Open Access

Irreversible inhibition of estrogen receptor α signaling and the emergence of hormonal resistance in MCF7 breast cancer cells induced by DNA damage agents

  • Authors:
    • Alexander M. Scherbakov
    • Danila V. Sorokin
    • Valeria E. Razuvaeva
    • Yuri Yu. Shchegolev
    • Olga E. Andreeva
    • Diana I. Salnikova
    • Timur I. Fetisov
    • Olga A. Vlasova
    • Kirill I. Kirsanov
    • Margarita V. Gudkova
    • Mikhail A. Krasil'nikov
  • View Affiliations

  • Published online on: January 19, 2024     https://doi.org/10.3892/br.2024.1727
  • Article Number: 42
  • Copyright : © Scherbakov et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Combining chemotherapy and hormone therapy is a prevalent approach in breast cancer treatment. While the cytotoxic impact of numerous chemotherapy drugs stems from DNA damage, the exact role of these DNA alterations in modulating estrogen receptor α (ERα) machinery remains elusive. The present study aimed to analyze the impact of DNA damage agents on ERα signaling in breast cancer cells and assess the signaling pathways mediating the influence of DNA damage drugs on the ERα machinery. Cell viability was assessed using the MTT method, while the expression of signaling proteins was analyzed by immunoblotting. ERα activity in the cells treated with various drugs (17β‑estradiol, tamoxifen, 5‑fluorouracil) was assessed through reporter gene assays. In vitro experiments were conducted on MCF7 breast cancer cells subjected to varying durations of 5‑fluorouracil (5‑FU) treatment. Two distinct cell responses to 5‑FU were identified based on the duration of the treatment. A singular dose of 5‑FU induces pronounced DNA fragmentation, temporally suppressing ERα signaling while concurrently activating AKT phosphorylation. This suppression reverses upon 5‑FU withdrawal, restoring normalcy within ten days. However, chronic 5‑FU treatment led to the emergence of 5‑FU‑resistant cells with irreversible alterations in ERα signaling, resulting in partial hormonal resistance. These changes mirror those observed in cells subjected to UV‑induced DNA damage, underscoring the pivotal role of DNA damage in shaping estrogen signaling alterations in breast cancer cells. In summary, the results of the present study suggested that the administration of DNA damage agents to cancer cells can trigger irreversible suppression of estrogen signaling, fostering the development of partial hormonal resistance. This outcome may ultimately impede the efficacy of combined or subsequent chemo‑ and hormone therapy strategies.

Introduction

The role of chemotherapy in the conservative treatment of malignant tumors is pivotal, representing a cornerstone in the therapeutic approach. The main objective in molecular oncology is the exploration of the mechanisms underlying chemotherapy-induced cellular changes and understanding the nature of cell death (1). In the past years, there has been an active exploration for agents and their synergistic combinations tailored to selectively target the pathways responsible for sustaining cancer resistance (2-5). The efficacy of combining different modalities of conservative therapy for breast cancer, especially the tandem use of chemotherapy and hormonal therapy, remains a largely unresolved question. While the impact of numerous chemotherapy drugs is linked to DNA damage, the precise role of these alterations in potentially influencing the estrogen receptor α (ERα) machinery and the hormonal response of tumors remains unclear. Current research in this domain heavily relies on the examination of clinical data, particularly the analysis of combined chemotherapy and hormone therapy effectiveness across diverse patient groups. However, findings in this area are often conflicting and contradictory. Specifically, evidence has revealed that incorporating tamoxifen into chemotherapy cycles enhances outcomes for ERα-positive breast cancer (6-9). Likewise, the combination of hormonal and chemotherapy treatments has been associated with improved survival among women aged over 60 years (10). Conversely, some studies have reported that additional hormonal therapy fails to yield a discernible impact on overall survival (8,11), while supplementary chemotherapy does not demonstrate enhanced outcomes when compared with hormonal therapy alone (12). Several studies have revealed changes in ERα status during neoadjuvant chemotherapy for breast tumors (13,14). Notably, a correlation has been established between neoadjuvant chemotherapy and increased expression of microRNA-18a, a member of the ERα suppressor family (15). The potential role of DNA damage in modulating ERα signaling was underscored in investigations exploring the effects of radiation on breast cancer. These studies revealed notable changes in hormonal signaling within irradiated cells (16,17).

In the present study, it was revealed for the first time that the treatment of MCF7 breast cancer cells with a single dose of 5-fluorouracil (5-FU) induces significant DNA fragmentation, which correlates with a transient suppression of estrogen signaling. Notably, continuous 5-FU treatment leads to the irreversible inhibition of ERα activity and the emergence of partial resistance to the antiestrogen tamoxifen. The pivotal role of DNA damage in altering estrogen signaling was further corroborated through parallel experiments involving ultraviolet-C (UVC)-irradiated cells. These irradiated cells exhibited a pronounced inhibition of estrogen machinery, mirroring the effects observed with 5-FU treatment. Chronic UVC irradiation, akin to prolonged 5-FU exposure, resulted in irreversible changes to estrogen receptor activity and a concomitant reduction in hormonal sensitivity. These findings strongly support the role of DNA damage in driving the progression of hormonal resistance.

Materials and methods

Cell cultures and reagents

Experiments were conducted on the MCF7 human breast cancer cell line (18) (cat. no. HTB-22; American Type Culture Collection), authenticated by morphology and STR profiling through ‘Gordiz’ (http://gordiz.ru/, accessed on February 1, 2022). The cells were cultured at 37˚C with 5% CO2 in DMEM containing 4.5 g/l glucose (cat. no. СC420-02; PanEco), alanyl-glutamine (cat. no. Ф005; PanEco) and 7% fetal bovine serum (FBS) (cat. no. SV30160.03; HyClone; Cytiva). The response of the cells to tamoxifen (cat. no. 27190; Cayman Chemical Company) was assessed by treating them with tamoxifen for 3 days, followed by evaluating viability using the MTT assay [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] (cat. no. A2231; PanReac AppliChem) (19) modified as previously described (20). Dimethyl sulfoxide (DMSO) (cat. no. 191954; PanReac AppliChem) served as the solvent for the assay. Ultrapure water for the experiments was prepared using a Milli-Q water purification system (Merck KGaA).

Treatment of MCF7 cells with 5-FU and the development of resistant clones

MCF7 breast cancer cells were seeded onto 24-well culture plates at a density of 40,000 cells per well. To assess cell sensitivity to 5-FU (cat. no. F6627; Sigma-Aldrich; Merck KGaA), the cells were exposed to 15 µM 5-FU for a 3-day period, followed by an analysis of the number of viable cells. To establish a 5-FU-resistant subline, MCF7 cells (at a density of 150,000 cells per well in 6-well plates) were cultured in DMEM medium with 7% FBS. The cells were exposed to increasing concentrations of 5-FU ranging from 5 to 30 µM over a span of 2 months, and this regimen was maintained for at least 1 month after withdrawal of 5-FU.

