Recent findings have established a connection between DNA methylation and transcriptionally inactive chromatin characterized by deacetylated histones. Because the absence of estrogen receptor α(ERα) gene expression has been associated with aberrant methylation of its CpG island in a significant fraction of breast cancers, we tested whether histone deacetylase activity contributes to the transcriptional inactivation of the methylated ER gene in a panel of ER-negative human breast cancer cells. Treatment of these cells with trichostatin A, a specific histone deacetylase inhibitor, led to dose- and time-dependent re-expression of ER mRNA as detected by reverse transcription-PCR without alteration in ERα CpG island methylation. Trichostatin A-induced ER re-expression was associated with increased sensitivity to DNase I at the ER locus in MDA-MB-231 cells. These data implicate inactive chromatin mediated by histone deacetylation as a critical component of ER gene silencing in human breast cancer cells. Therefore, histone deacetylation may be a potential target for therapeutic intervention in the treatment of a subset of ER-negative breast cancers.

Interaction between 17β-estradiol and ERα3 plays an important role in breast carcinogenesis and breast cancer treatment. That estrogen stimulates the growth of certain breast cancers is well established, and hormonal therapy via estrogen depletion or antiestrogen administration is widely used to block the action of estrogen in women with breast cancer. However, patients whose breast cancers lack ER seldom respond to endocrine therapy; therefore, a potential mechanism for hormone resistance is de novo or acquired loss of ER gene expression at the transcriptional level during disease progression (1, 2).

One possible mechanism for loss of ER in ER-negative breast cancers is cytosine methylation of the ER CpG island in the 5′regulatory region of the gene (3). Indeed the ER gene CpG island is extensively methylated in ER-negative breast cancer cells, and ∼50% of unselected primary breast tumors but remains unmethylated in normal breast tissue and many ER-positive tumors and ER-positive cancer cell lines (4, 5). The functional importance of this finding is demonstrated by the fact that treatment of ER-negative human breast cancer cells with the demethylating agent, 5-aza-dC, led to reactivation of expression of ER mRNA and functional ER protein (6). Recent studies indicate that silencing of a gene by methylation involves the generation of an inactive chromatin structure characterized by deacetylated histones. An abundant chromosomal methyl CpG-binding protein, MeCP2, was the first protein identified to link methylated DNA and a HDAC-containing transcriptionally repressive complex for gene silencing. Subsequently, several MBD proteins have been identified that, similar to MeCP2, couple methylated DNA to HDAC (7, 8). More recently, the well-known maintenance methyltransferase,Dnmt1, was found to interact physically with HDAC through its N terminus, thereby forming a transcriptionally inactive chromatin structure that represses transcription (9, 10). All of these findings demonstrate the important role of HDAC in transcriptional regulation. The HDACs deacetylate lysine groups of histones H3 and H4, allowing ionic interactions between positively charged lysines and negatively charged DNA, which result in a more compact nucleosome structure that limits transcription. The availability of specific HDAC inhibitors such as TSA (11)permits the study of the role of HDAC in silencing a variety of tissue-specific methylated genes (7, 12).

Here, we have tested whether loss of ER expression in some breast cancers is associated with transcriptional repression through HDAC activity on the methylated ER gene. Our data demonstrate that specific HDAC inhibition via TSA treatment can reactivate ER transcription in the presence of the methylated DNA. The activated gene transcription is associated with increased sensitivity of the ER promoter to DNase I treatment. These data suggest that inactive chromatin mediated by HDAC is critical to ER gene silencing.

Cell Lines, Reagents, and TSA Treatment.

