The human erythropoietin gene is expressed predominantly in the kidney and liver in response to hypoxia. Although the signaling cascade for hypoxia is present in many different cell types, the expression of erythropoietin is restricted to only a few tissues. The authors show that the promoter and 5′-untranslated region (5′-UTR) of the erythropoietin gene comprise a CpG island and that methylation of the CpG island correlates inversely with expression. Methylation represses the expression of the erythropoietin gene in 2 ways: high-density methylation of the 5′-UTR recruits a methyl-CpG binding protein to the promoter, and methylation of CpGs in the proximal promoter blocks the association of nuclear proteins. (Blood. 2000;95:111-119)

CpG dinucleotides (CpGs) are underrepresented in vertebrate DNA; they occur at 0.2 to 0.25 the frequency expected from the overall nucleotide composition.1 Despite the low average abundance of CpGs, the human genome contains approximately 45,000 limited regions of high-density CpGs known as CpG islands,2,3 most of which are associated with promoters, the 5′ end of genes, or both.4 Housekeeping genes regularly contain CpG islands, as do approximately 40% of genes with tissue-specific expression patterns.5,6 In vertebrate genomic DNA, 60% to 90% of CpGs are modified by symmetric methylation of the fifth position of the cytosine ring (methyl-CpGs).7In contrast, cytosines in CpG islands in the promoters of active genes are largely unmethylated.8 

Gene silencing is the legacy of CpG island methylation. Indeed the transcriptional activity of a promoter often correlates inversely with its methylation status,8,9 and agents that cause demethylation of DNA often induce the expression of previously silent genes.10,11 The best examples of gene silencing induced by CpG methylation are genomic imprinting12 and X-chromosome inactivation in females.13 More recently, DNA methylation has been implicated in the silencing of tumor-suppressor genes14-18 and other genes in cancer cells.19-21 In tissue-specific genes CpG island methylation suppresses expression in tissues that do not require those particular genes.22 Methylation of CpG islands may be epigenetic: the factors and conditions necessary to initiate methylation may not be required for the maintenance of methylation and the subsequent control of gene expression.

Two mechanisms have been proposed to explain the transcriptional repression resulting from CpG methylation. Methylation of cytosines in a DNA recognition element can block the binding of sequence-specifictrans-acting proteins.7 DNA methylation also promotes the binding of a family of proteins that interferes directly or indirectly with the transcriptional apparatus.23Methyl-CpG binding protein-1 (MeCP1) represses transcription from regions with high-density CpG methylation (> 10 methyl-CpGs),24,25 and low-density CpG methylation decreases transcription by recruiting histone deacetylase activity to chromatin through interactions with methyl-CpG-binding protein-2 (MeCP2).26-29 CpG methylation also enhances nucleosome stability by attracting histone H1 to linker regions containing methyl-CpGs.30 31 

The human erythropoietin gene is expressed predominantly in kidney32,33 and liver,34,35 in limited amounts by some hematopoietic tissue,36,37 and in small amounts in other tissues.38,39 Erythropoietin expression is stimulated by hypoxia,40 and the elements that control tissue-specific, hypoxia-induced expression have been studied in detail. When expressed as a transgene in mice, the basal human erythropoietin promoter (∼400 base pair [bp]) has little tissue specificity41—tissue-specific expression requires 6 and 22 kb human DNA upstream of the transcription start site.42,43A transgene driven by a 6-kb murine erythropoietin promoter is expressed predominantly in proximal convoluted tubular cells after hypoxic stress.44 In contrast, in situ hybridization experiments suggest that expression of the native erythropoietin gene in the kidney is limited to infrequent interstitial peritubular cells.45-47 Hepatocytes and Ito cells in the adult liver contribute a small fraction to the erythropoietin pool,35,39,48,49 whereas the extent of erythropoietin production by hematopoietic cells has not been extensively characterized.36,37 Hep3B and HepG2 are human hepatoma cell lines that produce small amounts of erythropoietin under normoxic conditions but that increase their erythropoietin production up to 100-fold after exposure to hypoxia or cobalt.50-52 

We hypothesize that methylation of the erythropoietin gene in nonexpressing cells contributes to tissue-specific expression. We show that the human erythropoietin gene contains a CpG island in its promoter and 5′-UTR that is differentially methylated in respect to gene expression. We demonstrate that CpG methylation of the erythropoietin gene blocks the binding of sequence-specific DNA binding proteins, recruits methyl-CpG binding proteins, and represses transcription.

Cell culture

Hep3B and HeLa cells were cultured in 5% CO2 in minimum essential medium (Gibco-BRL, Gaithersburg, MD) containing 10% iron-enriched bovine calf serum (Hyclone Laboratories, Logan, UT).

Southern blot analysis

Genomic DNA was digested with Xba1 and Eag1, separated on a 1% agarose gel in 1 × TAE buffer, and transferred to Nytran Plus membrane (Schleicher & Schuell, Keene, NH) in 0.4 mol/L NaOH. The blot was incubated overnight at 42°C in 50% formamide with the radiolabeled Xba1 fragment that flanks the transcription start site (Figure 1). The blot was washed in 0.1 × SSC and 0.1% sodium dodecyl sulfate at 65°C, and a digital image was acquired on a PhosphorImager (Molecular Dynamics, Sunnyvale, CA).

Fig. 1.

Schematic map of the erythropoietin gene.

(A) Distribution of CpG sites and a few restriction enzyme sites in an 8765-base pair (bp) fragment of the erythropoietin gene. (B) CpG island is exploded. (black rectangle) GATA box. (long bent arrow) Transcription start site. (short thick arrow) Position of the translation start site, arbitrarily numbered +1.

Fig. 1.

Schematic map of the erythropoietin gene.

(A) Distribution of CpG sites and a few restriction enzyme sites in an 8765-base pair (bp) fragment of the erythropoietin gene. (B) CpG island is exploded. (black rectangle) GATA box. (long bent arrow) Transcription start site. (short thick arrow) Position of the translation start site, arbitrarily numbered +1.

Close modal

Peripheral blood mononuclear cell isolation

Peripheral blood was obtained from normal human volunteers with their informed consent. Mononuclear cells were isolated on a Ficoll step gradient (density, 1.077).

Bisulphite conversion, polymerase chain reaction amplification, and thermocycler sequencing

Bisulphite conversion of DNA was performed as described.53 DNA was denatured for 15 minutes at 37°C in freshly prepared 0.3 mol/L NaOH, then incubated with 3.1 mmol/L sodium bisulphite, pH 5, and 0.5 mmol/L hydroquinone (Sigma Chemical, St. Louis, MO) for 16 hours at 55°C. Polymerase chain reaction (PCR) amplifications were conducted with the following primers: top strand, 5410FT versus 5893R; bottom strand, 5410FB versus 5904R. In some cases, nested PCR was performed with primers 5446F versus 5893R (top strand) and 5441F versus 5904R (bottom strand), and PCR products were extracted from agarose gels with the QIAquick gel extraction kit (QIAGEN, Chatsworth, CA). The purified PCR products were sequenced using the fmol DNA cycle sequencing system (Promega, Madison, WI) and [32P]end-labeled primers. Primer sequences are listed in the Table.

