Hostname: page-component-7c8c6479df-94d59 Total loading time: 0 Render date: 2024-03-29T15:55:49.488Z Has data issue: false hasContentIssue false

Epigenetic modulation of BRCA1 and BRCA2 gene expression by equol in breast cancer cell lines

Published online by Cambridge University Press:  05 January 2012

Rémy Bosviel
Affiliation:
Centre Jean Perrin, Département d'Oncogénétique, CBRV, 28 Place Henri Dunant,BP38, 63001Clermont-Ferrand, France Université d'Auvergne, EA 4233, Nutrition, Cancérogenèse et Thérapie anti-tumorale, 28 Place Henri Dunant, BP38, 63001Clermont-Ferrand, France
Julie Durif
Affiliation:
Centre Jean Perrin, Département d'Oncogénétique, CBRV, 28 Place Henri Dunant,BP38, 63001Clermont-Ferrand, France Université d'Auvergne, EA 4233, Nutrition, Cancérogenèse et Thérapie anti-tumorale, 28 Place Henri Dunant, BP38, 63001Clermont-Ferrand, France
Pierre Déchelotte
Affiliation:
Université d'Auvergne, EA 4233, Nutrition, Cancérogenèse et Thérapie anti-tumorale, 28 Place Henri Dunant, BP38, 63001Clermont-Ferrand, France CHU, Nouvel Hôpital Estaing, Service d'Anatomie Pathologique, 1 Place Lucie Aubrac,63100Clermont-Ferrand, France
Yves-Jean Bignon*
Affiliation:
Centre Jean Perrin, Département d'Oncogénétique, CBRV, 28 Place Henri Dunant,BP38, 63001Clermont-Ferrand, France Université d'Auvergne, EA 4233, Nutrition, Cancérogenèse et Thérapie anti-tumorale, 28 Place Henri Dunant, BP38, 63001Clermont-Ferrand, France
Dominique Bernard-Gallon
Affiliation:
Centre Jean Perrin, Département d'Oncogénétique, CBRV, 28 Place Henri Dunant,BP38, 63001Clermont-Ferrand, France Université d'Auvergne, EA 4233, Nutrition, Cancérogenèse et Thérapie anti-tumorale, 28 Place Henri Dunant, BP38, 63001Clermont-Ferrand, France
*
*Corresponding author: Y.-J. Bignon, email yves-jean.bignon@cjp.fr
Rights & Permissions [Opens in a new window]

Abstract

S-Equol is a metabolite resulting from the conversion of daidzein, a soya phyto-oestrogen, by the gut microflora. The potential protective effects of equol in breast cancer are still under debate. Consequently, we investigated the effects of equol on DNA methylation of breast cancer susceptibility genes (BRCA1 and BRCA2) and oncosuppressors in breast cancer cell lines (MDA-MB-231 and MCF-7) and in a dystrophic breast cell line (MCF-10a) following exposure to S-equol (2 μm) for 3 weeks. We demonstrated by quantitative analysis of methylated alleles a significant decrease in the methylation of the cytosine phosphate guanine (CpG) islands in the promoters of BRCA1 and BRCA2 after the S-equol treatment in MCF-7 and MDA-MB-231 cells and a trend in MCF-10a cells. We also showed that S-equol increases BRCA1 and BRCA2 protein expression in the nuclei and the cytoplasm in MCF-7, MDA-MB-231 and MCF-10a cell lines by immunohistochemistry. The increase in BRCA1 and BRCA2 proteins was also found after Western blotting in the studied cell lines. In summary, we demonstrated the demethylating effect of S-equol on the CpG islands inside the promoters of BRCA1 and BRCA2 genes, resulting in an increase in the level of expressed oncosuppressors in breast cancer cell lines.

Type
Full Papers
Copyright
Copyright © The Authors 2011

First found in equine urine(Reference Marrian and Haslewood1), equol is a non-steroidal oestrogen. Many years after its discovery, it was found that the soya isoflavone daidzein was a precursor to equol(Reference Axelson, Kirk and Farrant2), and that soya consumption increased the excretion of equol in some, but not all, adults. Studies have shown that gut microflora was responsible for the conversion of daidzein to S-equol(Reference Setchell, Borriello and Hulme3). More recently, particular bacteria capable of this conversion were even isolated(Reference Setchell and Clerici4). Multiple studies have shown that equol producers were more frequent in Asian countries than in Western countries, which led researchers to ask themselves whether particular diets would not favour equol-producing microflora(Reference Setchell and Cole5). Equol is a chiral molecule and two forms can coexist: R- and S-equol. Distinction between these two forms and purification of one of them is complex, so many studies have worked on the effects of racemic equol. Today, it has been shown that only S-equol is synthesised by gut bacteria(Reference Setchell, Brzezinski and Brown6) and S-equol is commercially available, leading to studies on the effect of S-equol alone.

Breast cancer is the most frequent cancer in women, with 1·38 million new cases and 458 000 deaths in 2008(Reference Jemal, Bray and Center7). The incidence of breast cancer is high in Western countries, and low in Asia. This difference has been attributed, at least in part, to the Asian traditional diet, containing larger amounts of soya than the Western diet. Particular chemicals in soya, namely phyto-oestrogens, are supposed to have protective effects on breast cancer, mainly because of their similarity of structure with 17-β-oestradiol, the natural human oestrogen, allowing them to bind and activate oestrogen receptors (ER)(Reference Kuiper, Carlsson and Grandien8Reference Pfitscher, Reiter and Jungbauer10), with, contrarily to 17-β-estradiol, a higher affinity for ERβ(Reference Kuiper, Lemmen and Carlsson9, Reference Takeuchi, Takahashi and Sawada11). This is also the case for S-equol(Reference Setchell, Brzezinski and Brown6, Reference Muthyala, Ju and Sheng12). More recently, special attention has been paid to S-equol, as some studies have shown that equol had a greater affinity for ER than its precursor, daidzein(Reference Muthyala, Ju and Sheng12).