Human breast cancer cells (Hs578t, MCF-7/WT,MCF-7/AdrR, T-47D, and MDA-MB-231) were grown in DMEM supplemented with 10% fetal bovine serum and 2 mml-glutamine. TSA was purchased from Wako Pure Chemical Industries, Ltd. (Osaka, Japan), dissolved in absolute ethanol at a stock concentration of 1 mg/ml (3.30 mm), and stored at−20°C. DNase I was purchased from Pharmacia Biotech (Pharmacia Biotech, NJ). Cells (MDA-MB-231, Hs578t, T-47D, or MCF-7/AdrR) were seeded at a density of 6 × 105 cells/100-mm tissue culture dish (8 × 103cells/cm2). After 24 h of incubation, the culture medium was changed to different concentrations of TSA- or vehicle (ethanol)-containing medium. Either total cellular RNA or genomic DNA was isolated after 0, 24, 48, or 72 h of TSA exposure.

RNA Isolation and RT-PCR Analysis of ER Expression.

Total cellular RNA was isolated from cell lines with TRIzol reagent according to the recommendations of the supplier (Life Technologies,Inc., Rockville, MD). RNA (3 μg) was reversibly transcribed by Moloney murine leukemia virus reverse transcriptase (Life Technologies)using OligoDT15 primer (Promega Corp., Madison,WI) in a final volume of 50 μl. Four % of synthesized cDNA (2 μl,derived from 150 ng of initial RNA) was used for PCR amplification of ER and the constitutively expressed housekeeping gene β-actin(13). Specific sense and antisense PCR primers used for the amplifications across the seventh intron of ER and the first intron of β-actingenes, yielding 470 and 400 bp of PCR products respectively, were described previously (6). PCR products were resolved by 2% agarose gel electrophoresis and visualized by ethidium bromide staining.

Quantitative Competitive PCR Analysis of ER Reactivation.

A quantitative assay was performed to determine the level of ER mRNA in TSA- and vehicle-treated MDA-MB-231 cells as compared with the expression levels in the known ER-positive MCF-7 and T-47D cells using the method of Wang and Rowley (14). This assay involves coamplification of a wild-type target cDNA (wER) of unknown amount and a competitive template (cER) in known amounts. A truncated competitive template was generated with a sense primer containing a 22-bp deletion(primer 2) and the same antisense primer (primer 3) as the wild-type(Fig. 2 A). After amplification, the competitive template was separated, gel purified, and quantified by Spectrophotometer DU 640(Beckman, CA). Thus, target wild-type and known amounts of competitive templates can be coamplified with the pair of wild-type primers (primers 1 and 3) and differentiated by size. Because the most accurate results are obtained when wild-type and competitive templates are amplified at nearly equivalent concentrations, resulting in the signal ratio of wER:cER equivalent to 1, we first performed an initial titration in log and then in 2-fold dilutions to determine the approximate concentration of the wild-type ER cDNA in our experimental samples. RNAs under comparison were simultaneously reversibly transcribed to achieve equal efficiency for reverse transcription. The PCR reactions were carried out with 0.5 μm of wild-type sense and antisense primers for 35 cycles. The wild-type and competitive PCR products were fractionated on 2.5% agarose gel,stained with ethidium bromide, and scanned by Densitometer (EagleSight Software of Eagle Eye II Imaging System; Stratagene, La Jolla, CA). The ratio between wild-type and control templates was determined and used to calculate the amount of target wild-type cDNA because the input of competitive template is known.

Genomic DNA Isolation and MSP Analysis of ER CpG Island.

DNA was isolated by standard phenol-chloroform extraction. Isolated DNA was subjected to modification by sodium bisulfite to convert unmethylated cytosines but not methylated cytosines to uracil as described previously (15). Methylation status of the bisulfite-modified DNA at the ER locus was characterized by methylation-specific PCR using a method described previously (5).

DNase I Sensitivity Assay.