Electrophoretic mobility shift assay

Synthetic oligonucleotides and DNA fragments are listed in the Table. Probes were [32P]end-labeled with T4 polynucleotide kinase (New England BioLabs, Beverly, MA) and extracted from polyacrylamide or agarose gels with QIAEXII gel extraction or QIAquick gel extraction kits (QIAGEN). Electrophoretic mobility shift assay (EMSA) reactions (20 μL) contained nuclear extract from Hep3B and HeLa (5–10 μg protein) and 0.1 pmol probe in binding buffer: 10 mmol/L Tris, pH 8, 1 mmol/L dithiothreitol, 5% glycerol ([vol/vol], 20 mmol/L NaCl, 2 mmol/L MgCl2, 0.01 mg/mL bovine serum albumin), and either 0.1 mg/mL sonicated salmon sperm DNA (CpG 30-mer) or 0.2 mg/mL Escherichia coli DNA (CG11 and EPO236).54 Binding reactions were incubated at room temperature for 30 minutes and were analyzed on 4% nondenaturing polyacrylamide gels.

In vitro methylation, transfection, and luciferase assays

The 117-bp erythropoietin promoter (P117) was inserted immediately upstream of the luciferase reporter gene in pGL2 (Promega), with or without the 126-bp erythropoietin enhancer (E126), distal to the luciferase coding region.54 pGL2-P117 + 5′-UTR+E126 was constructed by inserting the 236-bp Eag1 to Rsa1 fragment of the erythropoietin gene into the Eag1 and Sac 2 sites of pGL2-P117+E126. The blunted erythropoietin Rsa1 site was ligated to a blunted Sac 2 site in pGL2. The P117 promoter was released from pGL2-P117 and pGL2-P117+E126 by digestion with Asp718 and Bgl2, and the 5′-UTR was released from pGL2-P117 + 5′-UTR+E126 by digestion with Eag1 and Bgl2. DNA fragments were methylated or mock-methylated with Sss 1 methylase (New England BioLabs) and then were ligated into their respective unmethylated backbones. The plasmids were introduced into Hep3B cells as calcium phosphate–DNA precipitates, and the cells were incubated under normoxia or hypoxia (1% oxygen) for 48 hours. Cell lysates were prepared, and luciferase activity was measured as previously described.54 A plasmid containing the cytomegalovirus promoter driving the lac z gene was included to correct for transfection efficiency.

In vivo digestion with restriction endonucleases

After 8 hours of hypoxia (1% oxygen), cells from three 150-mm plates were suspended in 2 mL 150 mmol/L sucrose, 35 mmol/L HEPES, pH 7.4, 80 mmol/L KCl, 5 mmol/L K2HPO4, 5 mmol/L MgCl2, and 0.5 mmol/L CaCl2. Membranes were made permeable by incubating for 1 minute with 0.05% lysolecithin (Sigma Chemical) and then were washed with digestion buffer (10 mmol/L Tris, pH 7.7, 50 mmol/L NaCl, 5 mmol/L MgCl2, and 1 mmol/L dithiothreitol).55 Aliquots containing 3 × 106 cells were treated with Msp1 for 10 minutes at 37°C. Cells were collected by centrifugation and suspended in 40 μL digestion buffer. DNA was isolated with the Genomic Prep Cells and Tissue DNA Isolation Kit (Amersham Pharmacia Biotech, Piscataway, NJ) and digested with Xba1 (−678) to provide an internal reference for digestion and amplification.

Ligation-mediated polymerase chain reaction

Ligation-mediated PCR was performed with Vent DNA polymerase (New England BioLabs) in 5% dimethyl sulfoxide with gene-specific primers BP4, BP5, and BP6 (Table) as described.56 After 24 cycles of PCR, the products were subjected to 4 cycles of linear amplification using [32P]end-labeled primers. The annealing temperatures for first-strand synthesis, PCR amplification, and labeling reactions were 65°C, 68°C, and 70°C, respectively.

Graphics

Figures were produced with Adobe Illustrator (Adobe Systems, Mountain View, CA). PhosphorImager data were analyzed with ImageQuaNT (Molecular Dynamics) without modifying the linear response curve. The CpG maps were created with VectorNTI (InforMax, Bethesda, MD).

Several criteria should be met before it can be suggested that CpG methylation is involved in the tissue-specific regulation of a gene: (1) control elements of the gene should contain CpG residues, (2) methylation of the control regions of the gene should mirror gene activity in expressing and nonexpressing cells, (3) proteins that bind the control regions should be present in expressing and nonexpressing cells, (4) methylation of the control elements should affect protein binding, and (5) methylation of the control elements should affect transcriptional activity in functional assays. We provide the following experimental data in support of methylation controlling expression of the erythropoietin gene.

CpGs in the erythropoietin gene

Figure 1 is a schematic representation of 8765 bp of the human erythropoietin gene. The map begins 5788 bp upstream of the translation start site (arbitrarily defined as base pair + 1) at a BamH1 site, ends at a Pvu 2 site distal to the polyadenylation signal, and contains 203 CpGs that disproportionately cluster around the transcription start site (−240). The promoter has 25 CpGs in 320 bp, and the 5′-UTR has 31 CpGs in 240 bp, 7.8 and 12.9 CpGs per 100 bp, respectively. The highest density of CpGs is a 30-bp region in the 5′-UTR that contains 8 CpGs. The promoter and 5′-UTR comprise a CpG island based on criteria set forth by Gardiner-Garden and Frommer.6 Specifically, the GC content is 74.6%, the CpG rich region is >200 bp in length, and the observed-to-expected ratio of CpGs is 0.75.

Analysis of the erythropoietin gene with a methylation-sensitive restriction enzyme

We screened the CpG island in the erythropoietin gene for methylation by exploiting the sensitivity of Eag1 to CpG methylation within its recognition site. The human erythropoietin gene (Fig. 1) has an Eag1 site 220 bp proximal to the translation start site that is flanked by sites for Xba1 (cleaves irrespective of methylation). Genomic DNA was digested with Xba1 and Eag1, and restriction fragments were identified by Southern blot analysis. Cleavage by both enzymes generates restriction fragments of 461 and 789 bp; methylation of the Eag1 site precludes Eag1 digestion, resulting in a single 1250-bp Xba1 fragment. DNA from Hep3B (expressing cells) is digested to completion with Eag1, whereas DNA from HeLa (nonexpressing cells) is resistant to Eag1 digestion (Fig. 2), consistent with methylation-sensitive expression of the erythropoietin gene.

Fig. 2.

Southern blot analysis of methylation in Hep3B and HeLa cells.