Many studies have worked on the potential protective effect of soya over breast cancer, but mixed results have been found(Reference Satih, Rabiau and Bignon13). Studies on the effects of S-equol on breast cancer risk have led to the same mixed results. As epigenetic mechanisms are implied in cancer, a growing number of studies have investigated the effect of soya phyto-oestrogens on those mechanisms, particularly DNA methylation(Reference Fang, Jin and Wang14). In normal tissues, oncogenes and repeated sequences are globally methylated while oncosuppressors are hypomethylated, particularly at the level of cytosine phosphate guanine (CpG) islands found in the promoters of these genes(Reference Das and Singal15). In cancer, an inversion of this methylation profile is found, so it has been stated that soya phyto-oestrogens could have protective effects on cancer by reverting this methylation profile. Moreover, protective effects of breast cancer are observed in women consuming moderate amounts of soya since their childhood but not in women starting soya consumption after the menopause(Reference Guha, Kwan and Quesenberry16, Reference Korde, Wu and Fears17). This observation could be the result of a protective epigenetic effect with expression changes of genes implicated in the early events of carcinogenesis. Some studies have shown a demethylating action of genistein and daidzein on oncosuppressors in cancer cells(Reference Fang, Jin and Wang14). To our knowledge, only one study showed an effect of equol on DNA methylation: Lyn-Cook et al. (Reference Lyn-Cook, Blann and Payne18) showed that high doses of equol caused the hypermethylation of the c-H-ras proto-oncogene in the pancreas cells of neonatal rats. Here, we investigated the effects of equol on the methylation of two major breast cancer oncosuppressors: BRCA1 and BRCA2. The breast cancer susceptibility gene 1 (BRCA1) and the breast cancer susceptibility gene 2 (BRCA2) are the major high-penetrance genes in which mutations increase susceptibility to breast cancer. Mutations in these genes account together for 2–3 % of all breast cancers and about 30–40 % of all familial breast cancers(Reference Wooster and Weber19). The BRCA1 gene is located on chromosome 17q12-21. BRCA1 is involved in many transcriptional activation or transcriptional repression processes(Reference Cable, Wilson and Calzone20). It also plays a role in apoptosis, genomic stability maintenance, and DNA recognition and repair(Reference Jhanwar-Uniyal21). The BRCA2 gene is located on chromosome 13q12-13. The gene codes for proteins involved in DNA repair, cell-cycle control and transcription(Reference Kerr and Ashworth22), and may have a function in the terminal differentiation of breast epithelial cells(Reference Vidarsson, Mikaelsdottir and Rafnar23).

Although somatic mutations of these genes are rarely found in sporadic breast cancers(Reference Kerr and Ashworth22Reference Venkitaraman26), methylation of the promoter of BRCA1 coupled with a decrease in mRNA(Reference Rice, Ozcelik and Maxeiner27) or lower BRCA1 protein(Reference Matros, Wang and Lodeiro28, Reference Tapia, Smalley and Kohen29) can be found. BRCA2 promoter methylation has also been reported in sporadic breast cancer cases(Reference Cucer, Taheri and Ok30).

As a growing number of studies have shown the effects of soya phyto-oestrogens on DNA methylation(Reference Lyn-Cook, Blann and Payne18, Reference Day, Bauer and DesBordes31Reference Majid, Dar and Ahmad35), the protective effects of soya isoflavones on breast cancer could be due, at least in part, to an effect on DNA methylation.

We undertook the present study to examine changes in DNA methylation of the CpG islands in the promoters of BRCA1 and BRCA2 in breast cancer cells following exposure to S-equol at physiological doses during 3 weeks.

Materials and methods

Cell lines

MCF-7 and MDA-MB-231 breast tumour cell lines came from a pleural effusion of patients with invasive breast carcinoma(Reference Cailleau, Young and Olive36, Reference Soule, Vazguez and Long37). The MCF-10a cell line was established from the breast tissue of patients with fibrocystic breast disease(Reference Soule, Maloney and Wolman38). All three human cell lines were provided by the American Type Culture Collection. MCF-7 were cultured in Roswell Park Memorial Institute (RPMI)-1640 medium supplemented with 2 mm-l-glutamine (Invitrogen), gentamycin (20 μg/ml; Panpharma), 10 % fetal bovine serum (Invitrogen) and insulin (1·4 μg/ml; Novo Nordisk) in a humidified atmosphere at 37°C containing 5 % CO2. This cell line has a positive ER status (ERα+/ERβ+). MCF-10a cells were maintained in Dulbecco's modified Eagle's medium F12 (Invitrogen) containing 10 % horse serum (Invitrogen), 2 mm-l-glutamine, gentamycin (20 μg/ml; Panpharma), epidermal growth factor (20 ng/ml; Sigma), cholera toxin (100 ng/ml; Sigma), insulin (10 μg/ml; Novo Nordisk) and hydrocortisone (0·5 μg/ml; Sigma) held at 37°C with 5 % CO2. This cell line has a negative oestrogen receptor status (ERα − /ERβ − ). MDA-MB-231 cells were grown in Leibovitz L-15 medium with 15 % fetal bovine serum (Invitrogen), gentamycin (20 μg/ml; Panpharma) and 2 mm-l-glutamine in a 37°C humidified atmosphere without CO2. This cell line has a negative ER status (ERα − /ERβ+).