This assay was performed according to the method of Keshet et al.(16) using cells from TSA- or vehicle-treated MDA-MB-231 and MCF 7 cells (1 × 107 cells/each) with the following modifications. The isolated DNA was digested with EcoRI, the recognition sites of which flank the ER promoter region to yield a 3.1-kb fragment that was separated by 1% agarose gel electrophoresis. DNAs blotted on nylon membrane were probed with a PCR-amplified, 561-bp DNA fragment corresponding to the ER CpG island (Fig. 4 A). The sense and antisense oligonucleotides used to amplify the fragment are 5′-AGACCAGTACTTAAAGTTGGAGGCC-3′ and 5′-GGGAAACCCCCCAGG-3′. The amplified DNA was cloned into pCR2.1-TOPO vector (Invitrogen,CA) according to the manufacturer’s protocol. Colonies containing amplified DNA sequence, determined by Mini-Prep (Promega Wizard Mini-Prep kits), were grown and purified. The purified plasmids were sequenced via automated sequencing (Johns Hopkins Sequencing Core Facility). The specific ER CpG probe was prepared from sequence-confirmed plasmid and labeled with bio-16-dUTP (Boehringer Mannheim, IN) by PCR using the above-mentioned primer set. The 3.1-kb DNA band containing the ER promoter region was visualized by chemiluminescence using a streptavidin-conjugated,alkaline phosphatase-catalyzed substrate, CDP-star. The band signals that reflect the resistance to DNase I were quantified by densitometry(Stratagene), and the cumulative DNase I sensitivity was calculated as follows. The band density at each DNase I concentration was divided by the density of the control band and then multiplied by 100%. This value was subtracted from 100% to yield the percentage of DNase I sensitivity. The sum of the percentage of DNase I sensitivity at each dose was considered to be the cumulative DNase I sensitivity.

A growing body of data demonstrates the importance of histone acetylation and deacetylation and corresponding structural alteration of chromatin in gene transcriptional regulation (7, 8, 10). The ER-negative cell line, MDA-MB-231, the ERCpG island of which is densely methylated, was used as a cell model to test whether HDAC activity contributes to repression of ER expression in ER-negative breast cancer cells. TSA, a specific and potent HDAC inhibitor, was used as a pharmacological tool. Exposure of MDA-MB-231 cells to increasing concentrations of TSA led to induction of ER mRNA synthesis in a dose-dependent manner (Fig. 1,A). A detectable level of ER mRNA, as demonstrated by a RT-PCR product with predicted 470-bp size,was noted after treatment with 50 ng/ml (160 nm)TSA for 48 h, and ER transcript was clearly present after 100 ng/ml (330 nm) TSA for the same duration. A time course analysis showed that a weak signal could be seen after 24 h of 100 ng/ml TSA treatment, whereas ER transcript was readily observed after 48 or 72 h (Fig. 2 B). Multiple experiments were done to achieve optimal conditions for TSA induction of ER mRNA in MDA-MB-231 cells. These showed that maximal ER reactivation was achieved with 100 ng/ml TSA for 48 h using an initial seeding density of 8 × 103cells/cm2. Higher inoculating cell densities reduced ER transcript signal (data not shown).

To ascertain whether HDAC activity could play a role in repression of ER expression more generally, the dose response and time course studies described above were extended to other ER-negative human breast cancer cells lines with methylated ER CpG islands. As shown in Fig. 1 C, TSA treatment led to re-expression of ER mRNA in all three ER-negative cell lines tested. Optimal ER gene re-expression was observed after treatment of MDA-MB-231 cells with 100 ng/ml (330 nm), Hs578t cells with 400 ng/ml (1.32μ m), and MCF-7/AdrR with 25 ng/ml (82.5 nm) TSA for 48 h. Therefore,TSA treatment consistently induced ER re-expression in the panel of ER-negative cell lines, supporting a role for HDAC in ERgene silencing.

A quantitative competitive PCR assay was used to assess the magnitude of TSA-induced ER mRNA transcript in MDA-MB-231 cells. Fig. 2,A shows the design of the primer sets used, and validation of the quantitative competitive PCR is shown in Fig. 2,B. As shown in Fig. 2,C, a 5-fold increase in ER transcript was obtained after TSA exposure in MDA-MB-231 cells (100 ng/ml for 48 h). This effect was specific for the methylated ER promoter because TSA treatment (50 or 100 ng/ml for 48 h) of ER-positive, unmethylated MCF-7 cells had no effect on the level of ER mRNA expression using the same quantitative assay (data not shown). However, TSA treatment of MDA-MB-231 cells did not restore ER mRNA expression to the levels seen in cell lines with endogenous ER expression, as shown in Fig. 2 C. Quantitative assay suggested that the level of ER transcript seen with TSA treatment of MDA-MB-231 cells represented about 1 and 10% of that seen in the ER-positive MCF-7 and T-47D cell lines, respectively. Several possibilities might account for this:

(a) It is possible that only partial reactivation is seen because only a fraction of cells responded to the treatment. Indeed, a similar pattern of partial reactivation was seen in MDA-MB-231 cells exposed to a demethylating agent, 5-aza-dC, in our previous study(6).

(b) It has been shown that a component of the repression mediated by MeCP2 transcriptional repression domain is partially HDAC independent; mSin3A could retain some ability to repress transcription,even in the absence of associated HDACs (7).

(c) Simultaneous inhibition of several components in the methylation-associated repressive complexes might be necessary to achieve maximal reactivation of the repressed genes (12).

The ability of TSA to reactivate ER expression raised the question of whether the ER CpG island remained methylated. Our previous studies with 5-aza-dC demonstrated that ER re-expression was associated with demethylation of the ER CpG island. However, a parallel study of the progesterone receptor in progesterone receptor-negative human breast cancer cells showed that the ligand-bound ER could overcome methylation-related repression of the progesterone receptor,even in the continuing presence of a methylated progesterone receptor CpG island (17). We therefore examined the methylation status of the ER CpG island in TSA-treated MDA-MB-231 cells using a sensitive MSP assay that allowed examination of methylation status across the ER CpG island (Fig. 3,A). As shown in Fig. 3,B, the ER CpG island was completely methylated across the entire CpG island in MDA-MB-231 cells treated with vehicle or TSA (100 ng/ml for 48 h). A single primer set, ER 5, was used to confirm this finding in the other TSA-treated, ER-negative human breast cancer cells, Hs578t and MCF-7/AdrR (Fig. 3 C). As expected, the unmethylated ER-positive, MCF-7 cells demonstrated an unmethylated pattern using all four primer sets that span the ER CpG island. In summary, TSA treatment of ER-negative breast cancer cells can lead to re-expression of ER mRNA without an apparent alteration in the methylation status of the ER CpG island.

Because acetylated histones are generally associated with transcriptionally active chromatin whereas deacetylated histones are often found in conjunction with an inactive chromatin state(18), we next studied whether HDAC inhibition could alter chromatin structure at the ER gene locus. Because nuclease susceptibility is one of the characteristics of active chromatin(19), we used a DNase I sensitivity assay to examine chromatin conformation of the ER gene in ER-negative MDA-MB-231 cells in the presence or absence of the HDAC inhibitor, TSA. Cells were treated with 100 ng/ml of TSA for 48 h, a treatment course shown previously to result in optimal re-expression of ER mRNA. Equal amounts of purified nuclei from control and TSA-treated MDA-MB-231 cells were exposed to increasing concentrations of DNase I as described in “Materials and Methods.” Nuclei isolated from ER-positive MCF-7 cells served as a DNase I accessible control. As expected, the ER locus in MCF-7 cells was a highly DNase I sensitive region, whereas the ER locus in control MDA-MB-231 cells was relatively resistant to DNase I digestion (Fig. 4,B). TSA treatment of MDA-MB-231 cells resulted in an ∼2-fold increase in DNase I sensitivity (Fig. 4 C), suggesting that inhibition of HDAC activity leads to a more open chromatin conformation, even in the presence of CpG island methylation.