Genomic DNA from Hep3B and HeLa cells was digested with Xba1 and Eag1, fractionated by size, transferred to a nylon membrane, and hybridized to the full-length Xba1 fragment of the erythropoietin gene. The Xba1 fragment is 1250 bp long, and the combined Xba1/Eag1 fragments measure 461 and 789 bp. Nox, normoxia; Hyp, hypoxia for 8 hours.

Fig. 2.

Southern blot analysis of methylation in Hep3B and HeLa cells.

Genomic DNA from Hep3B and HeLa cells was digested with Xba1 and Eag1, fractionated by size, transferred to a nylon membrane, and hybridized to the full-length Xba1 fragment of the erythropoietin gene. The Xba1 fragment is 1250 bp long, and the combined Xba1/Eag1 fragments measure 461 and 789 bp. Nox, normoxia; Hyp, hypoxia for 8 hours.

Close modal

Analysis of CpG methylation by sodium bisulfite conversion

Most CpGs in the human erythropoietin gene are not in restriction enzyme recognition sites and cannot be analyzed by methylation-sensitive cleavage assays. Sodium bisulfite conversion displays the methylation status of all cytosines.53Treatment of DNA with sodium bisulfite converts cytosine to uracil; 5-methylcytosine is unaffected because methylation blocks deamination. During PCR amplification of bisulfite-treated DNA, thymidines are incorporated in place of converted cytosines. Converted cytosines are detected by DNA sequence analysis of pooled PCR products or individual clones.

The methylation status of the human erythropoietin promoter (−288 to +81) was analyzed in Hep3B and HeLa cells by sodium bisulfite conversion (Figures 3A, 3B). In normoxic Hep3B cells, all cytosines are converted to uracil except a single CpG on the coding strand, 238 bp proximal to the translation start site that is hemimethylated (Figures 3A, 3B, diamond). In hypoxic Hep3B cells, this CpG is not methylated (data not shown). In contrast each cytosine in CpG sites from −288 to +81 of the erythropoietin gene has some resistance to sodium bisulfite conversion in HeLa (Figs. 3A,3B). The proximal 5′-UTR containing the highest density of CpGs (26.7%) is heavily methylated. The CpGs in the upstream region of the human erythropoietin promoter (−558 to −288) are only sporadically methylated in HeLa (data not shown).

Fig. 3.

CpG methylation in the erythropoietin promoter and 5′-UTR.

DNA from normoxic Hep3B, HeLa cells, and peripheral blood mononuclear cells was treated with sodium bisulfite. Polymerase chain reaction amplification was performed, and the DNA sequence was obtained from the complementary strands (guanines converted to adenines). (A) Coding strand sequence from −288 to −133. (B) Noncoding strand sequence from −66 to −289. (C) Coding strand sequence from −288 to −133 from peripheral blood mononuclear cells (PMC1 and PMC2). (bent arrows) Transcription start site. (open circles) Partially methylated CpGs. (filled circles) Fully methylated CpGs. (diamonds) Hemimethylated site present in normoxic Hep3B cells.

Fig. 3.

CpG methylation in the erythropoietin promoter and 5′-UTR.

DNA from normoxic Hep3B, HeLa cells, and peripheral blood mononuclear cells was treated with sodium bisulfite. Polymerase chain reaction amplification was performed, and the DNA sequence was obtained from the complementary strands (guanines converted to adenines). (A) Coding strand sequence from −288 to −133. (B) Noncoding strand sequence from −66 to −289. (C) Coding strand sequence from −288 to −133 from peripheral blood mononuclear cells (PMC1 and PMC2). (bent arrows) Transcription start site. (open circles) Partially methylated CpGs. (filled circles) Fully methylated CpGs. (diamonds) Hemimethylated site present in normoxic Hep3B cells.

Close modal

We verified the methylation of the human erythropoietin promoter with genomic DNA from peripheral blood mononuclear cells, a heterogeneous mixture of T-lymphocytes, B-lymphocytes, natural killer cells, and monocytes. DNA from these normal human cells has a methylation pattern similar to that of HeLa DNA (Figure 3C).

Proteins bind methyl-CpGs in the erythropoietin 5′-UTR

We sought to demonstrate by EMSA that methyl-CpG binding proteins in Hep3B and HeLa extracts bind high-density CpGs in a methylation-dependent manner. The Eag1 to Rsa1 fragment of the erythropoietin gene (EPO236; see Figure 1) and a synthetic high-density CpG probe (CG11) were symmetrically methylated in vitro with a CpG methylase. Methylated CG11 is a ligand for methyl-CpG binding proteins such as MeCP1.24,57 Several protein complexes form on unmethylated CG11 (Figure 4A; lane 3) and EPO236 (Figure 4B; lane 3) in nuclear extracts from HeLa cells, but none of these complexes is methylation dependent (Figures 4A, 4B; lanes 4–8). When challenged with methylated CG11 and EPO236, new complexes are formed in both extracts (Figures 4A, 4B; lanes 2, 4; arrows). In HeLa extracts the shifted complexes form in the presence of excess unmethylated CG11 or EPO236 competitor (Figure 4; lanes 5, 6), but not when incubated with an excess of either methylated competitor (Figure4; lanes 7, 8): behavior consistent with MeCP binding proteins.23 Similar complexes form in Hep3B extracts (Fig. 4; lanes 1, 2).

Fig. 4.

Erythropoietin 5′-UTR recruits a methyl-CpG–binding protein.

Electrophoretic mobility shift assays were performed with methylated and unmethylated CG11 and EPO236 as probes and competitors. [32P]-end-labeled probes were incubated with nuclear extracts and, where indicated, with excess unlabeled competitors. (A) CG11 probe. (B) EPO236 probe. U, unmethylated; M, methylated probe or competitor. Probe identity and methylation status are noted below the panels, and competitors and sources of nuclear extracts are noted above the panels. (arrow) Methyl-CpG–binding complex. (asterisks) Complexes formed on unmethylated probes blocked by methylation. (arrowheads) Protein complexes formed on unmethylated and methylated probes, whose formation is blocked by methylated and unmethylated competitors.

Fig. 4.

Erythropoietin 5′-UTR recruits a methyl-CpG–binding protein.

Electrophoretic mobility shift assays were performed with methylated and unmethylated CG11 and EPO236 as probes and competitors. [32P]-end-labeled probes were incubated with nuclear extracts and, where indicated, with excess unlabeled competitors. (A) CG11 probe. (B) EPO236 probe. U, unmethylated; M, methylated probe or competitor. Probe identity and methylation status are noted below the panels, and competitors and sources of nuclear extracts are noted above the panels. (arrow) Methyl-CpG–binding complex. (asterisks) Complexes formed on unmethylated probes blocked by methylation. (arrowheads) Protein complexes formed on unmethylated and methylated probes, whose formation is blocked by methylated and unmethylated competitors.