The ER status of the three cell lines has previously been confirmed by immunohistochemistry(Reference Vissac-Sabatier, Bignon and Bernard-Gallon39).

Cell treatments

Cells (1 × 106 per T75 flask) were seeded in the medium and treated with 2 μm-S-equol provided by the ENITA Unité Micronutriments-Reproduction-Santé and dissolved in dimethyl sulfoxide. As controls, the cell lines were also conditioned in the medium with the solvent dimethyl sulfoxide.

During the 3 weeks, each 48 h and just before 80 % confluence, cells were trypsinised and cell number scored on a Malassez cell using Trypan blue, and then they were passed into three flasks and the treatments were added again.

DNA extraction

DNA was extracted using Millipore's non-organic DNA extraction kit as follows: after recovering the cells, 9 ml of wash buffer 1 ×  were added to resuspend the pellet. After 15 min of incubation at room temperature, the cells were centrifuged at 1000 g for 20 min. The supernatant was discarded and the cells were resuspended in 3 ml of suspension buffer I 1 × . Lysis buffer I (800 μl) and 50 μl of protein-digesting enzyme were added to the suspension. The samples were incubated for 2 h at 50°C. After adding 1 ml of a protein-precipitating agent, a 15 min centrifugation at 1000 g was carried out. The supernatant thus obtained was mixed with two volumes of absolute ethanol. The precipitated DNA was recovered using an inoculating needle, dried for 5 min at room temperature, and dipped in 5 ml of 70 % ethanol. DNA was resuspended in 300 μl of suspension buffer II. After vortexing them for 5 min, the samples were left in incubation overnight at 50°C. The quantity of DNA collected as well as the quality of the extraction was then determined by spectrometry using a NanoDrop™ 8-sample spectrophotometer (ND-8000, NanoDrop Technologies®).

Bisulfite treatment and quantitative analysis of methylated alleles

Conversion of unmethylated cytosines to uracil(Reference Frommer, McDonald and Millar40), leaving methylated cytosines unaltered, was achieved using the methylSEQr™ Bisulfite Modification Kit (Applied Biosystems) following the manufacturer's instructions. We measured the methylation of oncosuppressor promoters with the real-time PCR-based quantitative analysis of methylated alleles (QAMA) assay previously described by Zeschnigk et al. (Reference Zeschnigk, Bohringer and Price41) and adapted here by Bosviel et al. (Reference Bosviel, Michard and Lavediaux42). PCR was performed using a ninety-six-well optical tray with optical adhesive film at a final reaction volume of 20 μl. Samples contained 10 μl of TaqMan® Universal PCR Master Mix II, No AmpErase® UNG (uracil-N-glycosylase), 8 μl of bisulfite-treated DNA, an additional 5 U of FastStart Taq DNA Polymerase (Roche), 2·5 μm each of the primers and 150 nm of the fluorescently labelled methylated and unmethylated BRCA1 or methylated and unmethylated BRCA2 probes. Initial denaturation at 95°C for 10 min to activate DNA polymerase was followed by forty cycles of denaturation at 95°C for 15 s and annealing and extension at 60°C for 1 min (7900HT, Real-Time PCR System; Applied Biosystems). Primer and probe sequences were selected with the help of Primer Express software (ABI). PCR primers were designed to amplify the bisulfite-converted sense strand of the CpG island BRCA1 promoter sequence or the antisense strand of the CpG island BRCA2 promoter sequence, lacking any known nucleotide polymorphisms. The software designs primers with a melting temperature (T m) of 58–60°C and probes with a T m value of 68–69°C. The T m of both primers should be equal. The amplicon sizes were 79 bp for BRCA1 (located at chromosome 17: 41278096–41278175 on the Ensembl GRCh37/hg19 assembly) and 87 bp for BRCA2 (located at chromosome 13: 32889345–32889428). Primer and probe sequences are as follows: for BRCA1, forward primer – 5′-GGAGTTTGGGGTAAGTAGTTTTGTAAG-3′; reverse primer – 5′-TTCCCCTACCCCAAACAAATT-3′; methylated probe – 5′-VIC-ACTACGTCCCCGCAAA-MGBNFQ-3′; unmethylated probe – 5′-6FAM-ACTACATCCCCACAAAC-MGBNFQ-3′; for BRCA2, forward primer – 5′-GTTGGAGTAAAAAGAAAGGGATGG-3′; reverse primer – 5′-CCTTAAAAATCCCAAACCACCC-3′; methylated probe – 5′-VIC-AAACCGCCCCTATAC-MGBNFQ-3′; unmethylated probe – 5′-6FAM-AAAACCACCCCTATACC-MGBNFQ-3′. The primer binding sites lack CpG dinucleotides and, therefore, the nucleotide sequences in the methylated and unmethylated DNA are identical after the bisulfite treatment. Consequently, it is possible to amplify both alleles in the same reaction tube with one primer pair. Methylation discrimination occurs during probe hybridisation by the use of two different MGB Taqman® probes. The binding site of the BRCA1 and BRCA2 MGB Taqman® probes both cover two CpG dinucleotides. We used a VIC-labelled MGB Taqman® probe that specifically hybridises to the sequence derived from the methylated allele, and a 6-carboxyfluorescein (FAM)-labelled MGB Taqman® probe that binds to the sequence generated from the unmethylated allele. The amount of FAM and VIC fluorescence released during the PCR was measured by the real-time PCR system and is directly proportional to the amount of the PCR product generated. The cycle number at which the fluorescence signal crosses a detection threshold is referred to as C T and the difference of both C T values within a sample (ΔC T) is calculated (ΔC T = C T − FAM − C T − VIC). All samples were measured in duplicate using the mean for further analysis. For a precise quantification of the ratio of methylated:unmethylated alleles, the ΔC T value is determined and compared with a standard curve that exhibits a sigmoid shape with a linear part in the range of 10–90 % of methylated DNA (Fig. 1). To set up the curve, we mixed bisulfite-treated and methylated control human DNA (EpiTect, ref. 59 655; Qiagen) with defined ratios of bisulfite-treated and unmethylated control human DNA (EpiTect, ref. 59 665; Qiagen) implemented in each run. From this, we deduced an algorithm to calculate the methylation ratio of an unknown sample from its ΔC T value by the Mathematica software package version 5.2 from Wolfram Research (http://www.wolfram.com). Student's t test was performed using the data obtained with QAMA, and P < 0·05 was considered to be statistically significant compared with the cells treated with the solvent dimethyl sulfoxide.