Recently, the interaction between DNA methylation and histone deacetylation linked by methyl-binding proteins (MBDs), or the direct interaction of Dnmt1 with HDAC as well as other corepressors, has been an area of active study. More recently, a nucleosome-stimulated ATPase Mi2, a part of chromatin remodeling machinery, was also shown to bind the methylated DNA through MBD3 and deacetylase in Xenopus laevis and mammalian cells, further illustrating the role of HDAC on gene transcription regulation (8). In addition to evaluating the role of HDAC in in vitro studies, it is of importance to study its role in silencing endogenous methylated genes. In some cases, HDAC inhibition alone seems to be sufficient to reactivate a methylated gene. For example, both sodium butyrate and TSA can restore transcription from methylated and silenced plant rRNA genes(20). Also, reactivation of transcription of the methylated FMR1 gene was achieved by treatment with 4-phenylbutyrate, sodium butyrate or TSA (21). However, in other cases, both demethylation and HDAC inhibition appear to be necessary. For example, certain hypermethylated genes like MLH1,TIMP3, CDKN2B, and CDKN2A can be transcriptionally activated in colon cancer cells by TSA only after Dnmt1 inhibition by 5-aza-dC, suggesting an important role of Dnmt1 in transcription repression although recruiting HDAC is essential (12).

ER is a critical growth-regulatory gene in breast cancer, and its expression status is tightly linked to the prognosis and treatment outcome of breast cancer patients. Thus, it is important to understand its regulation. Our work suggests that histone deacetylation and DNA methylation may both play a role in ER transcription, and further studies will focus on the effects of TSA on ER protein expression. This is critical because it is possible that activation of the silenced ER by HDAC inhibition could open a new avenue for management of a subset of advanced breast cancer with hormonal resistance. Studies using primary breast cancers have shown that the antiestrogen, tamoxifen, confers a benefit to women whose breast cancer expresses ER by immunohistochemistry in as few as 1–10%of tumor cells. Thus, even partial re-expression of ER could be of clinical benefit (22).

Fig. 1.

Upper panels: A, dose response of TSA on ER transcription in MDA-MB-231 cells analyzed by RT-PCR. B,time course of TSA (100 ng/ml) on re-expression of ER mRNA for 48 h in MDA-MB-231 cells. C, RT-PCR analysis of ER mRNA re-expression by TSA for 48 h in a panel of known ER-negative breast cancer cell lines. The ER-positive prototype, MCF-7, was used as a RT-PCR-positive control. Lower panels: β-actin RT-PCR product provides a control for the amount of intact RNA used in the reactions. VC, vehicle-treated; 231,MDA-MB-231; Hs5, Hs578t; Adr,MCF-7/Adr.

Fig. 1.

Upper panels: A, dose response of TSA on ER transcription in MDA-MB-231 cells analyzed by RT-PCR. B,time course of TSA (100 ng/ml) on re-expression of ER mRNA for 48 h in MDA-MB-231 cells. C, RT-PCR analysis of ER mRNA re-expression by TSA for 48 h in a panel of known ER-negative breast cancer cell lines. The ER-positive prototype, MCF-7, was used as a RT-PCR-positive control. Lower panels: β-actin RT-PCR product provides a control for the amount of intact RNA used in the reactions. VC, vehicle-treated; 231,MDA-MB-231; Hs5, Hs578t; Adr,MCF-7/Adr.

Close modal
Fig. 2.

Quantitative competitive PCR analysis of ER expression in breast cancer cells. A, primer design. As shown above,the competitive sense primer 2 is a 40-bp oligonucleotide that harbors a 22-bp deletion in the middle of the primer. Its sequence at the 5′ end is the same as the wild-type sense primer 1. Thus, primers 2 and 3 amplify cER, but wild-type primer pairs 1 and 3 amplify both wER and cER. Arrow bars, locations of sense and antisense primers. wER, wild-type ER; cER, competitive ER. B, validation of the competitive PCR. wER and cER from MCF-7 cells were amplified with the wild-type primers using different dilutions of competitive template DNA. The cER PCR product decreases and the wER PCR product increases when decreasing known amounts of cER and a constant but unknown amount of wER cDNA is used for PCR (see“Materials and Methods”). The amount of wER can be ascertained from the titration curve at the point where an equal amount of cER and wER is, in this example, between 100 and 10 fg. C, using this quantitative assay, TSA treatment (100 ng/ml for 48 h) of MDA-MB-231 cells induced a 5-fold increase in ER transcript. Equivalence of wER/cER signal was seen at 0.25 fg cER for TSA-treated cells and 0.05 fg cER for control cells (signal ratio of wER:cER = 1 in both TSA-treated and control cells; see“Materials and Methods”), thus demonstrating a 5-fold induction. Using the same calculations (signal ratio of wER:cER multiplied by amount of cER), the TSA-induced transcript reached about 1 and 10% of that found in ER-positive MCF-7 and T-47D cells, respectively. A representative example of four experiments that gave similar results is shown.