UV irradiation and the selection of UV-resistant cells

Irradiation was conducted using 6W UV-lamp, emitting 254 nm light (model VL-6.LC; Vilber Lourmat). MCF7 cells were exposed to UVC irradiation (254 nm) at intensities of 50 J/m2. For the selection of UV-resistant cells, MCF7 cells were exposed to UVC once every three days for a duration of 4 weeks. Subsequently, cell growth was sustained for a minimum of 40 days following the conclusion of the last irradiation cycle.

Colony-forming test

MCF7 cells were initially plated on 60-mm culture dishes at a density of 2 million cells per dish (Corning, Inc.). The following day, the DMEM culture medium was removed, and the seeded cells underwent UV irradiation (254 nm, 3 sec). After UV exposure, varying cell quantities were immediately seeded onto a 6-well culture plate (Corning, Inc.) in DMEM culture medium, aiming to establish 50-2,000 colonies per well. After a 14-day growth in a cell culture incubator, the colonies were fixed and stained using a solution of 20% methanol and 0.2% crystal violet at room temperature for 10 min. Any colony comprising >50 cells was identified and recorded as a viable surviving clone. Colonies were counted manually.

Comet assay

The comet assay was conducted following established procedures outlined in a previous study (21). MCF7 cells were subjected to varying concentrations of 5-FU (15 and 30 µM) for a duration of 72 h or exposed to UVC irradiation, and subsequently, embedded in agarose on microscope slides. Following cell lysis and electrophoresis, the slides were stained with a DNA dye (SYBR Gold) for 5 min at room temperature. Observations were made using a Zeiss AxioVert 200 fluorescence microscope equipped with an EBQ isolated lamp at x10 magnification (Carl Zeiss AG). A minimum of 100 cells were captured for each sample and analyzed using CometScore 2.0 software (RexHoover) to quantify DNA damage.

Micronucleus assay

To inhibit microfilament assembly and cytokinesis, cytochalasin B (cat. no. Х095; PanEco) was introduced into the medium at a final concentration of 6 µg/ml, 28 h prior to fixation across all experimental groups. Following cultivation, cells were collected, centrifuged at 1,200 x g for 10 min, and exposed to 0.075 M KCl (cat. no. 60129-100; PanEco) for 2 min. Subsequently, cells were fixed in ethanol-acetic acid (3:1), followed by another centrifugation at 1,000 x g at 4˚C for 7 min. The fixed cells were then transferred onto clean glass slides. All slides underwent staining with Giemsa solution (cat. no. 0080; PanEco) for 1 min at room temperature. Light microscopic analysis was performed on encrypted preparations, studying 2,000 binuclear cells from each group at a magnification of x400. The significance of differences in cytogenetic damage levels between control and treated cells was determined using Pearson's χ2 test. P<0.05 was considered to indicate a statistically significant difference.

Transient transfection and the measurement of reporter gene activity

The transcriptional activity of ERα was assessed through reporter analysis, involving the transfection of ERE plasmids (luciferase-expressing reporter construct ERE-tk-LUC, which incorporates the estrogen response elements (EREs) from the vitellogenin A2 gene upstream of the thymidine kinase promoter) kindly provided by Professor George Reid from European Molecular Biology Laboratory (Heidelberg, Germany) (22,23). Transfection occurred under steroid-free conditions, utilizing DMEM without phenol red supplemented with 2% charcoal/dextran-treated fetal bovine serum (cat. no. SH30068.03; HyClone, Cytiva). This process was carried out for 6 h at 37˚C using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.). For the transfection of a single well in Costar® 24-well clear TC-treated plate (cat. no. 3524; Corning, Inc.), 0.8 µl of the transfection agent and 0.4 µg of plasmid DNA were employed. Co-transfection with a plasmid carrying the β-galactosidase gene served as a control to assess the efficiency and potential toxicity of the transfection process. 17β-Estradiol (E2) (cat. no. 3301; Sigma-Aldrich; Merck KGaA) in a concentration of 10 nM was used to treat cells during 24 h before determination of the luciferase and β-galactosidase activities. After 24 h post-transfection, cell lysis was carried out in 1x lysis buffer (cat. no. E1531; Promega Corporation), and luciferase activity was quantified using a Tecan Infinite M200 Pro luminometer (Tecan Group), following the manufacturer's protocol (Luciferase Assay System; cat. no. E1501; Promega Corporation) (24,25). β-Galactosidase activity was determined using ONPG (p-nitrophenyl β-D-galactopyranoside) (cat. no. 34055; Thermo Fisher Scientific, Inc.), the substrate for β-galactosidase. The cell lysates were combined with a phosphate buffer (pH 7.5, 0.1 M) containing ONPG (3.3 mM), MgCl2 (1 mM), and β-mercaptoethanol (53 mM). Absorbance at 405 nm was measured using the MultiScan FC reader (Thermo Fisher Scientific, Inc.) ERE reporter activity was calculated in arbitrary units as the luciferase/galactosidase activity ratio, following the method outlined in previous studies (24,25).

Western blot analysis

To prepare samples for immunoblotting, cells were lysed in a buffer (150 µl) comprising Tris-HCl pH 7.4 (50 mM), Igepal CA-630 (1%), ethylenediamine tetraacetate (1 mM), dithiothreitol (1 mM), aprotinin, pepstatin and leupeptin (1 µg/ml), as well as sodium fluoride and sodium orthovanadate (1 mM). Protein content was determined using the Bradford method. Prior to centrifugation (10,000 x g, 10 min, 4˚C), the samples were incubated on ice for 20 min. Electrophoresis was performed on a 10% polyacrylamide gel, loaded with 60 µg of protein per lane, followed by protein transfer to a nitrocellulose membrane (Santa Cruz Biotechnology) and subsequent immunoblotting as described in our previous study (25). The membranes were immersed in a 5% non-fat milk solution (cat. no. A0830,0500; PanReac AppliChem) in TBS buffer with pH 7.5, consisting of Tris (20 mM) and NaCl (500 mM), supplemented with Tween-20 (0.1%) at room temperature over a period of 30 min to prevent non-specific absorption. Subsequently, the membranes were incubated with primary antibodies overnight at 4˚C. The primary antibodies targeting phosphorylated (p)-AKT (cat. no. 9271), AKT (cat. no. 9272), p-Ribosomal Protein S6 Kinase B1(S6K) (cat. no. 9205), S6K (cat. no. 2708) and ERα (cat. no. 8644) (all diluted at 1:1,000; all from Cell Signaling Technology, Inc.) were employed, with antibodies against α-tubulin (1:1,000; cat. no. 2144; Cell Signaling Technology) serving as loading controls. Appropriate IgGs (1:10,000; cat. no. 111-035-003; Jackson ImmunoResearch Europe) conjugated with horseradish peroxidase at room temperature during an hour were used as secondary antibodies. Signal detection was achieved using ECL reagents prepared according to Mruk's protocol (26) by ourselves, and the ImageQuant LAS4000 system for chemiluminescence (GE HealthCare) was utilized. Densitometry for the tested proteins/α-tubulin ratio was carried out using ImageJ 1.53q software (National Institutes of Health). The protocol for densitometry was provided by The University of Queensland, with recommendations from the references (27,28).