Fig. 1 Example of a standard curve for breast cancer susceptibility gene 2 (BRCA2) quantitative analysis of methylated alleles. ΔC T values obtained for standard samples were plotted against their defined methylation ratio. The methylation ratio of the tested samples was found by plotting the ΔC T values obtained onto this standard curve. In the case where only one fluorescence signal crossed the threshold, indicating a relative absence of the opposite target, the methylation percentage was set to 0 or 100 %, depending on the nature of the fluorescence.

Western blotting

Proteins were extracted from the cells with lysis buffer containing 20 mm-Tris (pH 8), 50 mm-EDTA, 0·8 % NaCl, 0·1 % Triton X-100 and 1 % glycerol. Protease inhibitors (1 %, Protease Inhibitor Cocktail; Sigma) and phosphatase inhibitors (1 %, Phosphatase Inhibitor Cocktail 2; Sigma) were added to the basic buffer extemporaneously (1 % each). Then, 50 μg proteins were electrophoresed on a SDS-polyacrylamide gel and transferred onto a nitrocellulose membrane. After 1 h blocking in Tris-Buffered Saline Tween 0·1 % containing 5 % milk, membranes were incubated overnight at 4°C with anti-BRCA1 (1:150 Mouse (Ab-1); Calbiochem), anti-BRCA2 (1:50 Rabbit (H-300); Santa Cruz Biotechnology®) or anti-actin (1:120,000 Mouse (Ab-1); Calbiochem) antibodies. The membranes were then washed three times in Tris-buffered saline Tween and incubated for 1 h with alkaline phosphatase-conjugated secondary antibody (1:2000 goat anti-mouse IgG (H&L) AP conjugate or 1:2000 goat anti-rabbit IgG (Fc) AP conjugate; Promega). Detection was then performed with the Western Blue detection system (Promega). Relative quantification of immunoblotted proteins was achieved using Quantity One software (Bio-Rad) with the local background subtraction method. A ratio between the intensity of the protein of interest and a reference protein (actin) was then calculated. The relative ratio was then calculated between each condition and the reference condition (dimethyl sulfoxide-treated cells).

Immunohistochemistry

For immunohistochemical analysis, 4 μm alcohol–formalin–acetic acid-fixed and paraffin-embedded sections of MCF-7, MDA-MB-231 and MCF-10a cell pellets were cut using a microtome. They were mounted on silanised glass slides (Starfrost; Duiven) and dried overnight at 37°C. Slides were processed on an automated Benchmark XT immunohistochemical instrument (Ventana). In particular, sections were deparaffinised and rehydrated using EZ Prep (Ventana), and heat-induced antigen retrieval using CC1 (Ventana) was performed for 30 min. The slides were then incubated at 37°C for 44 min with anti-BRCA1 (1:20 mouse (8F7); GeneTeX®) or anti-BRCA2 (1:20 mouse (Ab-1); Calbiochem®) primary antibodies. For detection, we used the UltraView universal DAB detection kit (Ventana). Signal was amplified using the Ventana amplification kit. The slides were then counterstained with haematoxylin for 3 min, rinsed in distilled water and coverslipped with an aqueous Faramount mounting media (DAKO). The primary polyclonal antibody was omitted and replaced with PBS as a negative control.

Results

Effect of S-equol on BRCA1 and BRCA2 CpG promoter methylation

QAMA was used to study the effects of S-equol on BRCA1 and BRCA2 CpG islands. We showed a significant decrease in the methylation of the CpG islands in the promoters of BRCA1 and BRCA2 following the 2 μm-S-equol treatment during 3 weeks in MDA-MB-231 and MCF-7 cells compared with the control (Fig. 2(a) and (b), respectively). This demethylation was not significant in MCF-10a cells (Fig. 2(c)).

Fig. 2 Breast cancer susceptibility genes (BRCA1 and BRCA2) methylation in (a) MDA-MB-231, (b) MCF-7 or (c) MCF-10a cells treated for 3 weeks with 2 μm-S-equol compared with the dimethyl sulfoxide (DMSO) control. BRCA1 and BRCA2 methylation were decreased significantly following the S-equol treatment in the MDA-MB-231 and MCF-7 cells (P < 0·05).

Effect of S-equol on BRCA1 and BRCA2 protein expression

Western blotting was used to study the effects of S-equol on BRCA1 and BRCA2 protein expression. We showed an increase in BRCA1 and BRCA2 proteins following the 2 μm-S-equol treatment for 3 weeks in MCF-7, MDA-MB-231 and MCF-10a cell lines (Fig. 3(a)–(c), respectively). An extensive increase in BRCA1 staining was found by immunohistochemistry in the nuclei, the cytoplasm and nucleoli in MCF-7, MDA-MB-231 and MCF-10a cell lines after 2 μm-S-equol exposure for 3 weeks. For BRCA2, the increase in staining was exhibited preferentially in the cytoplasm (Fig. 4). The results of immunohistochemistry are compiled in Table 1.