Close modal

In vivo protection of methyl-CpGs in HeLa cells

Several CpGs in the human erythropoietin promoter and 5′-UTR fall within Msp1 sites. If these CpGs recruit methyl-CpG–binding proteins when methylated, the restriction enzyme recognition sites should be protected from Msp1 cleavage in vivo. To this end we rendered the cellular and nuclear membranes of HeLa and Hep3B cells permeable and exposed the cells to Msp1, a restriction enzyme that cleaves irrespective of methylation. Cleavage sites in the DNA were detected by ligation-mediated PCR. In Hep3B cells, Msp1 cleaves the DNA at each restriction site (Figure 5; lanes 1–3); however, in HeLa cells, 4 Msp1 sites in the 5′-UTR are resistant to digestion, especially the site at −219 (Figure 5; lanes 4, 5; arrow). The in vivo resistance of HeLa DNA to Msp1 digestion suggests that methyl-CpG–binding proteins interact with a region of dense methylation in the erythropoietin 5′-UTR.

Fig. 5.

Restriction endonuclease accessibility of the erythropoietin promoter and 5′-UTR.

Hypoxic Hep3B and HeLa cells were incubated in vivo with Msp1 after a brief treatment with lysolecithin. DNA was purified and digested with Xba1 to create an amplification standard. Restriction enzyme cleavage of the DNA was detected by ligation-mediated polymerase chain reaction. X, nonspecific band. (arrow and bracket) Sites with increased resistance to Msp1 cleavage in HeLa. (filled circle) Xba1 site. The schematic diagram shows (dumbbells) Msp1 sites, (bent arrow) transcription start site, and (rectangle) GATA box. The ramp corresponds to increasing Msp1 concentration.

Fig. 5.

Restriction endonuclease accessibility of the erythropoietin promoter and 5′-UTR.

Hypoxic Hep3B and HeLa cells were incubated in vivo with Msp1 after a brief treatment with lysolecithin. DNA was purified and digested with Xba1 to create an amplification standard. Restriction enzyme cleavage of the DNA was detected by ligation-mediated polymerase chain reaction. X, nonspecific band. (arrow and bracket) Sites with increased resistance to Msp1 cleavage in HeLa. (filled circle) Xba1 site. The schematic diagram shows (dumbbells) Msp1 sites, (bent arrow) transcription start site, and (rectangle) GATA box. The ramp corresponds to increasing Msp1 concentration.

Close modal

Methyl-CpGs in the proximal erythropoietin promoter

Low-density methylation inhibits transcription by recruiting a repressive protein complex to the promoter58 or interfering with the binding of essential trans-acting proteins. The promoter of the erythropoietin gene has numerous CpGs; however, the CpGs in the promoter (upstream of −240) are largely unmethylated (data not shown). A cluster of 3 CpGs immediately upstream of the GATA box (−267) in the erythropoietin proximal promoter is partially methylated in HeLa and peripheral blood mononuclear cells but is unmethylated in Hep3B cells (Figures 3,6). The differential methylation of these CpG sites and their proximity to the GATA box suggest this region may play a role in the tissue-specific regulation and basal activity of the promoter. DNA/protein interactions in this part of the promoter have been previously shown to affect erythropoietin expression.59 We next chose to evaluate the 30-bp region encompassing these CpGs (CpG 30-mer; Figure7D) for methylation-dependent DNA/protein interactions with extracts from Hep3B and HeLa cells.

Fig. 6.

CpG methylation in the erythropoietin promoter from −306 to −275.

DNA from Hep3B and HeLa cells was modified by bisulfite conversion. Converted DNA was amplified, and polymerase chain reaction products were sequenced. Coding strand: sequence from the complementary strand (guanines are converted to adenines). Noncoding strand: sequence from the original strand (cytosines converted to thymines). (open circles) Methylated CpGs.

Fig. 6.

CpG methylation in the erythropoietin promoter from −306 to −275.

DNA from Hep3B and HeLa cells was modified by bisulfite conversion. Converted DNA was amplified, and polymerase chain reaction products were sequenced. Coding strand: sequence from the complementary strand (guanines are converted to adenines). Noncoding strand: sequence from the original strand (cytosines converted to thymines). (open circles) Methylated CpGs.

Close modal
Fig. 7.

Specific DNA/protein interaction is blocked by CpG methylation.

A panel of complementary oligonucleotides comprising CpG 30-mer was synthesized with 5-methylcytosines in all 3 CpGs, in each CpG individually, and in none of the CpGs. Mixing the oligonucleotides creates probes and competitors for electrophoretic mobility shift assays with diverse methylation profiles. (A) Unmethylated CpG 30-mer (U) and CpG 30-mer symmetrically methylated on all 3 CpGs (M) were used as probes and competitors as indicated above panel A. (arrow) Position of major binding complexes. (lanes 10, 11) Probe contains 5-methylcytosine on the sense (S) or antisense (A) strands at CpG1, CpG2, and CpG3. (lanes 6–8) A murine antibody to human aryl hydrocarbon nuclear translocator, ARNT (2B10), was included in the reactions. (open circle) Position of an anti-ARNT supershift. (lane 8) No nuclear extract. (lane 9, asterisk) Addition of a murine antibody to an unrelated antibody. (B) The binding complex from HeLa extracts and unmethylated CpG 30-mer is challenged with a 50-fold excess of double-stranded competitors (see Table). (C) Shifted complexes form with Hep3B extracts and CpG 30-mer symmetrically methylated at either CpG1 (m1), CpG2 (m2), or CpG3 (m3). (lanes 5–15) Shifted complexes formed with Hep3B extracts and unmethylated CpG 30-mer are challenged with CpG 30-mer symmetrically methylated at each CpG (m1-3) or hemimethylated at each CpG on the sense (S1–S3) or antisense (A1–A3) strand. (D) Schematic map of CpG 30-mer.

Fig. 7.

Specific DNA/protein interaction is blocked by CpG methylation.

A panel of complementary oligonucleotides comprising CpG 30-mer was synthesized with 5-methylcytosines in all 3 CpGs, in each CpG individually, and in none of the CpGs. Mixing the oligonucleotides creates probes and competitors for electrophoretic mobility shift assays with diverse methylation profiles. (A) Unmethylated CpG 30-mer (U) and CpG 30-mer symmetrically methylated on all 3 CpGs (M) were used as probes and competitors as indicated above panel A. (arrow) Position of major binding complexes. (lanes 10, 11) Probe contains 5-methylcytosine on the sense (S) or antisense (A) strands at CpG1, CpG2, and CpG3. (lanes 6–8) A murine antibody to human aryl hydrocarbon nuclear translocator, ARNT (2B10), was included in the reactions. (open circle) Position of an anti-ARNT supershift. (lane 8) No nuclear extract. (lane 9, asterisk) Addition of a murine antibody to an unrelated antibody. (B) The binding complex from HeLa extracts and unmethylated CpG 30-mer is challenged with a 50-fold excess of double-stranded competitors (see Table). (C) Shifted complexes form with Hep3B extracts and CpG 30-mer symmetrically methylated at either CpG1 (m1), CpG2 (m2), or CpG3 (m3). (lanes 5–15) Shifted complexes formed with Hep3B extracts and unmethylated CpG 30-mer are challenged with CpG 30-mer symmetrically methylated at each CpG (m1-3) or hemimethylated at each CpG on the sense (S1–S3) or antisense (A1–A3) strand. (D) Schematic map of CpG 30-mer.