Statistical analysis

Each antiproliferative assay was independently replicated three times, with each replication comprising three technical replicates. Statistical analysis was performed using Microsoft Excel 2019 (Microsoft Corp.) and GraphPad 9.0 software (Dotmatics). The IC50 value was calculated to determine the concentration of tamoxifen to produce 50% inhibition of cell growth. The results were presented as the mean ± standard deviation (S.D.), unless otherwise specified. P<0.05 was considered to indicate a statistically significant difference.

Results

5-FU-induced DNA damage

The primary objective of this experiment was to investigate the impact of DNA damage agents on estrogen signaling and the subsequent sensitivity of breast tumors to hormonal therapy. Specifically, the authors focused on 5-FU (29-31), a cytostatic chemotherapeutic drug widely employed in breast cancer treatment. The experiments were conducted on in vitro-cultured MCF7 breast cancer cells. The effectiveness of 5-FU-induced DNA damage was assessed using the DNA fragmentation test, specifically the Comet assay, and by measuring the accumulation of micronuclei in cells as an outcome of DNA disruption. As demonstrated, a single exposure of MCF7 cells to 5-FU resulted in notable DNA fragmentation and the accumulation of micronuclei within cells (Figs. 1A and S1A and B), correlating with a substantial decrease in the number of viable cells (Fig. 1B). To elucidate whether such DNA damage can disrupt ERα signaling and to determine the duration of such alterations, an in-depth analysis of estrogen signaling and the responsiveness to hormone therapy in 5-FU-treated breast cancer cells was conducted.

Influence of 5-FU on ERα signaling and cell response to antiestrogen tamoxifen

MCF7 cells were subjected to a three-day treatment with 5-FU, followed by the assessment of ERα expression and activity. Western blot analysis revealed non-significant changes in ERα expression in 5-FU-treated cells, while reporter analysis of ERα transcriptional activity exhibited a significant suppression following 5-FU exposure. Simultaneously, an activation of AKT, p85 S6K and p70 S6K phosphorylation in 5-FU-treated cells was observed, suggesting a potential compensatory reaction to the inhibition of ERα signaling (Fig. 2A and B). In parallel, the analysis of cell sensitivity to the antiestrogen tamoxifen indicated a decrease in cell sensitivity to the growth inhibitory effects of tamoxifen (Fig. 2C). The IC50 values of tamoxifen were 7.2±0.9 and 12.1±1.3 µM for MCF7 and MCF7/5-FU respectively. Upon withdrawal of 5-FU and the transfer of cells to a standard medium for ten days, there was a notable restoration of ERα transcriptional activity and cell sensitivity to the antiestrogen tamoxifen (Fig. 2D and E). This restoration was concomitant with a reduction in AKT and p85 S6K and p70 S6K phosphorylation levels (Fig. 2F).

Figure 2

5-FU treatment and ERα signaling in MCF7 cells. (A) Western blot analysis of ERα, p-AKT, AKT, p-p85/p-p70 S6K and p85/p70 S6K in cell extracts. The MCF7 cells were treated with 15 µM 5-FU for three days and the cells were subjected to western blotting. Protein loading was controlled by membrane hybridization with α-tubulin antibodies. The blot represents the results of one of three similar experiments. Densitometry for the tested proteins/α-tubulin ratio was carried out using ImageJ software (right diagram). *P<0.05. (B) Reporter analysis of ERα transcriptional activity. The cells were treated with 15 µM 5-FU for three days, then the cells were transfected with the plasmid containing the luciferase reporter gene under estrogen-responsive elements, and β-galactosidase plasmid. The cells were treated with or without 10 nM 17β-estradiol (E2) for 24 h, and the luciferase and β-galactosidase activities were determined. The relative luciferase activity was calculated in arbitrary units as the ratio of luciferase to the β-galactosidase activity. A total of 100 relative units were set as the luciferase activity in MCF7 cells treated with E2. Data represent the mean values ± S.D. of three independent experiments: *P<0.05 vs. untreated samples; #P<0.05 vs. E2-treated MCF7 cells. (C) Cell sensitivity to antiestrogen tamoxifen. The cells were treated with 15 µM 5-FU for three days following 5-FU withdrawal for three days. Then MCF7 cells were treated with 5 µM tamoxifen for three days and the number of viable cells was assessed by the MTT-test. Data represent the mean value ± SD of three independent experiments. Percentage of 100% was set as the viability of untreated cells. *P<0.05 vs. untreated samples. (D) Analysis of luciferase activity in MCF7 cells after 5-FU withdrawal. The cells were treated with 15 µM 5-FU for three days following 5-FU withdrawal for ten days: *P<0.05 vs. untreated samples. (E) The cell response to tamoxifen. *P<0.05 vs. untreated samples. (F) Western blot analysis of ERα, p-AKT, AKT, p-S6K, and S6K in control MCF7 cells and MCF7 cells 10 days after treatment. 5-FU, 5-fluorouracil; ERα, estrogen receptor α; 5-FU, 5-fluorouracil; S6K, Ribosomal Protein S6 Kinase B1; p-, phosphorylated.

Effect of prolonged 5-FU treatment on the ERα machinery

To explore the impact of repeated courses of chemotherapy, the effect of prolonged 5-FU treatment was examined on ERα signaling in MCF7 cells. These cells underwent a two-month treatment with 5-FU, followed by withdrawal of 5-FU and cultivation in standard medium for an additional month. The resulting cell subline, designated as MCF7/FUR, exhibited a notable resistance to 5-FU (Fig. 3A), and significantly, demonstrated marked resistance to antiestrogen tamoxifen (Fig. 3B). The comparative analysis of 5-FU-induced DNA damage revealed a decreased response in the resistant subline to 5-FU treatment (Fig. 3C).

In the analysis of ERα machinery, a suppression of ERα transcriptional activity was evident in 5-FU-resistant cells (Fig. 3D). Subsequent examination of growth-related signaling proteins indicated no significant changes either in ERα expression or in the level of AKT and S6K signaling in the resistant cells (Fig. 3E).