Fig. 3 Western blots with breast cancer susceptibility genes (BRCA1 and BRCA2) and actin proteins extracted from (a) MCF-7, (b) MDA-MB-231 and (c) MCF-10a cells. Ratios shown correspond to relative ratios of optical densities of the bands (measured with Quantity One software; Bio-Rad) from interest proteins over actin, relatively to the control condition (dimethyl sulfoxide (DMSO)-treated cells). Cells were treated for 3 weeks with DMSO (control condition) or 2 μm-S-equol (E).

Fig. 4 Immunoperoxidase staining of MDA-MB-231 human breast cancer cell lines on paraffin-embedded sections (60 × ). (a) Cytoplasmic, nuclear and nucleolar staining were exhibited with 1:20 breast cancer susceptibility gene 2 (BRCA2) monoclonal antibody (Ab1), shown by arrowheads in untreated cells. (b) The BRCA2 staining after 2 μm-S-equol treatment was considerably increased. N, nucleus; Cyt, cytoplasm; NU, nucleoli.

Table 1 Effects of S-equol on breast cancer susceptibility genes (BRCA1 and BRCA2) expression in MCF-7, MDA-MB-231 and MCF-10a cell lines*

Cyt, cytoplasm; N, nucleus; Nu, nucleoli; DMSO, dimethyl sulfoxide.

* Cells were treated during 3 weeks with S-equol (2 μm). Cells were also treated with DMSO, the solvent in which S-equol was diluted. Then, the cells were immunostained with MoAb anti-BRCA1 (8F7) or anti-BRCA2 (Ab-1). Staining: negative ( − ); intermediate (+/ − ); less intensive (+); intensive (++); very intensive (+++).

Discussion

A growing number of studies have revealed the importance of DNA methylation in cancer, with a global hypomethylation of DNA and the hypermethylation of CpG islands of oncosuppressors, leading to chromosomic instability and loss of the expression of oncosuppressors. In breast cancer, hypermethylation of the BRCA1 and BRCA2 genes has been found, associated with a decrease in mRNA expression for BRCA1. S-equol, an intestinal bacterial metabolite of daidzein, is a putative protective molecule for breast cancer. The present study sustains the idea that this protective effect could pass through epigenetic modulation of BRCA1 and BRCA2 expression. The mechanism for this effect is not yet clearly known, although studies have shown that S-equol can bind and activate ER.

As more and more studies have shown the effects of soya phyto-oestrogens on DNA methylation, we decided to study the effects of S-equol in breast cancer cell lines on BRCA1 and BRCA2 methylation and consequent protein expression. We studied the effects of S-equol on the expression of the BRCA1 and BRCA2 genes that interact together in two human breast cancer cell lines (MCF-7 and MDA-MB-231) and in a fibrocystic cell line (MCF-10a). We chose an exposure of 3 weeks to S-equol, because this treatment has been shown to increase the number of cells blocked in the S phase(Reference Choi and Kim43), and BRCA1 and BRCA2 reach their maximal level in the late G1 and S phases in normal and tumour-derived breast epithelial cells(Reference Bertwistle and Ashworth44).

An important point in the design of the present study is the use of physiological doses of S-equol, in the same order of magnitude as plasma concentrations found in post-menopausal women(Reference Mathey, Lamothe and Coxam45, Reference Bennetau-Pelissero, Arnal-Schnebelen and Lamothe46). Long exposures were carried out to point out an eventually weak effect due to the use of such doses. The effects observed in the present study thus have better chances to be representative of real-life exposure.

We provide evidence that S-equol demethylates the promoters of the BRCA1 and BRCA2 genes in MDA-MB-231 and MCF-7 breast cancer cell lines, but not in the MCF-10a cell line. We also showed an increase in the expression of the BRCA1 and BRCA2 proteins in the studied cell lines following the S-equol treatment. The fact that demethylation occurred in MDA-MB-231 and MCF-7 cell lines but not in the MCF-10a cell line whereas protein expression increased in all the three cell lines could suggest that DNA methylation was not the only mechanism regulating BRCA1 and BRCA2 expression that can be modulated by S-equol, and thus studies on histone mark status following the S-equol treatment could be interesting. Indeed, many studies have shown the effects of soya phyto-oestrogens on histone modifications, and S-equol could have similar effects(Reference Majid, Dar and Ahmad35, Reference Jawaid, Crane and Nowers47Reference Majid, Kikuno and Nelles50). Hong et al. (Reference Hong, Nakagawa and Pan51) also showed that equol stimulates ER-mediated histone acetyl transferase activity. ER status and, more particularly, ERβ status may play a role in the action of S-equol on DNA methylation, as the MCF-10a cell line lacks the ERβ receptor. To our knowledge, only one study has reported an effect of equol on DNA methylation, showing a rise in the methylation of the proto-oncogene c-H-ras in rat pancreatic cells(Reference Lyn-Cook, Blann and Payne18), while more data are found for other soya phyto-oestrogens(Reference Fang, Jin and Wang14, Reference Day, Bauer and DesBordes31, Reference Dolinoy, Weidman and Waterland32, Reference King-Batoon, Leszczynska and Klein34, Reference Majid, Dar and Ahmad35, Reference Majid, Dar and Shahryari49, Reference Berner, Aumuller and Gnauck52Reference Wang and Chen58). Such effects on oncosuppressors could help prevent cancer by restoring their expression similar to the protein expression of BRCA1 and BRCA2 in the present experiment. Studies on whether this demethylating effect is limited to the CpG islands in the promoter of oncosuppressors or whether it also acts on the methylation of other CG sites could be interesting, as demethylating effects on global methylation and, more particularly, repeated elements or transposable elements would be a counter effect for cancer prevention(Reference Watanabe and Maekawa59).

