investigate the influence of hypoxia on TSA-induced effects, cells were treated with various concentrations of TSA or solvent and were immediately afterwards transferred to normoxic or hypoxic conditions for 24 h.

vegf-D gene expression and promoter activity in cancer cell lines. Using a specific anti–VEGF-D antibody, we detected a paranuclear granular staining pattern in all cell lines investigated (Fig. 1A). In line with staining results, real-time PCR analysis revealed low levels of vegf-D mRNA in AGS, KATO-III, and MKN-28 cells, whereas MKN-45 cells showed 5- to 8-fold higher amounts of vegf-D mRNA (Fig. 1B). Compared with the empty pGL3 construct, transfection of vegf-D(−2011/+110) into MKN-45 cells produced a 6-fold increase in luciferase activity (Fig. 1C). In contrast, the activity of vegf-D(−2011/+110) in other gastric cancer cell lines was close to pGL3 control.

Figure 1.

vegf-D gene expression and promoter activity in gastric cell lines. A, immunofluorescence staining of gastric carcinoma cell lines AGS, KATO-III, MKN-28, and MKN-45 with a VEGF-D–specific antibody. Insets, negative control stainings in absence of primary antibody. B, real-time RT-PCR quantification of total RNA of gastric carcinoma cell lines with vegf-D–specific primers. Expression levels are depicted relative to MKN-45 cells. Summary of three independent experiments. C, vegf-D gene promoter activity in gastric cancer cell lines. Cells were transiently transfected with the pGL3-based vegf-D reporter gene construct vegf-D(−2011/+110), which comprises nucleotides −2011/+110 of the human vegf-D gene. Cells were harvested 24 h after transfection and assayed for luciferase activity. Results were normalized for transfection efficiency by cotransfection of equal amounts of a renilla luciferase reporter gene construct and data are shown as fold increase of activities obtained with empty vector pGL3.

Figure 1.

vegf-D gene expression and promoter activity in gastric cell lines. A, immunofluorescence staining of gastric carcinoma cell lines AGS, KATO-III, MKN-28, and MKN-45 with a VEGF-D–specific antibody. Insets, negative control stainings in absence of primary antibody. B, real-time RT-PCR quantification of total RNA of gastric carcinoma cell lines with vegf-D–specific primers. Expression levels are depicted relative to MKN-45 cells. Summary of three independent experiments. C, vegf-D gene promoter activity in gastric cancer cell lines. Cells were transiently transfected with the pGL3-based vegf-D reporter gene construct vegf-D(−2011/+110), which comprises nucleotides −2011/+110 of the human vegf-D gene. Cells were harvested 24 h after transfection and assayed for luciferase activity. Results were normalized for transfection efficiency by cotransfection of equal amounts of a renilla luciferase reporter gene construct and data are shown as fold increase of activities obtained with empty vector pGL3.

Close modal

A DR element between −138 and −79 is indispensable for vegf-D promoter activity. To identify the basal enhancer element(s) controlling vegf-D gene promoter activity, functional 5′ deletion analysis was done in MKN-45 cells. Whereas progressive deletion from −2011 to −141 had no influence on vegf-D promoter activity, loss of additional 20 nucleotides [vegf-D(−122/+110)] reduced promoter activity by 50% (Fig. 2A). Shortening of the vegf-D 5′ sequence by additional 40 bp [construct vegf-D(−83/+110)] abolished vegf-D transcription (Fig. 2A). Because these results indicated that vegf-D −141 to −83 contains essential regulatory elements, this sequence was used as a radiolabeled probe (probe A) in EMSAs. In parallel, a single copy of the fragment was subcloned into the heterologous, enhancerless luciferase reporter construct pT81 and analyzed in functional transfection studies. For further analysis, the vegf-D −138/−79 fragment was split into a 5′ [−138 to −109 (probe B)] and a 3′ fragment [−108 to −79 (probe C)]. Computer-assisted sequence analysis (28) revealed the presence of two consensus binding sites for GATA transcription factors and a consensus DR half-site at −119 to −125 within the −138/−79 sequence (Fig. 2B). In EMSAs, incubation of probe A with nuclear extracts yielded two typical nuclear complexes (Fig. 2C, lane 1). The lower complex (complex II) bound to probe A was also formed at probe B (−138/−109; Fig. 2C, lane 2), whereas complex I was not found with probe B but was clearly detectable with probe C (Fig. 2C, lane 3). In functional transfection assays, vegf-D −138/−79 showed a 10- to 15-fold higher transcriptional activity when compared with empty pT81, showing that this sequence confers true enhancer characteristics (Fig. 2D). In contrast, although showing unaffected binding of complex II or complex I, the activity conferred by vegf-D −135/−109 and vegf-D −109/−81 in pT81 was drastically reduced (Fig. 2D), suggesting that enhancer elements exist within the −138/−79 region that interact in a cooperative fashion.

Figure 2.