Close modal

Protein interactions with CpG 30-mer

Protein interactions with CpG 30-mer were evaluated by EMSA. Unmethylated CpG 30-mer produces a single-shift complex with nuclear extract from Hep3B and a doublet with extracts from HeLa (Figure 7A; lane 3; arrow). Complex formation is blocked by a 50-fold excess of unmethylated self-competitor (Figure 7A; lane 4), but not by competitors containing an SP-1 site, the erythropoietin enhancer steroid receptor element direct repeat (DR2), or a fragment of the erythropoietin promoter (P56) (Figure 7B; Table). The HIF1 element from the erythropoietin enhancer partially blocks formation of the lower complex in HeLa extracts (Figure 7B; lane 5). These experiments confirm that nuclear proteins from Hep3B and HeLa bind an element in CpG 30-mer in a sequence-specific manner.

CpG 30-mer shares sequence homology with the aryl hydrocarbon nuclear translocator (ARNT)-binding element, suggesting that ARNT is involved in the DNA/protein complex. We tested this hypothesis by incubating HeLa and Hep3B nuclear extracts, unmethylated CpG 30-mer, and 2B10 (Affinity Bioreagents, Golden, CO), an antibody to ARNT. A small portion of the specific complex is further shifted, suggesting ARNT is a part of the complex (Figure 7A; lane 6; open circle). Control reactions—including an excess of self-competitor, omission of nuclear extract, and inclusion of an antibody to an unrelated antigen—confirm the specificity of the ARNT antibody (Figure 7A; lanes 7, 8, 9).

Effect of methylation of CpG 30-mer on protein binding

CpG 30-mer symmetrically methylated at all 3 CpGs does not bind protein from HeLa or Hep3B extracts (Figure 7A; lanes 1, 2), nor does it block the formation of specific complexes when used as an unlabeled competitor against unmethylated CpG 30-mer (Figure 7A; lane 5). Even hemimethylation of the 3 CpGs on the sense or antisense strand of CpG 30-mer blocks the formation of DNA/protein complexes (Figure 7A; lanes 10, 11). Because these CpGs are only partially methylated in cells that do not express erythropoietin (Figures 3, 6), we chose to test whether methylation of each individual CpG in CpG 30-mer could block the formation of sequence-specific protein complexes. Symmetric methylation of CpG2 prevents the formation of specific complexes with Hep3B extracts (Figure 7C; lane 3), and both symmetric methylation (Figure 7; lane 8) and hemimethylation (Figure 7; lanes 11, 14) of CpG2 render the DNA fragment ineffective as a competitor. Symmetric methylation of CpG1 or CpG3 has little effect on DNA/protein interactions (Figure 7C; lanes 2, 4 [probes] and lanes 7, 9 [competitors]). CpG 30-mer hemimethylated at CpG1 competes well for protein binding to unmethylated CpG 30-mer (Figure 7C; lanes 10, 13), but hemimethylation of CpG3 on the antisense strand impairs its ability to act as a competitor (Figure 7C; lane 15). Similar results were obtained with HeLa extracts (data not shown).

Methylation of the promoter or 5′-UTR versus transcriptional activity

We examined whether methylation of the promoter or 5′-UTR could block the activity of the erythropoietin promoter in transient expression assays. The minimal promoter was selectively methylated at 11 CpGs in the context of the unmethylated pGL2 backbone ± the 126-bp erythropoietin enhancer (the enhancer was not methylated). In a separate experiment, the 5′-UTR was selectively methylated at 31 CpGs in the context of the unmethylated pGL2-P117+E126 backbone.

Without an enhancer, methylation of the CpGs in the erythropoietin promoter has a minor repressive effect on normoxic expression of luciferase. Under hypoxic conditions, however, the expression of luciferase falls by 40% (Figure 8A). In the context of the erythropoietin enhancer (unmethylated), methylation of erythropoietin promoter produces a substantial decrease in luciferase expression—the basal activity of the promoter (normoxia) falls by 35%, and the expression of the minigene under hypoxic conditions falls by 85% (Figure 8B). Methylation of the erythropoietin 5′-UTR in the context of an unmethylated erythropoietin promoter and enhancer results in a fall in luciferase activity of 30% for basal expression and 60% for inducible expression (Figure 8C).

Fig. 8.

The effect of methylation on the transcriptional activity of the erythropoietin promoter and 5′-UTR.

The minimal inducible erythropoietin promoter (P117) was methylated or mock methylated, then ligated into unmethylated pGL2 (pGL2-P117) or pGL2 with the unmethylated erythropoietin enhancer (pGL2-P117+E126). The 5′-UTR was methylated or mock methylated and ligated in its natural position into unmethylated pGL2-P117+E126, creating pGL2-P117 + 5′-UTR+E126. Reporter vectors were introduced into Hep3B cells, and luciferase assays were performed. Relative light units are expressed as raw light units divided by mU/min of β-galactosidase activity. (error bars) ±1 SD. (A) pGL2-P117 (n = 4). (B) pGL2-P117+E126 (n = 4). (C) pGL2-P117 + 5′-UTR+E126 (n = 5). U, unmethylated; M, methylated; Nox, normoxia; Hyp, hypoxia.

Fig. 8.

The effect of methylation on the transcriptional activity of the erythropoietin promoter and 5′-UTR.

The minimal inducible erythropoietin promoter (P117) was methylated or mock methylated, then ligated into unmethylated pGL2 (pGL2-P117) or pGL2 with the unmethylated erythropoietin enhancer (pGL2-P117+E126). The 5′-UTR was methylated or mock methylated and ligated in its natural position into unmethylated pGL2-P117+E126, creating pGL2-P117 + 5′-UTR+E126. Reporter vectors were introduced into Hep3B cells, and luciferase assays were performed. Relative light units are expressed as raw light units divided by mU/min of β-galactosidase activity. (error bars) ±1 SD. (A) pGL2-P117 (n = 4). (B) pGL2-P117+E126 (n = 4). (C) pGL2-P117 + 5′-UTR+E126 (n = 5). U, unmethylated; M, methylated; Nox, normoxia; Hyp, hypoxia.