In summary, the present study shows that S-equol has a demethylating effect on the CpG islands in the promoters of BRCA1 and BRCA2 genes. This effect might be linked with the presence of ER but the increase in subsequent protein expression is independent of this parameter. Thus, we suppose that other mechanisms can also be implied, such as effects on histone modifications.

Acknowledgements

We thank Nicolas Sonnier and Christelle Picard for helpful technical assistance. R. B. is the recipient of a grant from the Auvergne Regional Council/CPER 2008+ FEDER no. 32316 – 0930FDBG – 106NL. R. B., P. D., Y.-J. B. and D. B.-G. contributed to the experimental design. R. B. and J. D. were responsible for performing the experiments. R. B., J. D. and P. D. contributed to the data analysis. R. B. and D. B.-G. contributed to manuscript preparation. The authors declare that they have no conflict of interest.

References

1Marrian, GF & Haslewood, GA (1932) Equol, a new inactive phenol isolated from the ketohydroxyoestrin fraction of mares’ urine. Biochem J 26, 12271232.CrossRefGoogle ScholarPubMed
2Axelson, M, Kirk, DN, Farrant, RD, et al. (1982) The identification of the weak oestrogen equol [7-hydroxy-3-(4′-hydroxyphenyl)chroman] in human urine. Biochem J 201, 353357.CrossRefGoogle Scholar
3Setchell, KD, Borriello, SP, Hulme, P, et al. (1984) Nonsteroidal estrogens of dietary origin: possible roles in hormone-dependent disease. Am J Clin Nutr 40, 569578.CrossRefGoogle ScholarPubMed
4Setchell, KD & Clerici, C (2010) Equol: history, chemistry, and formation. J Nutr 140, 1355S1362S.CrossRefGoogle ScholarPubMed
5Setchell, KD & Cole, SJ (2006) Method of defining equol-producer status and its frequency among vegetarians. J Nutr 136, 21882193.CrossRefGoogle ScholarPubMed
6Setchell, KD, Brzezinski, A, Brown, NM, et al. (2005) Pharmacokinetics of a slow-release formulation of soybean isoflavones in healthy postmenopausal women. J Agric Food Chem 53, 19381944.CrossRefGoogle ScholarPubMed
7Jemal, A, Bray, F, Center, MM, et al. (2011) Global cancer statistics. CA Cancer J Clin 61, 6990.CrossRefGoogle ScholarPubMed
8Kuiper, GG, Carlsson, B, Grandien, K, et al. (1997) Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology 138, 863870.CrossRefGoogle ScholarPubMed
9Kuiper, GG, Lemmen, JG, Carlsson, B, et al. (1998) Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology 139, 42524263.CrossRefGoogle ScholarPubMed
10Pfitscher, A, Reiter, E & Jungbauer, A (2008) Receptor binding and transactivation activities of red clover isoflavones and their metabolites. J Steroid Biochem Mol Biol 112, 8794.CrossRefGoogle ScholarPubMed
11Takeuchi, S, Takahashi, T, Sawada, Y, et al. (2009) Comparative study on the nuclear hormone receptor activity of various phytochemicals and their metabolites by reporter gene assays using Chinese hamster ovary cells. Biol Pharm Bull 32, 195202.CrossRefGoogle ScholarPubMed
12Muthyala, RS, Ju, YH, Sheng, S, et al. (2004) Equol, a natural estrogenic metabolite from soy isoflavones: convenient preparation and resolution of R- and Ss-equols and their differing binding and biological activity through estrogen receptors aslpha and beta. Bioorg Med Chem 12, 15591567.CrossRefGoogle Scholar
13Satih, S, Rabiau, N, Bignon, YJ, et al. (2008) Soy phytoestrogens and breast cancer chemoprevention: molecular mechanisms. Curr Nutr Food Sci 4, 259264.CrossRefGoogle Scholar
14Fang, MZ, Jin, Z, Wang, Y, et al. (2005) Promoter hypermethylation and inactivation of O(6)-methylguanine-DNA methyltransferase in esophageal squamous cell carcinomas and its reactivation in cell lines. Int J Oncol 26, 615622.Google Scholar
15Das, PM & Singal, R (2004) DNA methylation and cancer. J Clin Oncol 22, 46324642.CrossRefGoogle ScholarPubMed
16Guha, N, Kwan, ML, Quesenberry, CP, et al. (2009) Soy isoflavones and risk of cancer recurrence in a cohort of breast cancer survivors: the Life After Cancer Epidemiology study. Breast Cancer Res Treat 118, 395405.CrossRefGoogle Scholar
17Korde, LA, Wu, AH, Fears, T, et al. (2009) Childhood soy intake and breast cancer risk in Asian American women. Cancer Epidemiol Biomarkers Prev 18, 10501059.CrossRefGoogle ScholarPubMed
18Lyn-Cook, BD, Blann, E, Payne, PW, et al. (1995) Methylation profile and amplification of proto-oncogenes in rat pancreas induced with phytoestrogens. Proc Soc Exp Biol Med 208, 116119.CrossRefGoogle ScholarPubMed
19Wooster, R & Weber, BL (2003) Breast and ovarian cancer. N Engl J Med 348, 23392347.CrossRefGoogle ScholarPubMed
20Cable, PL, Wilson, CA, Calzone, FJ, et al. (2003) Novel consensus DNA-binding sequence for BRCA1 protein complexes. Mol Carcinog 38, 8596.CrossRefGoogle ScholarPubMed
21Jhanwar-Uniyal, M (2003) BRCA1 in cancer, cell cycle and genomic stability. Front Biosci 8, s1107s1117.CrossRefGoogle ScholarPubMed
22Kerr, P & Ashworth, A (2001) New complexities for BRCA1 and BRCA2. Curr Biol 11, R668R676.CrossRefGoogle ScholarPubMed
23Vidarsson, H, Mikaelsdottir, EK, Rafnar, T, et al. (2002) BRCA1 and BRCA2 bind Stat5a and suppress its transcriptional activity. FEBS Lett 532, 247252.CrossRefGoogle ScholarPubMed
24Lambie, H, Miremadi, A, Pinder, SE, et al. (2003) Prognostic significance of BRCA1 expression in sporadic breast carcinomas. J Pathol 200, 207213.CrossRefGoogle ScholarPubMed
25Lerebours, F & Lidereau, R (2002) Molecular alterations in sporadic breast cancer. Crit Rev Oncol Hematol 44, 121141.CrossRefGoogle ScholarPubMed
26Venkitaraman, AR (2002) Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell 108, 171182.CrossRefGoogle ScholarPubMed
27Rice, JC, Ozcelik, H, Maxeiner, P, et al. (2000) Methylation of the BRCA1 promoter is associated with decreased BRCA1 mRNA levels in clinical breast cancer specimens. Carcinogenesis 21, 17611765.CrossRefGoogle ScholarPubMed
28Matros, E, Wang, ZC, Lodeiro, G, et al. (2005) BRCA1 promoter methylation in sporadic breast tumors: relationship to gene expression profiles. Breast Cancer Res Treat 91, 179186.CrossRefGoogle ScholarPubMed
29Tapia, T, Smalley, SV, Kohen, P, et al. (2008) Promoter hypermethylation of BRCA1 correlates with absence of expression in hereditary breast cancer tumors. Epigenetics 3, 157163.CrossRefGoogle ScholarPubMed
30Cucer, N, Taheri, S, Ok, E, et al. (2008) Methylation status of CpG islands at sites (59 to +96 in exon 1 of the BRCA2 gene varies in mammary tissue among women with sporadic breast cancer. J Genet 87, 155158.CrossRefGoogle ScholarPubMed