Fig. 2.

Quantitative competitive PCR analysis of ER expression in breast cancer cells. A, primer design. As shown above,the competitive sense primer 2 is a 40-bp oligonucleotide that harbors a 22-bp deletion in the middle of the primer. Its sequence at the 5′ end is the same as the wild-type sense primer 1. Thus, primers 2 and 3 amplify cER, but wild-type primer pairs 1 and 3 amplify both wER and cER. Arrow bars, locations of sense and antisense primers. wER, wild-type ER; cER, competitive ER. B, validation of the competitive PCR. wER and cER from MCF-7 cells were amplified with the wild-type primers using different dilutions of competitive template DNA. The cER PCR product decreases and the wER PCR product increases when decreasing known amounts of cER and a constant but unknown amount of wER cDNA is used for PCR (see“Materials and Methods”). The amount of wER can be ascertained from the titration curve at the point where an equal amount of cER and wER is, in this example, between 100 and 10 fg. C, using this quantitative assay, TSA treatment (100 ng/ml for 48 h) of MDA-MB-231 cells induced a 5-fold increase in ER transcript. Equivalence of wER/cER signal was seen at 0.25 fg cER for TSA-treated cells and 0.05 fg cER for control cells (signal ratio of wER:cER = 1 in both TSA-treated and control cells; see“Materials and Methods”), thus demonstrating a 5-fold induction. Using the same calculations (signal ratio of wER:cER multiplied by amount of cER), the TSA-induced transcript reached about 1 and 10% of that found in ER-positive MCF-7 and T-47D cells, respectively. A representative example of four experiments that gave similar results is shown.

Close modal
Fig. 3.

MSP analysis for the ER CpG island in TSA-treated ER-negative breast cancer cells. A, map of ER CpG island and location of ER MSP primers. B, MSP analysis of ER gene CpG island in TSA-treated MDA-MB-231 cells. Both control (231) and TSA-treated (231 TSA) MDA-MB-231 cells showed evidence of ER CpG methylation across primer sets examined. DNA from the ER-positive MCF-7 cell line was used as an unmethylated control. C, MSP analysis of ER gene CpG island by using primer set ER 5 in a panel of ER-negative breast cancer cells. Both vehicle- and TSA-treated cell pairs (MDA-MB-231 C and TSA; Hs578t C and TSA; and MCF-7/AdrR C and TSA) showed methylated pattern, whereas ER-positive MCF-7 showed unmethylated pattern (MCF 7). Distilled water instead of bisulfite-treated genomic DNA was used as MSP-negative control (H2O). m,methylated products; u, unmethylated products.

Fig. 3.

MSP analysis for the ER CpG island in TSA-treated ER-negative breast cancer cells. A, map of ER CpG island and location of ER MSP primers. B, MSP analysis of ER gene CpG island in TSA-treated MDA-MB-231 cells. Both control (231) and TSA-treated (231 TSA) MDA-MB-231 cells showed evidence of ER CpG methylation across primer sets examined. DNA from the ER-positive MCF-7 cell line was used as an unmethylated control. C, MSP analysis of ER gene CpG island by using primer set ER 5 in a panel of ER-negative breast cancer cells. Both vehicle- and TSA-treated cell pairs (MDA-MB-231 C and TSA; Hs578t C and TSA; and MCF-7/AdrR C and TSA) showed methylated pattern, whereas ER-positive MCF-7 showed unmethylated pattern (MCF 7). Distilled water instead of bisulfite-treated genomic DNA was used as MSP-negative control (H2O). m,methylated products; u, unmethylated products.