UVC irradiation and ERα signaling

The question of whether the 5-FU-induced suppression of estrogen signaling is a shared event following DNA damage or if these alterations are unique to 5-FU was addressed in the subsequent experiments. The impact of UVC irradiation as a commonly used DNA damage agent was examined on the estrogen signaling of MCF7 cells. The results revealed that UVC irradiation leads to pronounced DNA fragmentation and a reduction in the number of viable cells (Figs. 4A and B and S2A and B), albeit to a different extent compared with the effects observed after 5-FU treatment.

The examination of ERα expression and transcriptional activity in UV-exposed cells revealed a reduction, coupled with the activation of AKT phosphorylation (Fig. 5A and B), no significant changes in the level of S6K phosphorylation were detected. Additionally, a concurrent analysis of the cell response to the growth-inhibitory action of tamoxifen revealed decreased tamoxifen sensitivity in UV-exposed cells, substantiating the suppression of ERα signaling in these cells (Fig. 5C).

The subsequent analysis conducted 30 days after UV irradiation demonstrated a complete restoration of ERα expression and activity, alongside an unchanged level of AKT phosphorylation. This restoration was correlated with the regained sensitivity of cells to tamoxifen (Fig. 6A-C).

Selection and characterization of UV-resistant clones

To explore the impact of continuous UV irradiation on estrogen signaling, MCF7 cells underwent repeated UV exposure once every three days for 4 weeks, followed by the maintenance of cell growth for at least 40 days after the last irradiation. The analysis of UV sensitivity in the selected cells, denoted as MCF7/UVR, revealed a significant increase of cell survival under UV compared with the UV-treated parent MCF7 cells (Fig. 7A). UVC irradiation of MCF7 induced pronounced DNA fragmentation, while no significant difference in DNA damage was observed in MCF7/UVR compared with the untreated control (Fig. 7B).

MCF7/UVR cells exhibited an irreversible reduction in ERα transcriptional activity (Fig. 8A), despite the restored level of ERα expression (Fig. 8B). Examination of the AKT pathway did not reveal changes in the corresponding signaling proteins. The analysis of the cell response to tamoxifen indicated that MCF7/UVR cells retained partial resistance to tamoxifen for at least 40 days after irradiation (Fig. 8C), in contrast to the parent MCF7 cells after a single UV dose.

Discussion

Hormone therapy (32-36) is extensively employed in the treatment of hormone-dependent breast tumors, either as a monotherapy or more frequently in combination with chemotherapy or radiotherapy. While the action of most chemotherapy drugs is linked to DNA damage, the exact role of DNA damage in influencing the estrogen receptor machinery in tumor cells remains unclear.

Several studies have highlighted alterations in the estrogen receptor status of breast tumors following neoadjuvant chemotherapy (13). Additionally, there has been evidence of the overexpression of miRNAs targeting the estrogen receptor after neoadjuvant chemotherapy (15). Furthermore, a correlation has been described between the decreased expression of DNA repair genes and the emergence of hormone resistance in breast cancer cells (37).

UV irradiation (38-41) serves as a widely utilized experimental model for investigating cellular responses to DNA damage treatment. Evidence has been accumulated regarding the influence of UV irradiation on the activity of various cell signaling proteins, including but not limited to p38 MAPK, Jun N-terminal kinase, extracellular signal-regulated kinase 1/2, NF-κB (42,43), eIF2α (44), Toll-like receptors (45), HER2/neu (46), death domain-associated protein (DAXX) (47), and others. In further studies, potential p42/44 ERKs-, AKT- and p38-mediated phosphorylation of ERα in UVC-treated cells is to be investigated. UV irradiation has been indicated to stimulate the bystander effect (48), with corresponding events such as apoptosis, premature senescence, single and double DNA strand breaks, and reduced clonogenic survival described in bystander cells (49).

However, contradictory findings exist, regarding the relationship between DNA damage and hormonal resistance. Various data suggest that radiation-induced DNA damage either does not lead to or is associated with only a marginal increase in overall survival for patients with ERα-negative breast cancer (50,51). In patients with ERα-positive breast cancer, no significant trend in this regard has been consistently identified (52-54). In studies involving in vitro-cultured breast cancer cells, previous studies have revealed a correlation between radiation exposure and disruptions in hormonal cell signaling. These disruptions include a partial loss of ERα and the development of resistance to antiestrogen (16,17). Furthermore, a relationship has been identified between the development of acquired radioresistance and hormonal resistance in breast cancer cells, providing general support for the possibility of impairment in hormonal signaling during irradiation (55-58).

The primary objective of the present study was to explore the impact of DNA damage agents on estrogen signaling and the sensitivity of breast cancer cells to hormonal drugs. The findings of the present study indicated that the response of MCF7 breast cancer cells to 5-FU was linked to alterations in estrogen signaling and the activation of the bypass AKT signaling pathway. A single treatment with 5-FU induces temporary changes in AKT signaling pathways, whereas chronic 5-FU exposure leads to the selection of 5-FU-resistant cells exhibiting irreversible alterations in ERα signaling, correlated with partial hormonal resistance. Similar alterations were observed in cells subjected to UV-induced DNA damage, emphasizing the pivotal role of DNA damage in modifying ERα signaling in breast cancer cells. These observed changes persist in cells for several months after drug treatment, suggesting the potential involvement of (epi)genetic machinery in maintaining the resistant phenotype. ERα and AKT kinase are among the key regulators of breast cancer cell proliferation. Significant efforts of researchers are directed towards the development of novel inhibitors of these targets. The development of such inhibitors also takes into account the significant overlap between signaling pathways. The signaling between ERα and AKT axis largely determines the formation of resistance to targeted and hormonal therapies, and the assessment of these parameters is important for disease prognosis and, in some cases, for changing treatment protocols (59-61). Interestingly, AKT overexpression leads to upregulation of estrogen-regulated pS2 gene, Bcl-2, and macrophage inhibitory cytokine 1(62). Moreover, AKT protects breast cancer cells from tamoxifen-induced apoptosis. The AKT-mediated activation of ERα in 5-FU treated cells has not been described in detail in the present study, and is of great interest for further study, including by means of CRISPR/Cas9 technology.

Additional investigations are required to elucidate the mechanism by which DNA damage agents deactivate estrogen receptors. The present findings suggested that the inhibition of ERα transcriptional activity induced by drugs/UV is not correlated with corresponding changes in ERα expression. This underscores the crucial role of post-translational modifications in the regulation of ERα. The reduction in ERα transcriptional activity may stem from an imbalance between ERα co-activators and corepressors induced by DNA damage agents. Evidence supporting this includes the observed suppression of ERα co-activator CBP/p300 in response to 5-FU (63) and the modulation of ERα coregulator MDC1 (mediator of DNA damage checkpoint 1) in response to DNA damage (64). Similarly, several studies have highlighted the involvement of ERα coregulators (65) and ERα-binding chaperones (66) in the cellular response to irradiation-induced DNA damage agents. Significantly, MCF7 cells are characterized as p53-positive tumor cells, suggesting potential interactions between p53 and ERα signaling. Currently, only few studies describe the interrelation between p53 and ERα, highlighting changes in p53 activity under estrogen stimulation (67,68). MCF7 cells are wtERα and wtp53 positive (69) but the interplay between the two transcription factors in 5-FU and UVC-treated cells has not been investigated in the present study. Additionally, a series of observations underscore the involvement of growth-related pathways, including PI3K/AKT and MAP cascades, in the regulation of ERα activity. Moreover, the role of ERα itself has been implicated in the regulation of cellular radioresistance (58,70,71). Further studies are required for the explanation of the mechanism of the inactivation of ERα by DNA damage agents and how DNA damage inhibits ERα transcription without affecting its expression. In addition, an extension of the study is possible with the use of a tamoxifen gradient, as it is known that the effects of tamoxifen vary greatly depending on the dose used.