The vegf-D −138 to −79 element is necessary and sufficient for regulation of basal transcriptional activity of the vegf-D gene. A, MKN-45 cells were transiently transfected with a series of vegf-D 5′ deletion constructs and luciferase reporter gene activity was determined. Columns, mean of three separate experiments; bars, SE. **, P < 0.01; ***, P < 0.001. B, nucleotide sequence of the −138 to −79 vegf-D element and derived fragments B and C as used in transcription and EMSA studies. C, EMSA analysis of nuclear proteins binding to the −138 to −79 vegf-D sequence. Crude nuclear protein extracts were prepared from MKN-45 cells and incubated with 32P-labeled double-stranded DNA probes representing the entire −138/−79 vegf-D sequence (A) or its fragment B or C. Complexes were resolved on nondenaturating polyacrylamide gels and data shown represent a typical result obtained from a series of three independent experiments. D, the −135/−81 vegf-D sequence confers transcriptional activity to the heterologous promoter system pT81. The entire −135/−81 vegf-D sequence or its fragments vegf-D(−135/−109) and vegf-D(−109/−81) were subcloned into the heterologous promoter system pT81-Luc and transiently transfected into MKN-45 cells. **, P < 0.01, between minimal promoter fragment and its half-sites.

Figure 2.

The vegf-D −138 to −79 element is necessary and sufficient for regulation of basal transcriptional activity of the vegf-D gene. A, MKN-45 cells were transiently transfected with a series of vegf-D 5′ deletion constructs and luciferase reporter gene activity was determined. Columns, mean of three separate experiments; bars, SE. **, P < 0.01; ***, P < 0.001. B, nucleotide sequence of the −138 to −79 vegf-D element and derived fragments B and C as used in transcription and EMSA studies. C, EMSA analysis of nuclear proteins binding to the −138 to −79 vegf-D sequence. Crude nuclear protein extracts were prepared from MKN-45 cells and incubated with 32P-labeled double-stranded DNA probes representing the entire −138/−79 vegf-D sequence (A) or its fragment B or C. Complexes were resolved on nondenaturating polyacrylamide gels and data shown represent a typical result obtained from a series of three independent experiments. D, the −135/−81 vegf-D sequence confers transcriptional activity to the heterologous promoter system pT81. The entire −135/−81 vegf-D sequence or its fragments vegf-D(−135/−109) and vegf-D(−109/−81) were subcloned into the heterologous promoter system pT81-Luc and transiently transfected into MKN-45 cells. **, P < 0.01, between minimal promoter fragment and its half-sites.

Close modal

After confirming the enhancer characteristics of vegf-D(−135/−79), this sequence was investigated by site-directed mutagenesis in context of a larger vegf-D fragment [vegf-D(-264/+110)]. Whereas individual or simultaneous disruption of GATA sites had no significant influence, mutation of the 5′ DR half-site at −125/−119 (mutant ΔDR5′) reduced vegf-D promoter activity by 50% (Fig. 3A). Moreover, mutation of the sequence vegf-D −99/−94, which resembles characteristics of an degenerated DR half-site (mutant ΔdDR3′), blunted vegf-D promoter activity, whereas simultaneous mutation of −125/−119 and −99/−94 abolished vegf-D transcription (Fig. 3A). EMSA competition experiments confirmed that the mutations used in transfection assays were sufficient to abrogate formation of complex I at the 3′ dDR element (competitor ΔdDR3′) as well as formation of complex II at the 5′ DR half-site (competitor ΔDR5; Fig. 3B).

Figure 3.

Two DR motifs are essential for transactivation of the human vegf-D promoter and binding of nuclear receptors to the minimal vegf-D promoter sequence. A, DR motifs and/or the putative GATA binding sites were point-mutated within the context of vegf-D(−264/+110) and used in transient transfection studies using MKN-45 cells. Results were calculated as luciferase activities relative to empty vector pGL3-Luc. Columns, mean obtained from three separate experiments; bars, SE. *, P < 0.05; **, P < 0.01; ***, P < 0.001. B, EMSA competition analysis of nuclear protein binding to DR element and mutants. Oligonucleotides representing the 5′ or 3′ halves of −138 /−79 vegf-D sequence or mutated DR elements were used as competitors (200-fold molar excess) in EMSA studies using 32P-labeled −138/−79 vegf-D sequence as a probe. C, EMSA competition analysis of nuclear proteins binding to the −138/−79 vegf-D sequence. Using 32P-labeled −138/−79 vegf-D nucleotides as a radiolabeled probe, competition studies with consensus binding sites for nuclear hormone receptors were carried out. Unlabeled nucleotides representing consensus PPAR, RAR, as well as RXR binding motifs were applied in 200-fold molar excess. D, EMSA supershift analysis of nuclear proteins binding to the −138/−79 vegf-D element. To identify transcription factors binding to the −138/−79 vegf-D sequence, nuclear proteins were prepared from MKN-45 cells and incubated with 32P-labeled oligonucleotides representing vegf-D −139/−79 as well as specific antibodies as indicated (top). In addition to vegf-D −139/−79, the 5′ (probe B) or vegf-D −108/−79 3′ half (probe C) was used as radiolabeled probe.

Figure 3.

Two DR motifs are essential for transactivation of the human vegf-D promoter and binding of nuclear receptors to the minimal vegf-D promoter sequence. A, DR motifs and/or the putative GATA binding sites were point-mutated within the context of vegf-D(−264/+110) and used in transient transfection studies using MKN-45 cells. Results were calculated as luciferase activities relative to empty vector pGL3-Luc. Columns, mean obtained from three separate experiments; bars, SE. *, P < 0.05; **, P < 0.01; ***, P < 0.001. B, EMSA competition analysis of nuclear protein binding to DR element and mutants. Oligonucleotides representing the 5′ or 3′ halves of −138 /−79 vegf-D sequence or mutated DR elements were used as competitors (200-fold molar excess) in EMSA studies using 32P-labeled −138/−79 vegf-D sequence as a probe. C, EMSA competition analysis of nuclear proteins binding to the −138/−79 vegf-D sequence. Using 32P-labeled −138/−79 vegf-D nucleotides as a radiolabeled probe, competition studies with consensus binding sites for nuclear hormone receptors were carried out. Unlabeled nucleotides representing consensus PPAR, RAR, as well as RXR binding motifs were applied in 200-fold molar excess. D, EMSA supershift analysis of nuclear proteins binding to the −138/−79 vegf-D element. To identify transcription factors binding to the −138/−79 vegf-D sequence, nuclear proteins were prepared from MKN-45 cells and incubated with 32P-labeled oligonucleotides representing vegf-D −139/−79 as well as specific antibodies as indicated (top). In addition to vegf-D −139/−79, the 5′ (probe B) or vegf-D −108/−79 3′ half (probe C) was used as radiolabeled probe.