Close modal
Fig. 4.

Chromatin conformation at ER gene locus in breast cancer cells analyzed by DNase I sensitivity assay. A, map of the ER promoter region flanked by EcoRI (E), showing the location of the ER CpG island in the region and probe position. B, DNase I digestion profiles with the indicated concentrations of DNase I in TSA-treated (100 ng/ml for 48 h(231TSA) and vehicle-treated (231C)MDA-MB-231 cells are shown. A highly DNase I sensitive region was found in MCF-7 cells (MCF7). Presented is one of three experiments that showed similar results. C, quantitative analysis of DNase I sensitivity at 48 h after TSA treatment in MDA-MB-231 cells. The concentration of DNase I was plotted against the percentage of DNase I resistance. Densitometric analysis demonstrated a 1.8-fold increased sensitivity in TSA-treated (•, 231 TSA, cumulative DNase I sensitivity of 135) versus vehicle-treated (♦,231C, cumulative DNase I sensitivity of 75) MDA-MB-231 cells (see“Materials and Methods”).

Fig. 4.

Chromatin conformation at ER gene locus in breast cancer cells analyzed by DNase I sensitivity assay. A, map of the ER promoter region flanked by EcoRI (E), showing the location of the ER CpG island in the region and probe position. B, DNase I digestion profiles with the indicated concentrations of DNase I in TSA-treated (100 ng/ml for 48 h(231TSA) and vehicle-treated (231C)MDA-MB-231 cells are shown. A highly DNase I sensitive region was found in MCF-7 cells (MCF7). Presented is one of three experiments that showed similar results. C, quantitative analysis of DNase I sensitivity at 48 h after TSA treatment in MDA-MB-231 cells. The concentration of DNase I was plotted against the percentage of DNase I resistance. Densitometric analysis demonstrated a 1.8-fold increased sensitivity in TSA-treated (•, 231 TSA, cumulative DNase I sensitivity of 135) versus vehicle-treated (♦,231C, cumulative DNase I sensitivity of 75) MDA-MB-231 cells (see“Materials and Methods”).

Close modal

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

This work was supported by Grants CA78352 (to N. E. D.) and 2-T32CA09110 (to X. Y. and S. J. N.) from the NIH,Contracts DAMD17-00-1-0301 (to X. Y.) and DAMD17-98-1-8116 (to S. J. N.) from the Department of Defense Breast Cancer Program, and Grant PF4231 (to A. T. F.) from the American Cancer Society.

3

The abbreviations used are: ERα, estrogen receptor α; 5-aza-dC, 5-aza-2′-deoxycytidine; MBD, methyl-CpG-binding domain; Dnmt, DNA methyltransferase; HDAC, histone deacetylase; MeCP2,methyl-CpG binding protein 2; MSP, methylation-specific PCR; TSA,trichostatin A; RT-PCR, reverse transcription-PCR.