In conclusion, the present findings suggested that the treatment of cancer cells with DNA damage agents may lead to the irreversible suppression of estrogen signaling and the progression of partial hormonal resistance, thus limiting the efficiency of combined or subsequent chemo- and hormone therapy.

Supplementary Material

Effects of 5-FU on MCF7 cells. (A) Light microscopy images demonstrating morphology of MCF7 cells after 5-FU treatment [Inverted microscope Diavert (Leitz), phase-contrast objective Phaco x20, camera DP-70 with software DP-controller (Olympus Corporation)]. (B) Comet assay indicating DNA damage in 5-FU-treated MCF7 cells. The slides were observed with a Zeiss AxioVert 200 (Carl Zeiss AG) fluorescence microscope with an EBQ isolated lamp at x10 magnification. 5-FU, 5-fluorouracil.
Effects of UVC on MCF7 cells. (A) Light microscopy images demonstrating morphology of MCF7 cells after UVC exposure. (B) Comet assay indicating DNA damage in MCF7 cells after UVC exposure. The slides were observed with a Zeiss AxioVert 200 fluorescence microscope with an EBQ isolated lamp at x10 magnification. UV, ultraviolet.

Acknowledgements

The plasmids used in the present study were kindly provided by Professor George Reid from European Molecular Biology Laboratory (Heidelberg, Germany). The authors would like to thank Ms. Alexandra A. Drobysheva (Department of Experimental Tumor Biology), Mr. Fedor B. Bogdanov (Department of Experimental Tumor Biology) and Dr Antonina Yu. Alexandrova (Laboratory of Carcinogenesis Mechanisms; all of three are from Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, the Ministry of Health of Russia, Moscow, Russian Federation) for their kind assistance in cell experiments. The authors would also like to thank Ms. Alvina I. Khamidullina (Laboratory of Molecular Oncobiology, Institute of Gene Biology) and the Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, for providing the equipment of research facilities (electrophoresis unit).

Funding

Funding: The present study was supported (grant no. 22-25-00368) by the Russian Scientific Foundation, (M.V.G., https://rscf.ru/project/22-25-00368/, accessed on November 24, 2023).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Authors' contributions

MAK and MVG conceived and designed experiments. MVG managed the project. MAK and AMS wrote original draft. AMS prepared the manuscript and figures for submission. MAK and AMS confirm the authenticity of all the raw data. AMS, MVG, KIK and MAK contributed reagents/materials/analysis tools, analyzed data and critically revised manuscript. DVS, VER, YYS, OEA, DIS, TIF, OAV and KIK performed the experiments, analyzed all data, and amended the revised version. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Tiezzi DG, De Andrade JM, Cândido dos Reis FJ, Marana HR, Ribeiro-Silva A, Tiezzi MG and Pereira AP: Apoptosis induced by neoadjuvant chemotherapy in breast cancer. Pathology. 38:21–27. 2006.PubMed/NCBI View Article : Google Scholar

2 

Khan M, Rasul A, Yi F, Zhong L and Ma T: Jaceosidin induces p53-dependent G2/M phase arrest in U87 glioblastoma cells. Asian Pac J Cancer Prev. 12:3235–3238. 2011.PubMed/NCBI

3 

Rasul A, Khan M, Yu B, Ma T and Yang H: Xanthoxyletin, a coumarin induces S phase arrest and apoptosis in human gastric adenocarcinoma SGC-7901 cells. Asian Pac J Cancer Prev. 12:1219–1223. 2011.PubMed/NCBI

4 

Ding M, Shao Y, Sun D, Meng S, Zang Y, Zhou Y, Li J, Lu W and Zhu S: Design, synthesis, and biological evaluation of BRD4 degraders. Bioorg Med Chem. 78(117134)2023.PubMed/NCBI View Article : Google Scholar

5 

Chrienova Z, Rysanek D, Oleksak P, Stary D, Bajda M, Reinis M, Mikyskova R, Novotny O, Andrys R, Skarka A, et al: Discovery of small molecule mechanistic target of rapamycin inhibitors as anti-aging and anti-cancer therapeutics. Front Aging Neurosci. 14(1048260)2022.PubMed/NCBI View Article : Google Scholar

6 

Davidson NE, O'Neill AM, Vukov AM, Osborne CK, Martino S, White DR and Abeloff MD: Chemoendocrine therapy for premenopausal women with axillary lymph node-positive, steroid hormone receptor-positive breast cancer: Results from INT 0101 (E5188). J Clin Oncol. 23:5973–5982. 2005.PubMed/NCBI View Article : Google Scholar

7 

International Breast Cancer Study Group. Colleoni M, Gelber S, Goldhirsch A, Aebi S, Castiglione-Gertsch M, Price KN, Coates AS and Gelber RD: Tamoxifen after adjuvant chemotherapy for premenopausal women with lymph node-positive breast cancer: International breast cancer study group trial 13-93. J Clin Oncol. 24:1332–1341. 2006.PubMed/NCBI View Article : Google Scholar

8 

Morales L, Canney P, Dyczka J, Rutgers E, Coleman R, Cufer T, Welnicka-Jaskiewicz M, Nortier J, Bogaerts J, Therasse P, et al: Postoperative adjuvant chemotherapy followed by adjuvant tamoxifen versus nil for patients with operable breast cancer: A randomised phase III trial of the European organisation for research and treatment of cancer breast group. Eur J Cancer. 43:331–340. 2007.PubMed/NCBI View Article : Google Scholar

9 

Fukuda M, Yamaguchi S, Ohta T, Nakayama Y, Ogata H, Shimizu K, Nishikawa T, Adachi Y and Fukuma E: Combination therapy for advanced breast cancer: Cyclophosphamide, doxorubicin, UFT, and tamoxifen. Oncology (Williston Park). 13 (7 Suppl 3):S77–S81. 1999.PubMed/NCBI