Close modal

HNF-4α and COUP-TF transcription factors bind to the vegf-D DR element. DR elements are typically bound by nuclear receptors and/or orphan receptors (29). To evaluate whether such proteins interact with the vegf-D DR element, EMSA competition experiments using consensus binding sequences for PPAR, RAR, and RXR as unlabeled competitors and vegf-D −138/−79 as radiolabeled probe were conducted. Application of PPAR and RXR competitors abolished formation of complex II at the vegf-D −138/−79 probe (Fig. 3C, lanes 2 and 6), whereas addition RAR oligonucleotides disrupted binding of both complex I and complex II (Fig. 3C, lane 4). The lack of effectiveness observed for mutated competitors confirmed the specificity of results obtained with wild-type oligonucleotides (Fig. 3C, lanes 1, 3, 5, and 7). To further confirm findings from competition studies, EMSA supershifts with antibodies recognizing PPARγ, RARγ, RXRα, HNF-4α, and/or COUP-TF1/COUP-TF2 were done. At the vegf-D 138/−79 probe, α-PPARγ and/or α-RXRα antibodies reduced formation of complex II, whereas complex I was not altered (Fig. 3D, lanes 2, 4, and 5). In contrast, α-RARγ antibody did not influence complex formation (Fig. 3D, lane 3) unless it was applied together with α-PPARγ or α-RXRα antibodies, which disrupted complex II (Fig. 3D, lanes 5 and 7). In contrast, application of HNF-4α antibody diminished formation of complexes I and II at the vegf-D −138/−79 probe and produced a supershift with probes B and C (Fig. 3D, lane 8). Similarly, anti–COUP-TF1/COUP-TF2 antibody reduced complex formation at the vegf-D −138/−79 probe (Fig. 3D, lane 9, top) and produced supershifts from probes B and C (Fig. 3D, lane 9, middle and bottom). Previous studies suggested that transcription factor AP-1 may regulate vegf-D gene transcription via an AP-1 consensus site located in the human vegf-D 5′-flanking region (12). Our studies revealed that the loss of a putative AP-1 site at −192 had no influence on basal vegf-D promoter activity (Fig. 2A). Similarly, an additional AP-1 core motif at −54 was not able to compensate for the loss of −136/−79 (Fig. 2A), confirming that these AP-1 binding sites are not relevant for control of basal vegf-D gene transcription.

Ectopic expression of HNF-4α and COUP-TF1/COUP-TF2 stimulates vegf-D promoter activity. In contrast to PPARγ, RARγ, or RXRα, which had no influence on vegf-D transcription regardless whether experiments were carried out with or without stimulation, ectopic expression of HNF-4α as well as COUP-TF1 and COUP-TF2 significantly stimulated vegf-D promoter activity (Fig. 4A, data for COUP-TF2 not shown). In accordance with results obtained in gastric cancer cells, enforced expression of wild-type HNF-4α concentration-dependently stimulated vegf-D promoter activity in HNF-4α–deficient HeLa cells, confirming the direct effects of HNF-4α on the vegf-D gene promoter. Expression of mutant HNF4-DBD, which lacks a functional DNA binding domain (30), had no effect (Fig. 4B). Similarly, ectopic expression of COUP-TF1 or COUP-TF2 stimulated vegf-D promoter activity 4- to 5-fold in HNF-4α–negative HeLa cells (Fig. 4C), whereas coexpression of COUP-TF1 or COUP-TF2 along with HNF-4α was not more effective. To confirm that HNF-4α activates the vegf-D promoter via the vegf-D DR element, reporter constructs carrying mutated or wild-type vegf-D DR sequences were used (Fig. 4D). Mutation of the 5′ DR half-site abrogated HNF-4α–triggered promoter activity, whereas the mutant targeting the 3′ DR half-site reduced vegf-D promoter activity by ∼50%. Loss of both DR half-sites (mutant ΔDR5′ + ΔDR3′) abrogated the stimulatory effect of HNF-4α.

Figure 4.