31Day, JK, Bauer, AM, DesBordes, C, et al. (2002) Genistein alters methylation patterns in mice. J Nutr 132, 2419S2423S.CrossRefGoogle ScholarPubMed
32Dolinoy, DC, Weidman, JR, Waterland, RA, et al. (2006) Maternal genistein alters coat color and protects Avy mouse offspring from obesity by modifying the fetal epigenome. Environ Health Perspect 114, 567572.CrossRefGoogle ScholarPubMed
33Fang, M, Chen, D & Yang, CS (2007) Dietary polyphenols may affect DNA methylation. J Nutr 137, 223S228S.CrossRefGoogle ScholarPubMed
34King-Batoon, A, Leszczynska, JM & Klein, CB (2008) Modulation of gene methylation by genistein or lycopene in breast cancer cells. Environ Mol Mutagen 49, 3645.CrossRefGoogle ScholarPubMed
35Majid, S, Dar, AA, Ahmad, AE, et al. (2009) BTG3 tumor suppressor gene promoter demethylation, histone modification and cell cycle arrest by genistein in renal cancer. Carcinogenesis 30, 662670.CrossRefGoogle ScholarPubMed
36Cailleau, R, Young, R, Olive, M, et al. (1974) Breast tumor cell lines from pleural effusions. J Natl Cancer Inst 53, 661674.CrossRefGoogle ScholarPubMed
37Soule, HD, Vazguez, J, Long, A, et al. (1973) A human cell line from a pleural effusion derived from a breast carcinoma. J Natl Cancer Inst 51, 14091416.CrossRefGoogle ScholarPubMed
38Soule, HD, Maloney, TM, Wolman, SR, et al. (1990) Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Res 50, 60756086.Google ScholarPubMed
39Vissac-Sabatier, C, Bignon, YJ & Bernard-Gallon, DJ (2003) Effects of the phytoestrogens genistein and daidzein on BRCA2 tumor suppressor gene expression in breast cell lines. Nutr Cancer 45, 247255.CrossRefGoogle ScholarPubMed
40Frommer, M, McDonald, LE, Millar, DS, et al. (1992) A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci U S A 89, 18271831.CrossRefGoogle ScholarPubMed
41Zeschnigk, M, Bohringer, S, Price, EA, et al. (2004) A novel real-time PCR assay for quantitative analysis of methylated alleles (QAMA): analysis of the retinoblastoma locus. Nucleic Acids Res 32, e125.CrossRefGoogle ScholarPubMed
42Bosviel, R, Michard, E, Lavediaux, G, et al. (2011) Peripheral blood DNA methylation detected in the BRCA1 or BRCA2 promoter for sporadic ovarian cancer patients and controls. Clin Chim Acta 412, 14721475.CrossRefGoogle ScholarPubMed
43Choi, EJ & Kim, T (2008) Equol induced apoptosis via cell cycle arrest in human breast cancer MDA-MB-453 but not MCF-7 cells. Mol Med Report 1, 239244.Google Scholar
44Bertwistle, D & Ashworth, A (1998) Functions of the BRCA1 and BRCA2 genes. Curr Opin Genet Dev 8, 1420.CrossRefGoogle ScholarPubMed
45Mathey, J, Lamothe, V, Coxam, V, et al. (2006) Concentrations of isoflavones in plasma and urine of post-menopausal women chronically ingesting high quantities of soy isoflavones. J Pharm Biomed Anal 41, 957965.CrossRefGoogle ScholarPubMed
46Bennetau-Pelissero, C, Arnal-Schnebelen, B, Lamothe, V, et al. (2003) ELISA as a new method to measure genistein and daidzein in food and human fluids. Food Chem 82, 645658.CrossRefGoogle Scholar
47Jawaid, K, Crane, SR, Nowers, JL, et al. (2010) Long-term genistein treatment of MCF-7 cells decreases acetylated histone 3 expression and alters growth responses to mitogens and histone deacetylase inhibitors. J Steroid Biochem Mol Biol 120, 164171.CrossRefGoogle ScholarPubMed
48Kikuno, N, Shiina, H, Urakami, S, et al. (2008) Genistein mediated histone acetylation and demethylation activates tumor suppressor genes in prostate cancer cells. Int J Cancer 123, 552560.CrossRefGoogle ScholarPubMed
49Majid, S, Dar, AA, Shahryari, V, et al. (2010) Genistein reverses hypermethylation and induces active histone modifications in tumor suppressor gene B-Cell translocation gene 3 in prostate cancer. Cancer 116, 6676.CrossRefGoogle ScholarPubMed
50Majid, S, Kikuno, N, Nelles, J, et al. (2008) Genistein induces the p21WAF1/CIP1 and p16INK4a tumor suppressor genes in prostate cancer cells by epigenetic mechanisms involving active chromatin modification. Cancer Res 68, 27362744.CrossRefGoogle ScholarPubMed
51Hong, T, Nakagawa, T, Pan, W, et al. (2004) Isoflavones stimulate estrogen receptor-mediated core histone acetylation. Biochem Biophys Res Commun 317, 259264.CrossRefGoogle ScholarPubMed
52Berner, C, Aumuller, E, Gnauck, A, et al. (2010) Epigenetic control of estrogen receptor expression and tumor suppressor genes is modulated by bioactive food compounds. Ann Nutr Metab 57, 183189.CrossRefGoogle ScholarPubMed
53Guerrero-Bosagna, CM, Sabat, P, Valdovinos, FS, et al. (2008) Epigenetic and phenotypic changes result from a continuous pre and post natal dietary exposure to phytoestrogens in an experimental population of mice. BMC Physiol 8, 17.CrossRefGoogle Scholar
54Qin, W, Zhu, W, Shi, H, et al. (2009) Soy isoflavones have an antiestrogenic effect and alter mammary promoter hypermethylation in healthy premenopausal women. Nutr Cancer 61, 238244.CrossRefGoogle ScholarPubMed
55Tang, WY, Newbold, R, Mardilovich, K, et al. (2008) Persistent hypomethylation in the promoter of nucleosomal binding protein 1 (Nsbp1) correlates with overexpression of Nsbp1 in mouse uteri neonatally exposed to diethylstilbestrol or genistein. Endocrinology 149, 59225931.CrossRefGoogle ScholarPubMed
56Vanhees, K, Coort, S, Ruijters, EJ, et al. (2011) Epigenetics: prenatal exposure to genistein leaves a permanent signature on the hematopoietic lineage. FASEB J 25, 797807.CrossRefGoogle Scholar
57Vardi, A, Bosviel, R, Rabiau, N, et al. (2010) Soy phytoestrogens modify DNA methylation of GSTP1, RASSF1A, EPH2 and BRCA1 promoter in prostate cancer cells. In vivo 24, 393400.Google ScholarPubMed
58Wang, Z & Chen, H (2010) Genistein increases gene expression by demethylation of WNT5a promoter in colon cancer cell line SW1116. Anticancer Res 30, 45374545.Google ScholarPubMed
59Watanabe, Y & Maekawa, M (2010) Methylation of DNA in cancer. Adv Clin Chem 52, 145167.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 Example of a standard curve for breast cancer susceptibility gene 2 (BRCA2) quantitative analysis of methylated alleles. ΔCT values obtained for standard samples were plotted against their defined methylation ratio. The methylation ratio of the tested samples was found by plotting the ΔCT values obtained onto this standard curve. In the case where only one fluorescence signal crossed the threshold, indicating a relative absence of the opposite target, the methylation percentage was set to 0 or 100 %, depending on the nature of the fluorescence.