1
Hortobagyi G. N. Treatment of breast cancer.
N. Engl. J. Med.
,
339
:
974
-984,  
1998
.
2
Weigel R. J., de Coninck E. C. Transcriptional control of estrogen receptor in estrogen receptor-negative breast carcinoma.
Cancer Res.
,
53
:
3472
-3474,  
1993
.
3
Ottaviano Y. L., Issa J. P., Parl F. F., Smith H. S., Baylin S. B., Davidson N. E. Methylation of the estrogen receptor gene CpG island marks loss of estrogen receptor expression in human breast cancer cells.
Cancer Res.
,
54
:
2552
-2555,  
1994
.
4
Nass S. J., Herman J. G., Gabrielson E., Iversen P. W., Parl F. F., Davidson N. E., Graff J. R. Aberrant methylation of the estrogen receptor and E-cadherin 5′ CpG islands increases with malignant progression in human breast cancer.
Cancer Res.
,
60
:
4346
-4348,  
2000
.
5
Lapidus R. G., Nass S. J., Butash K. A., Parl F. F., Weitzman S. A., Graff J. G., Herman J. G., Davidson N. E. Mapping of ER gene CpG island methylation-specific polymerase chain reaction.
Cancer Res.
,
58
:
2515
-2519,  
1998
.
6
Ferguson A. T., Lapidus R. G., Baylin S. B., Davidson N. E. Demethylation of the estrogen receptor gene in estrogen receptor-negative breast cancer cells can reactivate estrogen receptor gene expression.
Cancer Res.
,
55
:
2279
-2283,  
1995
.
7
Nan X., Ng H. H., Johnson C. A., Laherty C. D., Turner B. M., Eisenman R. N., Bird A. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex.
Nature (Lond.)
,
393
:
386
-389,  
1998
.
8
Wade P. A., Gegonne A., Jones P. L., Ballestar E., Aubry F., Wolffe A. P. Mi-2 complex couples DNA methylation to chromatin remodelling and histone deacetylation.
Nat. Genet.
,
23
:
62
-66,  
1999
.
9
Rountree M. R., Bachman K. E., Baylin S. B. DNMT1 binds HDAC2 and a new co-repressor, DMAP1, to form a complex at replication foci.
Nat. Genet.
,
25
:
269
-277,  
2000
.
10
Robertson K. D., Ait-Si-Ali S., Yokochi T., Wade P. A., Jones P. L., Wolffe A. P. DNMT1 forms a complex with Rb, E2F1 and HDAC1 and represses transcription from E2F-responsive promoters.
Nat. Genet.
,
25
:
338
-342,  
2000
.
11
Yoshida M., Kijima M., Akita M., Beppu T. Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro by trichostatin A.
J. Biol. Chem.
,
265
:
17174
-17179,  
1990
.
12
Cameron E. E., Bachman K. E., Myohanen S., Herman J. G., Baylin S. B. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer.
Nat. Genet.
,
21
:
103
-107,  
1999
.
13
Yang X., Uziely B., Groshen S., Lukas J., Israel V., Russell C., Dunnington G., Formenti S., Muggia F., Press M. F. MDR1 gene expression in primary and advanced breast cancer.
Lab. Investig.
,
79
:
271
-280,  
1999
.
14
Wang S. M., Rowley J. D. A strategy for genome-wide gene analysis: integrated procedure for gene identification.
Proc. Natl. Acad. Sci. USA
,
95
:
11909
-11914,  
1998
.
15
Herman J. G., Graff J. R., Myohanen S., Nelkin B. D., Baylin S. B. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands.
Proc. Natl. Acad. Sci. USA
,
93
:
9821
-9826,  
1996
.
16
Keshet I., Lieman-Hurwitz J., Cedar H. DNA methylation affects the formation of active chromatin.
Cell
,
44
:
535
-543,  
1986
.
17
Ferguson A. T., Lapidus R. G., Davidson N. E. Demethylation of the progesterone receptor CpG island is not required for progesterone receptor gene expression.
Oncogene
,
17
:
577
-583,  
1998
.
18
Luo R. X., Dean D. C. Chromatin remodeling and transcriptional regulation.
J. Natl. Cancer Inst.
,
91
:
1288
-1294,  
1999
.
19
Eden S., Hashimshony T., Keshet I., Cedar H., Thorne A. W. DNA methylation models histone acetylation.
Nature (Lond.)
,
394
:
842
1998
.
20
Chen Z. J., Pikaard C. S. Epigenetic silencing of RNA polymerase I transcription: a role for DNA methylation and histone modification in nucleolar dominance.
Genes Dev.
,
11
:
2124
-2136,  
1997
.
21
Chiurazzi P., Pomponi M. G., Pietrobono R., Bakker C. E., Neri G., Oostra B. A. Synergistic effect of histone hyperacetylation and DNA demethylation in the reactivation of the FMR1 gene.
Hum. Mol. Genet.
,
8
:
2317
-2323,  
1999
.
22
Harvey J. M., Clark G. M., Osborne C. K., Allred D. C. Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer.
J. Clin. Oncol.
,
17
:
1474
-1481,  
1999
.