10 

Hupperets PS, Wils JA, Volovics L, Schouten LJ, Fickers MM, Bron HN, Jager JJ, de Jong JM and Blijham GH: Adjuvant chemo-hormonal therapy with cyclophosphamide, doxorubicin and 5-fluorouracil (CAF) with or without medroxyprogesterone acetate (MPA) for node-positive cancer patients, update at 12 years follow up. Breast. 10:35–37. 2001.PubMed/NCBI View Article : Google Scholar

11 

Gordon NH, Silverman P, Lasheen W, Meinert J and Siminoff LA: Thirty-year follow-up of chemo/hormonal therapy in node-positive breast cancer. Breast Cancer Res Treat. 102:301–312. 2007.PubMed/NCBI View Article : Google Scholar

12 

Alramadhan M, Ryu JM, Rayzah M, Nam SJ, Kim SW, Yu J, Lee SK, Bae SY, Park S, Paik HJ and Lee JE: Goserelin plus tamoxifen compared to chemotherapy followed by tamoxifen in premenopausal patients with early stage-, lymph node-negative breast cancer of luminal A subtype. Breast. 30:111–117. 2016.PubMed/NCBI View Article : Google Scholar

13 

De La Cruz LM, Harhay MO, Zhang P and Ugras S: Impact of neoadjuvant chemotherapy on breast cancer subtype: Does subtype change and, if so, how?: IHC profile and neoadjuvant chemotherapy. Ann Surg Oncol. 25:3535–3540. 2018.PubMed/NCBI View Article : Google Scholar

14 

Zhang N, Moran MS, Huo Q, Haffty BG and Yang Q: The hormonal receptor status in breast cancer can be altered by neoadjuvant chemotherapy: a meta-analysis. Cancer Invest. 29:594–598. 2011.PubMed/NCBI View Article : Google Scholar

15 

Luengo-Gil G, García-Martínez E, Chaves-Benito A, Conesa-Zamora P, Navarro-Manzano E, González-Billalabeitia E, García-Garre E, Martínez-Carrasco A, Vicente V and Ayala de la Peña F: Clinical and biological impact of miR-18a expression in breast cancer after neoadjuvant chemotherapy. Cell Oncol (Dordr). 42:627–644. 2019.PubMed/NCBI View Article : Google Scholar

16 

Paulsen GH, Strickert T, Marthinsen AB and Lundgren S: Changes in radiation sensitivity and steroid receptor content induced by hormonal agents and ionizing radiation in breast cancer cells in vitro. Acta Oncol. 35:1011–1019. 1996.PubMed/NCBI View Article : Google Scholar

17 

Bravatà V, Cava C, Minafra L, Cammarata FP, Russo G, Gilardi MC, Castiglioni I and Forte GI: Radiation-induced gene expression changes in high and low grade breast cancer cell types. Int J Mol Sci. 19(1084)2018.PubMed/NCBI View Article : Google Scholar

18 

Rose HN and McGrath CM: Alpha-lactalbumin production in human mammary carcinoma. Science. 190:673–675. 1975.PubMed/NCBI View Article : Google Scholar

19 

Iselt M, Holtei W and Hilgard P: The tetrazolium dye assay for rapid in vitro assessment of cytotoxicity. Arzneimittelforschung. 39:747–749. 1989.PubMed/NCBI

20 

Volkova YA, Antonov YS, Komkov AV, Scherbakov AM, Shashkov AS, Menchikov LG, Chernoburova LI and Zavarzin IV: Access to steroidal pyridazines via modified thiohydrazides. RSC Adv. 6:42863–42868. 2016.

21 

Singh NP, McCoy MT, Tice RR and Schneider EL: A simple technique for quantitation of low levels of DNA damage in individual cells. Exp Cell Res. 175:184–191. 1988.PubMed/NCBI View Article : Google Scholar

22 

Reid G, Hübner MR, Métivier R, Brand H, Denger S, Manu D, Beaudouin J, Ellenberg J and Gannon F: Cyclic, proteasome-mediated turnover of unliganded and liganded ERalpha on responsive promoters is an integral feature of estrogen signaling. Mol Cell. 11:695–707. 2003.PubMed/NCBI View Article : Google Scholar

23 

Reid G, Métivier R, Lin CY, Denger S, Ibberson D, Ivacevic T, Brand H, Benes V, Liu ET and Gannon F: Multiple mechanisms induce transcriptional silencing of a subset of genes, including oestrogen receptor alpha, in response to deacetylase inhibition by valproic acid and trichostatin A. Oncogene. 24:4894–4907. 2005.PubMed/NCBI View Article : Google Scholar

24 

Scherbakov AM, Komkov AV, Komendantova AS, Yastrebova MA, Andreeva OE, Shirinian VZ, Hajra A, Zavarzin IV and Volkova YA: Steroidal pyrimidines and dihydrotriazines as novel classes of anticancer agents against hormone-dependent breast cancer cells. Front Pharmacol. 8(979)2018.PubMed/NCBI View Article : Google Scholar

25 

Semina SE, Scherbakov AM, Vnukova AA, Bagrov DV, Evtushenko EG, Safronova VM, Golovina DA, Lyubchenko LN, Gudkova MV and Krasil'nikov MA: Exosome-mediated transfer of cancer cell resistance to antiestrogen drugs. Molecules. 23(829)2018.PubMed/NCBI View Article : Google Scholar

26 

Mruk DD and Cheng CY: Enhanced chemiluminescence (ECL) for routine immunoblotting: An inexpensive alternative to commercially available kits. Spermatogenesis. 1:121–122. 2011.PubMed/NCBI View Article : Google Scholar

27 

Taylor SC, Berkelman T, Yadav G and Hammond M: A defined methodology for reliable quantification of Western blot data. Mol Biotechnol. 55:217–226. 2013.PubMed/NCBI View Article : Google Scholar

28 

Butler TAJ, Paul JW, Chan EC, Smith R and Tolosa JM: Misleading Westerns: Common quantification mistakes in western blot densitometry and proposed corrective measures. Biomed Res Int. 2019(5214821)2019.PubMed/NCBI View Article : Google Scholar

29 

Kho MC and Parsonnet V: Observations on the use of 5-fluorouracil in metastatic carcinoma of the breast. J Newark Beth Isr Hosp. 12:93–100. 1961.PubMed/NCBI

30 

Curreri AR and Ansfield FJ: Comparison of 5-fluorouracil and 5-fluoro-2'-deoxyuridine in the treatment of far-advanced breast and colon lesions. Cancer Chemother Rep. 16:387–388. 1962.PubMed/NCBI

31 

Del Mastro L, Poggio F, Blondeaux E, De Placido S, Giuliano M, Forestieri V, De Laurentiis M, Gravina A, Bisagni G, Rimanti A, et al: Fluorouracil and dose-dense adjuvant chemotherapy in patients with early-stage breast cancer (GIM2): End-of-study results from a randomised, phase 3 trial. Lancet Oncol. 23:1571–1582. 2022.PubMed/NCBI View Article : Google Scholar

32 

Bogush TA, Polezhaev BB, Mamichev IA, Bogush EA, Polotsky BE, Tjulandin SA and Ryabov AB: Tamoxifen never ceases to amaze: New findings on non-estrogen receptor molecular targets and mediated effects. Cancer Invest. 36:211–220. 2018.PubMed/NCBI View Article : Google Scholar

33 

Semiglazov VF, Semiglazov VV, Klemtsel AA, Barash NIu, Zhil'tsova EK, Bozhok AA, Mel'nikova OA, Paltuev RM, Dashian GA, Petrovskiĭ SG, et al: New results of endocrine therapy of breast cancer (the role of Aromasin). Vopr Onkol. 50:729–736. 2004.PubMed/NCBI(In Russian).