Effect of ectopic expression of nuclear receptor and orphan receptors on vegf-D promoter activity. A, MKN-45 were transfected with vegf-D(−135/−81) along with constructs encoding nuclear receptors RARα, RXRα, and/or PPARγ as well as orphan receptors HNF-4α or COUP-TF1. In addition, transfected cells were stimulated by PPAR agonist prostaglandin J2 (PGJ2) and/or 9-cis retinoic acid (9cisRA) or all-trans retinoic acid (atRA). Results are shown as increase of activities determined in MKN-45 cells transfected with vegf-D promoter construct vegf-D(−135/−81) alone. Columns, mean obtained from three separate experiments; bars, SE. B, cotransfection of HeLa cells with the vegf-D promoter construct vegf-D(−135/−81) and HNF-4α displays a concentration-dependent reporter gene transactivation. Same concentrations of the HNF-4α DNA-binding mutant (HNF4-DBD) showed no transactivation. C, COUP-TF1 and COUP-TF2 stimulate in a concentration-dependent fashion HNF-4α–independent vegf-D gene promoter activity in HeLa cells. HNF-4α–deficient HeLa cells in 24 wells were cotransfected with HNF-4α and/or increasing amounts of COUP-TF1 and COUP-TF2 in the presence of vegf-D(−264/+110). D, HNF-4α–deficient HeLa cells were transfected with the vegf-D(−264/+110) construct or its DR5′ and/or DR3′ mutants alone or along with an expression construct encoding human wild-type HNF-4α. Columns, mean increase of activity determined in vegf-D(−264/+110)–transfected cells obtained from three separate experiments; bars, SE. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 4.

Effect of ectopic expression of nuclear receptor and orphan receptors on vegf-D promoter activity. A, MKN-45 were transfected with vegf-D(−135/−81) along with constructs encoding nuclear receptors RARα, RXRα, and/or PPARγ as well as orphan receptors HNF-4α or COUP-TF1. In addition, transfected cells were stimulated by PPAR agonist prostaglandin J2 (PGJ2) and/or 9-cis retinoic acid (9cisRA) or all-trans retinoic acid (atRA). Results are shown as increase of activities determined in MKN-45 cells transfected with vegf-D promoter construct vegf-D(−135/−81) alone. Columns, mean obtained from three separate experiments; bars, SE. B, cotransfection of HeLa cells with the vegf-D promoter construct vegf-D(−135/−81) and HNF-4α displays a concentration-dependent reporter gene transactivation. Same concentrations of the HNF-4α DNA-binding mutant (HNF4-DBD) showed no transactivation. C, COUP-TF1 and COUP-TF2 stimulate in a concentration-dependent fashion HNF-4α–independent vegf-D gene promoter activity in HeLa cells. HNF-4α–deficient HeLa cells in 24 wells were cotransfected with HNF-4α and/or increasing amounts of COUP-TF1 and COUP-TF2 in the presence of vegf-D(−264/+110). D, HNF-4α–deficient HeLa cells were transfected with the vegf-D(−264/+110) construct or its DR5′ and/or DR3′ mutants alone or along with an expression construct encoding human wild-type HNF-4α. Columns, mean increase of activity determined in vegf-D(−264/+110)–transfected cells obtained from three separate experiments; bars, SE. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

Transcriptional coactivators GRIP-1 and CBP bind and activate the vegf-D gene promoter. Previous reports showed that HNF-4α and COUP-TFs cooperate with p160 transcriptional coactivator GRIP-1 (31, 32) and/or CBP (33) to drive target promoters. To evaluate whether these two cofactors may be involved in vegf-D regulation, chromatin immunoprecipitations were carried out. In accordance with EMSA studies, HNF-4α was found at the proximal vegf-D promoter; however, we were not able to find COUP-TFs although several commercial antibodies and experimental conditions were tested. In addition to HNF-4α, coactivators CBP and GRIP-1 were found to be associated with the vegf-D promoter (Fig. 5A). Interestingly, in HNF-4α–negative HeLa cells, CBP, but not GRIP-1, was detected in the vegf-D binding nuclear complex, suggesting that interaction of CBP may require the presence of HNF-4α (Fig. 5A). To investigate the functional effect of GRIP-1 and CBP on vegf-D regulation, ectopic expression experiments were carried out. In HNF-4α– and COUP-TF–competent AGS cancer cells, expression of CBP or GRIP-1 significantly elevated vegf-D promoter activity (Fig. 5B), showing a more robust transcriptional response when both coactivators were coexpressed. In HNF-4α–deficient HeLa cells, neither GRIP-1 nor CBP significantly stimulated vegf-D promoter activity (Fig. 5C). Expression of GRIP-1 and/or CBP together with HNF-4α resulted in a 3- to 4-fold elevation of vegf-D promoter activity (Fig. 5C), strongly suggesting that the presence of HNF-4α is required for GRIP-1 and CBP to exert their transcriptional effects on the vegf-D gene.

Figure 5.

Transcriptional coactivators GRIP-1 and CBP bind and activate the vegf-D promoter. A, chromatin immunoprecipitation assay of nuclear proteins and transcriptional coactivators recruited to the proximal vegf-D promoter. Chromatin immunoprecipitation analysis was done according to the procedure outlined in Materials and Methods. To determine the functional influence of GRIP-1 and CBP on vegf-D promoter regulation, transfection studies in AGS (B) and HNF-4α–deficient HeLa cells (C) were carried out. For this purpose, cells were transiently transfected with the vegf-D(−264/+110) luciferase reporter gene construct along with indicated amounts of HNF-4α, GRIP-1, and/or CBP. Luciferase activity is depicted as fold increase relative to results obtained after transfection of corresponding empty vectors. Columns, mean from three separate experiments; bars, SE. *, P < 0.05.

Figure 5.

Transcriptional coactivators GRIP-1 and CBP bind and activate the vegf-D promoter. A, chromatin immunoprecipitation assay of nuclear proteins and transcriptional coactivators recruited to the proximal vegf-D promoter. Chromatin immunoprecipitation analysis was done according to the procedure outlined in Materials and Methods. To determine the functional influence of GRIP-1 and CBP on vegf-D promoter regulation, transfection studies in AGS (B) and HNF-4α–deficient HeLa cells (C) were carried out. For this purpose, cells were transiently transfected with the vegf-D(−264/+110) luciferase reporter gene construct along with indicated amounts of HNF-4α, GRIP-1, and/or CBP. Luciferase activity is depicted as fold increase relative to results obtained after transfection of corresponding empty vectors. Columns, mean from three separate experiments; bars, SE. *, P < 0.05.