Close modal

The minimal inducible promoter of the human erythropoietin gene consists of a 117-bp region upstream of the transcription start site (−240 bp proximal to the translation start site),54is highly G and C rich, and lacks a canonical TATA box. A GATA box located 25 to 30 bp upstream of the transcription start site may serve as the dock for TFIID/TBP and may bind a GATA family member.60 61 Despite the simplicity of the erythropoietin promoter and its similarity to housekeeping genes, erythropoietin expression is exquisitely tissue specific.

The cis-acting and trans-acting elements that control the hypoxic regulation of the erythropoietin gene have been carefully dissected. A hypoxia-inducible enhancer62 lies 3′ of the polyadenylation signal of the erythropoietin gene and cooperates with the minimal inducible erythropoietin promoter.54 Both the enhancer and the promoter interact with hypoxia-inducible factor-1 (HIF1),63 a heterodimer of HIF1-α, and ARNT. Other genes are activated by hypoxia through HIF1, including glycolytic enzymes,64,65 glucose transporters, 64-67 and vascular endothelial-derived growth factor.68 An intact hypoxia-signaling pathway is present in most cells69,70; hence, the silence of the erythropoietin gene in most tissues is perplexing. We previously demonstrated that sequence-specific binding proteins do not occupy the HIF1 element in the erythropoietin enhancer in HeLa cells,56 despite their expression of active HIF1 during hypoxia. The exclusion of HIF1 and other proteins from the enhancer suggests that the erythropoietin gene must assume a tissue-specific permissive state before activation by hypoxia. We believe the accessibility of the enhancer depends in part on the activity of the promoter, and we doubt that the permissive state of the promoter is determined by the ubiquitous factor HIF1. The presence of a CpG island in the promoter prompted us to study the methylation of the erythropoietin gene and to search for proteins that bind the gene in a methylation-dependent fashion.

The 5′-UTR of the erythropoietin gene has a region of high CpG density that is methylated in HeLa but is unmethylated in Hep3B cells. Although methylation of CpG islands in nonessential genes may be commonplace in cells lines and may not reflect the situation in primary tissues,71 we have shown that the methylation pattern of the erythropoietin 5′-UTR in normal peripheral blood mononuclear cells and HeLa cells is similar. Proteins from HeLa and Hep3B nuclear extracts bind to the methylated 5′-UTR in a manner consistent with methyl-CpG–binding proteins. Interactions between proteins and methyl-CpGs in the erythropoietin 5′-UTR were confirmed by in vivo footprints that prove the erythropoietin gene has different architectures in expressing and nonexpressing cells. Ex vivo methylation of the CpGs in the 5-UTR impedes transcription from the erythropoietin promoter during transient expression in Hep3B cells, consistent with the known repressive effects of methyl-CpG–binding proteins.

In contrast to the 5′-UTR, methylation of CpGs in the proximal erythropoietin promoter interferes with the binding of a sequence-specific DNA-binding protein. The binding element contains 3 CpGs that are methylated only in nonexpressing cells, and symmetric methylation or hemimethylation of the middle CpGs blocks formation of the DNA/protein complexes. The proteins in the complexes have not been identified, but they may include ARNT based on antibody recognition. Ex vivo methylation of the CpGs in the minimal erythropoietin promoter subverts the function of the promoter and substantially blocks the functional cooperation between the promoter and the enhancer.54 Recently, methylation of the HIF1 site in the erythropoietin enhancer has been shown to block HIF1 binding and to decrease the transcriptional activation conferred by the enhancer.72 

A CpG near the transcription start site is hemimethylated in normoxic, but not in hypoxic, Hep3B cells. We have not determined whether hemimethylation at this site affects the transcriptional activity of the promoter. The role of asymmetric methylation in eukaryotic gene regulation is largely undefined.73-75 

We have presented evidence that methylation of the promoter and 5′-UTR of the human erythropoietin gene contributes to tissue-specific regulation, and we propose the following model to explain the silencing of erythropoietin expression by methylation (Figure 9). Methylation provides 2 levels of control, both dependent on DNA/protein interactions. Methylation of 3 CpGs in the proximal promoter blocks the binding of essentialtrans-acting proteins, thereby indirectly repressing transcription. High-density CpG methylation in the 5′-UTR permits the binding of a methyl-CpG binding protein that either directly represses transcription or recruits corepressors, histone deacetylases, or both. Additional studies of primary human tissues will be necessary to verify that methylation is essential for the tissue-specific regulation of the erythropoietin gene and to identify the factors and control elements necessary for initiation and maintenance of the methylated state.

Fig. 9.

A model for silencing the erythropoietin gene by methylation.

Methylation of CpGs in a cis element upstream of the GATA box prevents the binding of a trans-acting protein, impairing transcriptional activation. The sequence-specific binding activity may contain aryl hydrocarbon nuclear translocator (ARNT). Methylation of high-density CpGs in the erythropoietin 5′-UTR recruits a methyl-CpG–binding protein (MeCP) that represses transcription by engaging corepressors (Co-R), histone deacetylases, or both.

Fig. 9.

A model for silencing the erythropoietin gene by methylation.

Methylation of CpGs in a cis element upstream of the GATA box prevents the binding of a trans-acting protein, impairing transcriptional activation. The sequence-specific binding activity may contain aryl hydrocarbon nuclear translocator (ARNT). Methylation of high-density CpGs in the erythropoietin 5′-UTR recruits a methyl-CpG–binding protein (MeCP) that represses transcription by engaging corepressors (Co-R), histone deacetylases, or both.

Close modal

The authors thank Jeanne Housman for her emotional and financial support.

Supported by Public Health Service grant DK-46967 from the National Institute of Diabetes and Digestive and Kidney Diseases (KLB).