34 

Lukina SS, Burdennyy AM, Zavarykina TM, Riabchikov DA, Kazubskaya TP, Kruglova MP and Loginov VI: The role of ESR1 gene polymorphic markers in the development of breast cancer and resistance to tamoxifen therapy. Bull Exp Biol Med. 170:350–355. 2021.PubMed/NCBI View Article : Google Scholar

35 

Jordan VC: 50th anniversary of the first clinical trial with ICI 46,474 (tamoxifen): Then what happened? Endocr Relat Cancer. 28:R11–R30. 2021.PubMed/NCBI View Article : Google Scholar

36 

Babyshkina N, Vtorushin S, Dronova T, Patalyak S, Slonimskaya E, Kzhyshkowska J, Cherdyntseva N and Choynzonov E: Impact of estrogen receptor α on the tamoxifen response and prognosis in luminal-A-like and luminal-B-like breast cancer. Clin Exp Med. 19:547–556. 2019.PubMed/NCBI View Article : Google Scholar

37 

Anurag M, Punturi N, Hoog J, Bainbridge MN, Ellis MJ and Haricharan S: Comprehensive profiling of DNA repair defects in breast cancer identifies a novel class of endocrine therapy resistance drivers. Clin Cancer Res. 24:4887–4899. 2018.PubMed/NCBI View Article : Google Scholar

38 

Wang SC, Chang HS, Tang JY, Farooqi AA, Kuo YT, Hsuuw YD, Lee JW and Chang HW: Combined treatment with cryptocaryone and ultraviolet C promotes antiproliferation and apoptosis of oral cancer cells. Int J Mol Sci. 23(2981)2022.PubMed/NCBI View Article : Google Scholar

39 

Chen W, Tang Z, Hu S, Yi X and Wang J: Ultraviolet C irradiation-induced dehybridization of double-stranded oligonucleotides: mechanism investigation and label-free measurement of the photodamage level. Langmuir. 38:15190–15197. 2022.PubMed/NCBI View Article : Google Scholar

40 

Bangruwa N, Srivastava M and Mishra D: CISS-based label-free novel electrochemical impedimetric detection of UVC-induced DNA damage. ACS Omega. 7:37705–37713. 2022.PubMed/NCBI View Article : Google Scholar

41 

Potapovich AI, Kostyuk TV, Shman TV, Ermilova TI, Shutava TG and Kostyuk VA: DNA repair activation and cell death suppression by plant polyphenols in keratinocytes exposed to ultraviolet irradiation. Rejuvenation Res. 26:1–8. 2023.PubMed/NCBI View Article : Google Scholar

42 

Muthusamy V and Piva TJ: The UV response of the skin: A review of the MAPK, NFkappaB and TNFalpha signal transduction pathways. Arch Dermatol Res. 302:5–17. 2010.PubMed/NCBI View Article : Google Scholar

43 

László CF and Wu S: Mechanism of UV-induced IkappaBalpha-independent activation of NF-kappaB. Photochem Photobiol. 84:1564–1568. 2008.PubMed/NCBI View Article : Google Scholar

44 

Lu W, László CF, Miao Z, Chen H and Wu S: The role of nitric-oxide synthase in the regulation of UVB light-induced phosphorylation of the alpha subunit of eukaryotic initiation factor 2. J Biol Chem. 284:24281–24288. 2009.PubMed/NCBI View Article : Google Scholar

45 

Fishelevich R, Zhao Y, Tuchinda P, Liu H, Nakazono A, Tammaro A, Meng TC, Lee J and Gaspari AA: Imiquimod-induced TLR7 signaling enhances repair of DNA damage induced by ultraviolet light in bone marrow-derived cells. J Immunol. 187:1664–1673. 2011.PubMed/NCBI View Article : Google Scholar

46 

Madson JG and Hansen LA: Multiple mechanisms of Erbb2 action after ultraviolet irradiation of the skin. Mol Carcinog. 46:624–628. 2007.PubMed/NCBI View Article : Google Scholar

47 

Shan Z, Liu L, Shen J, Hao H, Zhang H, Lei L, Liu F and Wang Z: Enhanced UV resistance role of death domain-associated protein in human MDA-MB-231 breast cancer cells by regulation of G2 DNA damage checkpoint. Cell Transplant. 29(963689720920277)2020.PubMed/NCBI View Article : Google Scholar

48 

Wu S, Jin C, Lu X, Yang J, Liu Q, Qi M, Lu S, Zhang L and Cai Y: Bystander effect induced by UVC radiation in Chinese hamster V79 cells. Photochem Photobiol. 90:837–844. 2014.PubMed/NCBI View Article : Google Scholar

49 

Krzywon A and Widel M: Bystander Me45 melanoma cells increase damaging effect in UVC-irradiated cells. Photochem Photobiol. 95:1019–1028. 2019.PubMed/NCBI View Article : Google Scholar

50 

Eaton BR, Jiang R, Torres MA, Kahn ST, Godette K, Lash TL and Ward KC: Benefit of adjuvant radiotherapy after breast-conserving therapy among elderly women with T1-T2N0 estrogen receptor-negative breast cancer. Cancer. 122:3059–3068. 2016.PubMed/NCBI View Article : Google Scholar

51 

Yang PS, Chen CM, Liu MC, Jian JM, Horng CF, Liu MJ, Yu BL, Lee MY and Chi CW: Radiotherapy can decrease locoregional recurrence and increase survival in mastectomy patients with T1 to T2 breast cancer and one to three positive nodes with negative estrogen receptor and positive lymphovascular invasion status. Int J Radiat Oncol Biol Phys. 77:516–522. 2010.PubMed/NCBI View Article : Google Scholar

52 

Chen JX, Zhang WW, Dong Y, Sun JY, He ZY and Wu SG: The effects of postoperative radiotherapy on survival outcomes in patients under 65 with estrogen receptor positive tubular breast carcinoma. Radiat Oncol. 13(226)2018.PubMed/NCBI View Article : Google Scholar