Close modal

Regulation of vegf-D gene transcription by histone H3 and H4 acetylation. Because previous reports showed that application of histone deacetylase inhibitors can influence expression of angiogenesis genes including vegf-A (34, 35), we investigated the effect of TSA on vegf-D mRNA levels in various cancer cell lines. We found that TSA treatment potently stimulated vegf-D mRNA levels (results for AGS cells shown in Fig. 6A and B). Similarly, vegf-C mRNA levels were potently up-regulated by TSA (Fig. 6B). Application of TSA potently stimulated the activity of a transfected vegf-D reporter gene construct in HeLa and AGS cells (Fig. 6C). Additionally, chromatin immunoprecipitation assays revealed that TSA treatment resulted in a 10- and 20-fold increase of acetylated histones H3 and H4 at the vegf-D promoter (Fig. 6D), strongly supporting the concept that histone deacetylase inhibition stimulates vegf-D transcription through histone-dependent chromatin remodeling.

Figure 6.

Influence of promoter acetylation on vegf-D gene expression. A, AGS gastric cancer cells were incubated with different concentrations of TSA for 24 h and vegf-D mRNA levels were determined by RT-PCR using appropriate primers. Columns, mean vegf-D mRNA abundance relative to solvent-treated controls obtained from three experiments; bars, SE. *, P < 0.05. B, AGS gastric cancer cells were incubated under normoxic conditions for 24 h with 100 ng/mL TSA and endogenous vegf-C and vegf-D mRNA levels were determined by quantitative real-time RT-PCR. After normalization to β-actin as housekeeping gene, the expression of vegf-C and vegf-D is expressed relative to the expression in solvent-treated cells. Representative data from three independent experiments. C, AGS and HeLa cells were transiently transfected with vegf-D(−2011/+110) or empty pGL3 vector. After 24 h of 100 ng/mL TSA treatment, cells were harvested and luciferase activity was measured. Columns, mean from three separate experiments; bars, SE. **, P < 0.01; ***, P < 0.001. D, quantitative chromatin immunoprecipitation analysis of histone H3 and H4 acetylation at the vegf-D promoter. AGS cells were incubated with or without 100 ng/mL TSA for 24 h. Bound DNA was quantitatively amplified by SYBR Green PCR and normalized to the input control and depicted as fold increase of treated versus untreated controls. PCR products were additionally separated on an agarose gel for illustration. Representative measurement of three independent experiments.

Figure 6.

Influence of promoter acetylation on vegf-D gene expression. A, AGS gastric cancer cells were incubated with different concentrations of TSA for 24 h and vegf-D mRNA levels were determined by RT-PCR using appropriate primers. Columns, mean vegf-D mRNA abundance relative to solvent-treated controls obtained from three experiments; bars, SE. *, P < 0.05. B, AGS gastric cancer cells were incubated under normoxic conditions for 24 h with 100 ng/mL TSA and endogenous vegf-C and vegf-D mRNA levels were determined by quantitative real-time RT-PCR. After normalization to β-actin as housekeeping gene, the expression of vegf-C and vegf-D is expressed relative to the expression in solvent-treated cells. Representative data from three independent experiments. C, AGS and HeLa cells were transiently transfected with vegf-D(−2011/+110) or empty pGL3 vector. After 24 h of 100 ng/mL TSA treatment, cells were harvested and luciferase activity was measured. Columns, mean from three separate experiments; bars, SE. **, P < 0.01; ***, P < 0.001. D, quantitative chromatin immunoprecipitation analysis of histone H3 and H4 acetylation at the vegf-D promoter. AGS cells were incubated with or without 100 ng/mL TSA for 24 h. Bound DNA was quantitatively amplified by SYBR Green PCR and normalized to the input control and depicted as fold increase of treated versus untreated controls. PCR products were additionally separated on an agarose gel for illustration. Representative measurement of three independent experiments.

Close modal

In this study, we identify a novel, atypical DR element located in the proximal vegf-D gene promoter as indispensable for vegf-D gene transcription. DR elements were initially identified as binding sites for ligand-activated hormone receptors such as RAR, RXR, and PPARs as well as orphan receptors like HNF-4α and COUP-TFs (36). Typical DR elements consist of two consensus 5′-AGGTCA-3′ half-sites spaced by one to five nucleotides (36, 37). In addition, atypical DR sites comprising degenerated variants of the consensus motif, palindromic configuration, and/or spacing sequences of up to 150 bp were described (36, 37). Importantly, the nucleotide sequence of DR half-sites as well as their spacing sequence determines the selectivity of DR elements toward binding nuclear factors (29). Based on its primary structure comprising a degenerated 3′ DR half-site sequence separated from a consensus half-site by a 20-bp spacer, the vegf-D DR element has to be classified as an atypical DR site. To our knowledge, structure and sequence of the vegf-D DR element are unique and DR elements with this configuration have not yet been described. In accordance with the concept that the half-sites of a given DR element cooperate functionally, we found that individual vegf-D DR half-sites conferred only very weak transcriptional activity, whereas the complete vegf-D DR element potently elevated (10–15-fold) transcription in a heterologous promoter system (Fig. 2). Accordingly, site-directed mutagenesis experiments showed that the integrity of both DR half-sites is indispensable for full vegf-D gene promoter activity (Fig. 3). Previous studies showed that DR elements can bind nuclear receptors without showing responsiveness toward corresponding receptor agonists (38). In line with this observation, we detected binding of PPARγ, RARγ, and RXRα to the vegf-D DR element, whereas application of appropriate agonists failed to stimulate vegf-D gene transcription. This may be explained by DNA-dependent allosteric effects influencing the activating capacity and/or ligand-binding abilities of hormone receptors involved (38). Alternatively, such effects are due to the lack of transcriptional cofactors required for the inducibility of a given complex forming at a DR element (38).