Reprints:Kerry L. Blanchard, Feist–Weiller Cancer Center, Louisiana State University Medical Center, 1501 Kings Highway, Shreveport, LA 71103; e-mail: kblanc@lsumc.edu.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Swartz
MN
Trautner
TA
Kornberg
A
Enzymatic synthesis of deoxyribonucleic acid.
J Biol Chem.
237
1962
1961
1967
2
Antequera
F
Bird
A
Number of CpG islands and genes in human and mouse.
Proc Natl Acad Sci U S A.
90
1993
11995
11999
3
Antequera
F
Bird
A
Predicting the total number of human genes [letter; comment].
Nat Genet.
8
1994
114
4
Cross
SH
Bird
AP
CpG islands and genes.
Curr Opin Genet Dev.
5
1995
309
314
5
Larsen
F
Gundersen
G
Lopez
R
Prydz
H
CpG islands as gene markers in the human genome.
Genomics.
13
1992
1095
1107
6
Gardiner-Garden
M
Frommer
M
CpG islands in vertebrate genomes.
J Mol Biol.
196
1987
261
282
7
Siegfried
Z
Cedar
H
DNA methylation: a molecular lock.
Curr Biol.
7
1997
R305
307
8
Naveh-Many
T
Cedar
H
Active gene sequences are undermethylated.
Proc Natl Acad Sci U S A.
78
1981
4246
4250
9
Meehan
R
Lewis
J
Cross
S
Nan
X
Jeppesen
P
Bird
A
Transcriptional repression by methylation of CpG.
J Cell Sci.
16(suppl)
1992
9
14
10
Taylor
SM
Jones
PA
Multiple new phenotypes induced in 10T1/2 and 3T3 cells treated with 5-azacytidine.
Cell.
17
1979
771
779
11
Harris
M
Induction of thymidine kinase in enzyme-deficient Chinese hamster cells.
Cell.
29
1982
483
492
12
Razin
A
Shemer
R
DNA methylation in early development.
Hum Mol Genet.
4
1995
1751
1755
13
Riggs
AD
Pfeifer
GP
X-chromosome inactivation and cell memory.
Trends Genet.
8
1992
169
174
14
Schroeder
M
Mass
MJ
CpG methylation inactivates the transcriptional activity of the promoter of the human p53 tumor suppressor gene.
Biochem Biophys Res Commun.
235
1997
403
406
15
Herman
JG
Latif
F
Weng
Y
et al
Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma.
Proc Natl Acad Sci U S A.
91
1994
9700
9704
16
Herman
JG
Merlo
A
Mao
L
et al
Inactivation of the CDKN2/p16/MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers.
Cancer Res.
55
1995
4525
4530
17
Bird
AP
The relationship of DNA methylation to cancer.
Cancer Surv.
28
1996
87
101
18
Stirzaker
C
Millar
DS
Paul
CL
et al
Extensive DNA methylation spanning the Rb promoter in retinoblastoma tumors.
Cancer Res.
57
1997
2229
2237
19
Lapidus
RG
Ferguson
AT
Ottaviano
YL
et al
Methylation of estrogen and progesterone receptor gene 5′ CpG islands correlates with lack of estrogen and progesterone receptor gene expression in breast tumors.
Clin Cancer Res.
2
1996
805
810
20
Rice
JC
Massey-Brown
KS
Futscher
BW
Aberrant methylation of the BRCA1 CpG island promoter is associated with decreased BRCA1 mRNA in sporadic breast cancer cells.
Oncogene.
17
1998
1807
1812
21
Graff
JR
Herman
JG
Lapidus
RG
et al
E-cadherin expression is silenced by DNA hypermethylation in human breast and prostate carcinomas.
Cancer Res.
55
1995
5195
5199
22
Bird
AP
Gene number, noise reduction and biological complexity [see comments].
Trends Genet.
11
1995
94
100
23
Hendrich
B
Bird
A
Identification and characterization of a family of mammalian methyl-CpG binding proteins.
Mol Cell Biol.
18
1998
6538
6547
24
Meehan
RR
Lewis
JD
McKay
S
Kleiner
EL
Bird
AP
Identification of a mammalian protein that binds specifically to DNA containing methylated CpGs.
Cell.
58
1989
499
507
25
Boyes
J
Bird
A
Repression of genes by DNA methylation depends on CpG density and promoter strength: evidence for involvement of a methyl-CpG binding protein.
EMBO J.
11
1992
327
333
26
Jones
PL
Veenstra
GJ
Wade
PA
et al
Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription.
Nat Genet.
19
1998
187
191
27
Meehan
RR
Lewis
JD
Bird
AP
Characterization of MeCP2, a vertebrate DNA binding protein with affinity for methylated DNA.
Nucl Acids Res.
20
1992
5085
5092
28
Nan
X
Ng
HH
Johnson
CA
et al
Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex [see comments].
Nature
393
1998
386
389
29
Razin
A
CpG methylation, chromatin structure and gene silencing: a three-way connection.
EMBO J.
17
1998
4905
4908
30
McArthur
M
Thomas
JO
A preference of histone H1 for methylated DNA.
EMBO J.
15
1996
1705
1714
31
Ball
DJ
Gross
DS
Garrard
WT
5-methylcytosine is localized in nucleosomes that contain histone H1.
Proc Natl Acad Sci U S A.
80
1983
5490
5494
32
Ratcliffe
PJ
Jones
RW
Phillips
RE
Nicholls
LG
Bell
JI
Oxygen-dependent modulation of erythropoietin mRNA levels in isolated rat kidneys studied by RNAse protection.
J Exp Med.
172
1990
657
660
33
Jacobson
LO
Goldwasser
E
Fried
W
Pizak
L
Role of the kidney in erythropoiesis.
Nature (London)
179
1957
633
634
34
Zanjani
ED
Poster
J
Burlington
H
Mann
LI
Wasserman
LR
Liver as the primary site of erythropoietin formation in the fetus.
J Lab Clin Med.
89
1977
640
644
35
Eckardt
KU
Pugh
CW
Meier
M
Tan
CC
Ratcliffe
PJ
Kurtz
A
Production of erythropoietin by liver cells in vivo and in vitro.
Ann N Y Acad Sci.
718
1994
50
60
36
Rich
IN
Vogt
C
Pentz
S
Erythropoietin gene expression in vitro and in vivo detected by in situ hybridization.
Blood Cells.
14
1988
505
520
37
Stopka
T
Zivny
JH
Stopkova
P
Prchal
JF
Prchal
JT
Human hematopoietic progenitors express erythropoietin.
Blood.
91
1998
3766
3772
38
Tan
CC
Eckardt
KU
Firth
JD
Ratcliffe
PJ
Feedback modulation of renal and hepatic erythropoietin mRNA in response to graded anemia and hypoxia.
Am J Physiol.
263
1992
F474
481
39
Gopfert
T
Eckardt
KU
Geb
B
Kurtz
A
Oxygen-dependent regulation of erythropoietin gene expression in rat hepatocytes.
Kidney Int.
51
1997
502
506
40
Jelkmann
W
Erythropoietin: structure, control of production, and function.
Physiol Rev.
72
1992
449
489
41
Semenza
GL
Traystman
MD
Gearhart
JD
Antonarakis
SE
Polycythemia in transgenic mice expressing the human erythropoietin gene.
Proc Natl Acad Sci U S A.
86
1989
2301
2305
42
Semenza
GL
Koury
ST
Nejfelt
MK
Gearhart
JD
Antonarakis
SE
Cell-type-specific and hypoxia-inducible expression of the human erythropoietin gene in transgenic mice.
Proc Natl Acad Sci U S A.
88