Figure 1

Fig. 2 Breast cancer susceptibility genes (BRCA1 and BRCA2) methylation in (a) MDA-MB-231, (b) MCF-7 or (c) MCF-10a cells treated for 3 weeks with 2 μm-S-equol compared with the dimethyl sulfoxide (DMSO) control. BRCA1 and BRCA2 methylation were decreased significantly following the S-equol treatment in the MDA-MB-231 and MCF-7 cells (P < 0·05).

Figure 2

Fig. 3 Western blots with breast cancer susceptibility genes (BRCA1 and BRCA2) and actin proteins extracted from (a) MCF-7, (b) MDA-MB-231 and (c) MCF-10a cells. Ratios shown correspond to relative ratios of optical densities of the bands (measured with Quantity One software; Bio-Rad) from interest proteins over actin, relatively to the control condition (dimethyl sulfoxide (DMSO)-treated cells). Cells were treated for 3 weeks with DMSO (control condition) or 2 μm-S-equol (E).

Figure 3

Fig. 4 Immunoperoxidase staining of MDA-MB-231 human breast cancer cell lines on paraffin-embedded sections (60 × ). (a) Cytoplasmic, nuclear and nucleolar staining were exhibited with 1:20 breast cancer susceptibility gene 2 (BRCA2) monoclonal antibody (Ab1), shown by arrowheads in untreated cells. (b) The BRCA2 staining after 2 μm-S-equol treatment was considerably increased. N, nucleus; Cyt, cytoplasm; NU, nucleoli.

Figure 4

Table 1 Effects of S-equol on breast cancer susceptibility genes (BRCA1 and BRCA2) expression in MCF-7, MDA-MB-231 and MCF-10a cell lines*