In contrast to PPARγ, RARγ, and RXRα, we found that orphan receptors HNF-4α and COUP-TF1/COUP-TF2 are potent activators of the human vegf-D gene promoter. Because mutations abrogating the binding of HNF-4α and COUP-TF1/COUP-TF2 to the vegf-D DR element also blunted vegf-D transcription, the presence of HNF-4α and COUP-TF1/COUP-TF2 at the proximal DR element seems to be indispensable for vegf-D transcription. Transcriptional effects of HNF-4α are typically mediated via DR1-like response elements (13, 14), but interaction of HNF-4α with DR2-like elements or single atypical DR half-sites has also been shown (39, 40). Similar to our findings with the vegf-D DR element, HNF-4α–activated DR sites were previously shown to be regulated by orphan receptors COUP-TF1/COUP-TF2 (16, 17). COUP-TFs preferentially interact with DR1 elements, but interaction with DR0, DR2, DR6, DR7, and DR9 sites as well as everted DRs has been reported (for review, see ref. 41). Interestingly, COUP-TFs were initially considered as transcriptional repressors of nuclear receptor action (41), but more recently, it became clear that COUP-TFs can also act as transcriptional activators via activation of DR elements (42, 43). In the light of in vivo studies showing that targeted disruption of HNF-4 and/or COUP-TF2 in mice influences endothelial morphogenesis and angiogenesis, our results make it tempting to speculate that at least some of the vascular effects of HNF-4 and/or COUP-TF2 are mediated by VEGF-D.

Activation of gene transcription by DNA-binding transcription factors is mediated by coactivators recruiting RNA polymerase II and its transcription initiation complex to target promoters (44). Complexes formed on stimulation of nuclear receptors frequently comprise p160 transcriptional coactivators such as GRIP, SRC-1, and/or pCIP (4446). Moreover, coactivator CBP and/or its functional homologue p300, both of which possess histone acetylase activity, were detected in such complexes (45). p160 proteins typically link other coactivators to the transcription initiation complex, whereas CBP/p300 can directly acetylate histones and/or other components of the transcription initiation complex (47). Similarly, orphan receptor HNF-4α was shown to interact with CBP/p300 and GRIP-1 (48, 49). In line with these studies, we found that CBP and GRIP-1 were associated with HNF-4α at the proximal vegf-D promoter (Fig. 5A). Functional expression studies confirmed that GRIP-1 and CBP contribute to the regulation of the human vegf-D promoter and strongly suggested that the presence of HNF-4α is required for GRIP-1 and CBP to exert their transcriptional effects. To our knowledge, our study for the first time provides an analysis of coactivator proteins regulating the transcription of a gene of the VEGF ligand/receptor family.

The acetylation/deacetylation of histones plays an important role in the regulation of gene expression (50). The acetylation status of histones is controlled by the opposing activity of two types of enzymes: histone acetylases and histone deacetylases (50). Most of the identified histone acetylases act as transcriptional coactivators, whereas histone deacetylases function in opposition to histone acetylases by deacetylating lysine residues on histone tails. Inappropriate recruitment of histone deacetylases in malignant cells has been identified to result in activation of transcriptional programs supporting the onset and progression of cancer. Conversely, histone deacetylase inhibitors such as TSA were shown to display potential as anticancer agents (50), an effect that could be partially mediated through inhibition of proangiogenic factors such as VEGF-A (34). As a potential mechanism underlying TSA-dependent suppression of the vegf-A gene, interruption of hypoxia-inducible factor 1α–dependent vegf-A activation was proposed (34, 35). Interestingly, in our study, application of TSA stimulated accumulation of acetylated histones H3 and H4 at the vegf-D promoter and potently induced vegf-D gene transcription (Fig. 6). How reversible epigenetic silencing of the vegf-D gene contributes to control of vegf-D expression in the setting of vascular differentiation and/or cancer, however, remains to be elucidated in future investigations.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