1991
8725
8729
43
Madan
A
Lin
C
Hatch
SL
II
Curtin
PT
Regulated basal, inducible, and tissue-specific human erythropoietin gene expression in transgenic mice requires multiple cis DNA sequences.
Blood.
85
1995
2735
2741
44
Loya
F
Yang
Y
Lin
H
Goldwasser
E
Albitar
M
Transgenic mice carrying the erythropoietin gene promoter linked to lacZ express the reporter in proximal convoluted tubule cells after hypoxia.
Blood.
84
1994
1831
1836
45
Lacombe
C
Da Silva
JL
Bruneval
P
et al
Peritubular cells are the site of erythropoietin synthesis in the murine hypoxic kidney.
J Clin Invest.
81
1988
620
623
46
Koury
ST
Bondurant
MC
Semenza
GL
Koury
MJ
The use of in situ hybridization to study erythropoietin gene expression in murine kidney and liver.
Microsc Res Tech.
25
1993
29
39
47
Fisher
JW
Koury
S
Ducey
T
Mendel
S
Erythropoietin production by interstitial cells of hypoxic monkey kidneys.
Br J Haematol.
95
1996
27
32
48
Caro
J
Zon
LI
Silver
R
Miller
O
Erslev
AJ
Erythropoietin in liver tissue extracts and in liver perfusates from hypoxic rats.
Am J Physiol.
244
1983
E431
434
49
Maxwell
PH
Ferguson
DJ
Osmond
MK
et al
Expression of a homologously recombined erythopoietin-SV40 T antigen fusion gene in mouse liver: evidence for erythropoietin production by Ito cells.
Blood.
84
1994
1823
1830
50
Goldberg
MA
Glass
GA
Cunningham
JM
Bunn
HF
The regulated expression of erythropoietin by two human hepatoma cell lines.
Proc Natl Acad Sci U S A.
84
1987
7972
7976
51
Goldberg
MA
Gaut
CC
Bunn
HF
Erythropoietin mRNA levels are governed by both the rate of gene transcription and posttranscriptional events.
Blood.
77
1991
271
277
52
Fandrey
J
Bunn
HF
In vivo and in vitro regulation of erythropoietin mRNA: measurement by competitive polymerase chain reaction.
Blood.
81
1993
617
623
53
Clark
SJ
Harrison
J
Paul
CL
Frommer
M
High sensitivity mapping of methylated cytosines.
Nucl Acids Res.
22
1994
2990
2997
54
Blanchard
KL
Acquaviva
AM
Galson
DL
Bunn
HF
Hypoxic induction of the human erythropoietin gene: cooperation between the promoter and enhancer, each of which contains steroid receptor response elements.
Mol Cell Biol.
12
1992
5373
5385
55
Pfeifer
GP
Riggs
AD
Chromatin differences between active and inactive X chromosomes revealed by genomic footprinting of permeabilized cells using DNase I and ligation-mediated PCR.
Genes Dev.
5
1991
1102
1113
56
Hu
B
Wright
E
Campbell
L
Blanchard
KL
In vivo analysis of DNA-protein interactions on the human erythropoietin enhancer.
Mol Cell Biol.
17
1997
851
856
57
Singal
R
Ferris
R
Little
JA
Wang
SZ
Ginder
GD
Methylation of the minimal promoter of an embryonic globin gene silences transcription in primary erythroid cells.
Proc Natl Acad Sci U S A.
94
1997
13724
13729
58
Lewis
JD
Meehan
RR
Henzel
WJ
et al
Purification, sequence, and cellular localization of a novel chromosomal protein that binds to methylated DNA.
Cell.
69
1992
905
914
59
Gupta
M
Goldwasser
E
The role of the near upstream sequence in hypoxia-induced expression of the erythropoietin gene.
Nucl Acids Res.
24
1996
4768
4774
60
Imagawa
S
Yamamoto
M
Miura
Y
GATA transcription factors negatively regulate erythropoietin gene expression.
Acta Haematol.
95
1996
248
256
61
Imagawa
S
Yamamoto
M
Miura
Y
Negative regulation of the erythropoietin gene expression by the GATA transcription factors.
Blood.
89
1997
1430
1439
62
Semenza
GL
Nejfelt
MK
Chi
SM
Antonarakis
SE
Hypoxia-inducible nuclear factors bind to an enhancer element located 3′ to the human erythropoietin gene.
Proc Natl Acad Sci U S A.
88
1991
5680
5684
63
Wang
GL
Semenza
GL
Purification and characterization of hypoxia-inducible factor 1.
J Biol Chem.
270
1995
1230
1237
64
Semenza
GL
Jiang
BH
Leung
SW
et al
Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1.
J Biol Chem.
271
1996
32529
32537
65
Firth
JD
Ebert
BL
Pugh
CW
Ratcliffe
PJ
Oxygen-regulated control elements in the phosphoglycerate kinase 1 and lactate dehydrogenase A genes: similarities with the erythropoietin 3′ enhancer.
Proc Natl Acad Sci U S A.
91
1994
6496
6500
66
Ebert
BL
Firth
JD
Ratcliffe
PJ
Hypoxia and mitochondrial inhibitors regulate expression of glucose transporter-1 via distinct Cis-acting sequences.
J Biol Chem.
270
1995
29083
29089
67
Ebert
BL
Gleadle
JM
JF
OR
Bartlett
SM
Poulton
J
Ratcliffe
PJ
Isoenzyme-specific regulation of genes involved in energy metabolism by hypoxia: similarities with the regulation of erythropoietin.
Biochem J.
313
1996
809
814
68
Minchenko
A
Salceda
S
Bauer
T
Caro
J
Hypoxia regulatory elements of the human vascular endothelial growth factor gene.
Cell Mol Biol Res.
40
1994
35
39
69
Wang
GL
Semenza
GL
General involvement of hypoxia-inducible factor 1 in transcriptional response to hypoxia.
Proc Natl Acad Sci U S A.
90
1993
4304
4308
70
Maxwell
PH
Pugh
CW
Ratcliffe
PJ
Inducible operation of the erythropoietin 3' enhancer in multiple cell lines: evidence for a widespread oxygen-sensing mechanism.
Proc Natl Acad Sci U S A.
90
1993
2423
2427
71
Antequera
F
Boyes
J
Bird
A
High levels of de novo methylation and altered chromatin structure at CpG islands in cell lines.
Cell.
62
1990
503
514
72
Wenger
RH
Kvietikova
I
Rolfs
A
Camenisch
G
Gassmann
M
Oxygen-regulated erythropoietin gene expression is dependent on a CpG methylation-free hypoxia-inducible factor-1 DNA-binding site.
Eur J Biochem.
253
1998
771
777
73
Ingelbrecht
I
Van Houdt
H
Van Montagu
M
Depicker
A
Posttranscriptional silencing of reporter transgenes in tobacco correlates with DNA methylation.
Proc Natl Acad Sci U S A.
91
1994
10,502
10,506
74
Toth
M
Muller
U
Doerfler
W
Establishment of de novo DNA methylation patterns: transcription factor binding and deoxycytidine methylation at CpG and non-CpG sequences in an integrated adenovirus promoter.
J Mol Biol.
214
1990
673
683
75
Woodcock
DM
Lawler
CB
Linsenmeyer
ME
Doherty
JP
Warren
WD
Asymmetric methylation in the hypermethylated CpG promoter region of the human L1 retrotransposon.
J Biol Chem.
272
1997
7810
7816
Sign in via your Institution