C. Wißmann and G. Schäfer contributed equally to this work.

We thank Anja Beyer and Feng Gao for excellent technical assistance, Dr. S. Tarivas (Division of Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg, Germany) for providing the HNFα expression plasmids, Dr. M.M. Moasser (Laboratory of Molecular Medicine, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY) for providing the RARγ and RXRα plasmids, Dr. M. Gurnell (Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom) for providing the PPARγ plasmid, and Dr. M.J. Tsai (Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX) for the expression constructs encoding COUP-TF1 and COUP-TF2 and for the antibody against COUP-TFs.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1
Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific growth factors and blood vessel formation.
Nature
2000
;
407
:
242
–8.
2
Stacker SA, Achen MG, Jussila L, Baldwin ME, Alitalo K. Lymphangiogenesis and cancer metastasis.
Nat Rev Cancer
2002
;
2
:
573
–83.
3
Duff SE, Li C, Jeziorska M, et al. Vascular endothelial growth factors C and D and lymphangiogenesis in gastrointestinal tract malignancy.
Br J Cancer
2003
;
89
:
426
–30.
4
Juttner S, Wissmann C, Jons T, et al. Vascular endothelial growth factor-D and its receptor VEGFR-3: two novel independent prognostic markers in gastric adenocarcinoma.
J Clin Oncol
2006
;
24
:
228
–40.
5
Niki T, Iba S, Tokunou M, Yamada T, Matsuno Y, Hirohashi S. Expression of vascular endothelial growth factors A, B, C, and D and their relationships to lymph node status in lung adenocarcinoma.
Clin Cancer Res
2000
;
6
:
2431
–9.
6
O-charoenrat P, Rhys-Evans P, Eccles SA. Expression of vascular endothelial growth factor family members in head and neck squamous cell carcinoma correlates with lymph node metastasis.
Cancer
2001
;
92
:
556
–68.
7
Enholm B, Paavonen K, Ristimaki A, et al. Comparison of VEGF, VEGF-B, VEGF-C and Ang-1 mRNA regulation by serum, growth factors, oncoproteins and hypoxia.
Oncogene
1997
;
14
:
2475
–83.
8
Orlandini M, Oliviero S. In fibroblasts Vegf-D expression is induced by cell-cell contact mediated by cadherin-11.
J Biol Chem
2001
;
276
:
6576
–81.
9
Ristimaki A, Narko K, Enholm B, Joukov V, Alitalo K. Proinflammatory cytokines regulate expression of the lymphatic endothelial mitogen vascular endothelial growth factor-C.
J Biol Chem
1998
;
273
:
8413
–8.
10
Tsai PW, Shiah SG, Lin MT, Wu CW, Kuo ML. Up-regulation of vascular endothelial growth factor C in breast cancer cells by heregulin-β1. A critical role of p38/nuclear factor-κB signaling pathway.
J Biol Chem
2003
;
278
:
5750
–9.
11
Orlandini M, Semboloni S, Oliviero S. β-Catenin inversely regulates vascular endothelial growth factor-D mRNA stability.
J Biol Chem
2003
;
278
:
44650
–6.
12
Orlandini M, Marconcini L, Ferruzzi R, Oliviero S. Identification of a c-fos-induced gene that is related to the platelet-derived growth factor/vascular endothelial growth factor family.
Proc Natl Acad Sci U S A
1996
;
93
:
11675
–80.
13
Tirona RG, Lee W, Leake BF, et al. The orphan nuclear receptor HNF4α determines PXR- and CAR-mediated xenobiotic induction of CYP3A4.
Nat Med
2003
;
9
:
220
–4.
14
Yamagata K, Furuta H, Oda N, et al. Mutations in the hepatocyte nuclear factor-4α gene in maturity-onset diabetes of the young (MODY1).
Nature
1996
;
384
:
458
–60.
15
Parviz F, Matullo C, Garrison WD, et al. Hepatocyte nuclear factor 4α controls the development of a hepatic epithelium and liver morphogenesis.
Nat Genet
2003
;
34
:
292
–6.
16
Mietus-Snyder M, Sladek FM, Ginsburg GS, et al. Antagonism between apolipoprotein AI regulatory protein 1, Ear3/COUP-TF, and hepatocyte nuclear factor 4 modulates apolipoprotein CIII gene expression in liver and intestinal cells.
Mol Cell Biol
1992
;
12
:
1708
–18.
17
Pineda T, I, Jamshidi Y, Flavell DM, Fruchart JC, Staels B. Characterization of the human PPARα promoter: identification of a functional nuclear receptor response element.
Mol Endocrinol
2002
;
16
:
1013
–28.
18
Pereira FA, Tsai MJ, Tsai SY. COUP-TF orphan nuclear receptors in development and differentiation.
Cell Mol Life Sci
2000
;
57
:
1388
–98.
19
Zhou C, Tsai SY, Tsai M. From apoptosis to angiogenesis: new insights into the roles of nuclear orphan receptors, chicken ovalbumin upstream promoter-transcription factors, during development.
Biochim Biophys Acta
2000
;
1470
:
M63
–68.
20
Schäfer G, Cramer T, Suske G, Kemmner W, Wiedenmann B, Höcker M. Oxidative stress regulates vascular endothelial growth factor-A gene transcription through Sp1- and Sp3-dependent activation of two proximal GC-rich promoter elements.
J Biol Chem
2003
;
278
:
8190
–8.
21
Moasser MM, Reuter VE, Dmitrovsky E. Overexpression of the retinoic acid receptor-γ directly induces terminal differentiation of human embryonal carcinoma cells.
Oncogene
1995
;
10
:
1537
–43.
22
Qiu Y, Krishnan V, Zeng Z, et al. Isolation, characterization, and chromosomal localization of mouse and human COUP-TF I and II genes.
Genomics
1995
;
29
:
240
–6.
23
Taraviras S, Monaghan AP, Schutz G, Kelsey G. Characterization of the mouse HNF-4 gene and its expression during mouse embryogenesis.
Mech Dev
1994
;
48
:
67
–79.
24
Gurnell M, Wentworth JM, Agostini M, et al. A dominant-negative peroxisome proliferator-activated receptor γ (PPARγ) mutant is a constitutive repressor and inhibits PPARγ-mediated adipogenesis.
J Biol Chem
2000
;
275
:
5754
–9.
25
Sladek FM, Zhong WM, Lai E, Darnell JE, Jr. Liver-enriched transcription factor HNF-4 is a novel member of the steroid hormone receptor superfamily.
Genes Dev
1990
;
4
:
2353
–65.
26
Wang LH, Tsai SY, Cook RG, Beattie WG, Tsai MJ, O'Malley BW. COUP transcription factor is a member of the steroid receptor superfamily.
Nature
1989
;
340
:
163
–6.
27
Lunyak VV, Burgess R, Prefontaine GG, et al. Corepressor-dependent silencing of chromosomal regions encoding neuronal genes.
Science
2002
;
298
:
1747
–52.
28
Wingender E, Dietze P, Karas H, Knuppel R. TRANSFAC: a database on transcription factors and their DNA binding sites.
Nucleic Acids Res
1996
;
24
:
238
–41.
29
Nakshatri H and Bhat-Nakshatri P. Multiple parameters determine the specificity of transcriptional response by nuclear receptors HNF-4, ARP-1, PPAR, RAR and RXR through common response elements.
Nucleic Acids Res
1998
;
26
:
2491
–9.
30
Taraviras S, Schutz G, Kelsey G. Generation of inhibitory mutants of hepatocyte nuclear factor 4.
Eur J Biochem
1997
;
244
:
883
–9.
31
Sugiyama T, Wang JC, Scott DK, Granner DK. Transcription activation by the orphan nuclear receptor, chicken ovalbumin upstream promoter-transcription factor I (COUP-TFI). Definition of the domain involved in the glucocorticoid response of the phosphoenolpyruvate carboxykinase gene.
J Biol Chem
2000
;
275
:
3446
–54.
32
Wang JC, Stafford JM, Granner DK. SRC-1 and GRIP1 coactivate transcription with hepatocyte nuclear factor 4.
J Biol Chem
1998
;
273
:
30847
–50.
33
Yoshida E, Aratani S, Itou H, et al. Functional association between CBP and HNF4 in trans-activation.
Biochem Biophys Res Commun
1997
;
241
:
664
–9.
34
Kim MS, Kwon HJ, Lee YM, et al. Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes.
Nat Med
2001
;
7
:
437
–43.
35
Mie LY, Kim SH, Kim HS, et al. Inhibition of hypoxia-induced angiogenesis by FK228, a specific histone deacetylase inhibitor, via suppression of HIF-1α activity.
Biochem Biophys Res Commun
2003
;
300
:
241
–6.
36
Mangelsdorf DJ, Thummel C, Beato M, et al. The nuclear receptor superfamily: the second decade.
Cell
1995
;
83
:
835
–9.
37
Aranda A and Pascual A. Nuclear hormone receptors and gene expression.
Physiol Rev
2001
;
81
:
1269
–304.
38
Kato S, Sasaki H, Suzawa M, et al. Widely spaced, directly repeated PuGGTCA elements act as promiscuous enhancers for different classes of nuclear receptors.
Mol Cell Biol
1995
;
15
:
5858
–67.
39
Fraser JD, Martinez V, Straney R, Briggs MR. DNA binding and transcription activation specificity of hepatocyte nuclear factor 4.
Nucleic Acids Res
1998
;
26
:
2702
–7.
40
Jiang G and Sladek FM. The DNA binding domain of hepatocyte nuclear factor 4 mediates cooperative, specific binding to DNA and heterodimerization with the retinoid X receptor α.
J Biol Chem
1997
;
272
:
1218
–25.
41
Tsai SY and Tsai MJ. Chick ovalbumin upstream promoter-transcription factors (COUP-TFs): coming of age.
Endocr Rev
1997
;
18
:
229
–40.
42
Hwung YP, Crowe DT, Wang LH, Tsai SY, Tsai MJ. The COUP transcription factor binds to an upstream promoter element of the rat insulin II gene.
Mol Cell Biol
1988
;
8
:
2070
–7.
43
Lu XP, Salbert G, Pfahl M. An evolutionary conserved COUP-TF binding element in a neural-specific gene and COUP-TF expression patterns support a major role for COUP-TF in neural development.
Mol Endocrinol
1994
;
8
:
1774
–88.
44
Glass CK and Rosenfeld MG. The coregulator exchange in transcriptional functions of nuclear receptors.
Genes Dev
2000
;
14
:
121
–41.
45
Hermanson O, Glass CK, Rosenfeld MG. Nuclear receptor coregulators: multiple modes of modification.
Trends Endocrinol Metab
2002
;
13
:
55
–60.
46
Xu J, Li Q. Review of the in vivo functions of the p160 steroid receptor coactivator family.
Mol Endocrinol
2003
;
17
:
1681
–92.
47
Chen HW, Lin RJ, Schiltz RL, et al. Nuclear receptor coactivator ACTR is a novel histone acetyltransferase and forms a multimeric activation complex with P/CAF and CBP/p300.
Cell
1997
;
90
:
569
–80.
48
Dell H and Hadzopoulou-Cladaras M. CREB-binding protein is a transcriptional coactivator for hepatocyte nuclear factor-4 and enhances apolipoprotein gene expression.
J Biol Chem
1999
;
274
:
9013
–21.
49
Ruse MD, Jr., Privalsky ML, Sladek FM. Competitive cofactor recruitment by orphan receptor hepatocyte nuclear factor 4α1: modulation by the F domain.
Mol Cell Biol
2002
;
22
:
1626
–38.
50
Marks PA, Richon VM, Breslow R, Rifkind RA. Histone deacetylase inhibitors as new cancer drugs.
Curr Opin Oncol
2001
;
13
:
477
–83.

Supplementary data