53 

Hughes KS, Schnaper LA, Berry D, Cirrincione C, McCormick B, Shank B, Wheeler J, Champion LA, Smith TJ, Smith BL, et al: Lumpectomy plus tamoxifen with or without irradiation in women 70 years of age or older with early breast cancer. N Engl J Med. 351:971–977. 2004.PubMed/NCBI View Article : Google Scholar

54 

Chesney TR, Yin JX, Rajaee N, Tricco AC, Fyles AW, Acuna SA and Scheer AS: Tamoxifen with radiotherapy compared with Tamoxifen alone in elderly women with early-stage breast cancer treated with breast conserving surgery: A systematic review and meta-analysis. Radiother Oncol. 123:1–9. 2017.PubMed/NCBI View Article : Google Scholar

55 

Steelman LS, Navolanic P, Chappell WH, Abrams SL, Wong EW, Martelli AM, Cocco L, Stivala F, Libra M, Nicoletti F, et al: Involvement of Akt and mTOR in chemotherapeutic- and hormonal-based drug resistance and response to radiation in breast cancer cells. Cell Cycle. 10:3003–3015. 2011.PubMed/NCBI View Article : Google Scholar

56 

Jang H, Baek J, Nam KS and Kim S: Determination of the optimal time for tamoxifen treatment in combination with radiotherapy. Int J Oncol. 49:2147–2154. 2016.PubMed/NCBI View Article : Google Scholar

57 

Luzhna L, Lykkesfeldt AE and Kovalchuk O: Altered radiation responses of breast cancer cells resistant to hormonal therapy. Oncotarget. 6:1678–1694. 2015.PubMed/NCBI View Article : Google Scholar

58 

Semina SE, Scherbakov AM, Kovalev SV, Shevchenko VE and Krasil'nikov MA: Horizontal transfer of tamoxifen resistance in MCF-7 cell derivates: Proteome study. Cancer Invest. 35:506–518. 2017.PubMed/NCBI View Article : Google Scholar

59 

Khatpe AS, Adebayo AK, Herodotou CA, Kumar B and Nakshatri H: Nexus between PI3K/AKT and estrogen receptor signaling in breast cancer. Cancers (Basel). 13(369)2021.PubMed/NCBI View Article : Google Scholar

60 

Golden E, Rashwan R, Woodward EA, Sgro A, Wang E, Sorolla A, Waryah C, Tie WJ, Cuyàs E, Ratajska M, et al: The oncogene AAMDC links PI3K-AKT-mTOR signaling with metabolic reprograming in estrogen receptor-positive breast cancer. Nat Commun. 12(1920)2021.PubMed/NCBI View Article : Google Scholar

61 

du Rusquec P, Blonz C, Frenel JS and Campone M: Targeting the PI3K/Akt/mTOR pathway in estrogen-receptor positive HER2 negative advanced breast cancer. Ther Adv Med Oncol. 12(1758835920940939)2020.PubMed/NCBI View Article : Google Scholar

62 

Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S and Nakshatri H: Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: A new model for anti-estrogen resistance. J Biol Chem. 276:9817–9824. 2001.PubMed/NCBI View Article : Google Scholar

63 

Du C, Huang D, Peng Y, Yao Y, Zhao Y, Yang Y, Wang H, Cao L, Zhu WG and Gu J: 5-Fluorouracil targets histone acetyltransferases p300/CBP in the treatment of colorectal cancer. Cancer Lett. 400:183–193. 2017.PubMed/NCBI View Article : Google Scholar

64 

Zou R, Zhong X, Wang C, Sun H, Wang S, Lin L, Sun S, Tong C, Luo H, Gao P, et al: MDC1 enhances Estrogen receptor-mediated transactivation and contributes to breast cancer suppression. Int J Biol Sci. 11:992–1005. 2015.PubMed/NCBI View Article : Google Scholar

65 

Chen J, Liu J, Zeng P, Zhao C, Liu X, Sun J, Wang J, Fang P, Chen W and Ding J: Estrogen and BRCA1 deficiency synergistically induce breast cancer mutation-related DNA damage. Biochem Biophys Res Commun. 613:140–145. 2022.PubMed/NCBI View Article : Google Scholar

66 

Ashrafizadeh M, Farhood B, Eleojo Musa A, Taeb S and Najafi M: Damage-associated molecular patterns in tumor radiotherapy. Int Immunopharmacol. 86(106761)2020.PubMed/NCBI View Article : Google Scholar

67 

Thibodeau PA, Bissonnette N, Bédard SK, Hunting D and Paquette B: Induction by estrogens of methotrexate resistance in MCF-7 breast cancer cells. Carcinogenesis. 19:1545–1552. 1998.PubMed/NCBI View Article : Google Scholar

68 

Molinari AM, Bontempo P, Schiavone EM, Tortora V, Verdicchio MA, Napolitano M, Nola E, Moncharmont B, Medici N, Nigro V, et al: Estradiol induces functional inactivation of p53 by intracellular redistribution. Cancer Res. 60:2594–2597. 2000.PubMed/NCBI

69 

Pairawan S, Zhao M, Yuca E, Annis A, Evans K, Sutton D, Carvajal L, Ren JG, Santiago S, Guerlavais V, et al: First in class dual MDM2/MDMX inhibitor ALRN-6924 enhances antitumor efficacy of chemotherapy in TP53 wild-type hormone receptor-positive breast cancer models. Breast Cancer Res. 23(29)2021.PubMed/NCBI View Article : Google Scholar

70 

Paramanantham A, Jung EJ, Go SI, Jeong BK, Jung JM, Hong SC, Kim GS and Lee WS: Activated ERK signaling is one of the major hub signals related to the acquisition of radiotherapy-resistant MDA-MB-231 breast cancer cells. Int J Mol Sci. 22(4940)2021.PubMed/NCBI View Article : Google Scholar

71 

Sun XY, Li HZ, Xie DF, Gao SS, Huang X, Guan H, Bai CJ and Zhou PK: LPAR5 confers radioresistance to cancer cells associated with EMT activation via the ERK/Snail pathway. J Transl Med. 20(456)2022.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-2024
Volume 20 Issue 3

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Scherbakov AM, Sorokin DV, Razuvaeva VE, Shchegolev YY, Andreeva OE, Salnikova DI, Fetisov TI, Vlasova OA, Kirsanov KI, Gudkova MV, Gudkova MV, et al: Irreversible inhibition of estrogen receptor α signaling and the emergence of hormonal resistance in MCF7 breast cancer cells induced by DNA damage agents. Biomed Rep 20: 42, 2024
APA
Scherbakov, A.M., Sorokin, D.V., Razuvaeva, V.E., Shchegolev, Y.Y., Andreeva, O.E., Salnikova, D.I. ... Krasil'nikov, M.A. (2024). Irreversible inhibition of estrogen receptor α signaling and the emergence of hormonal resistance in MCF7 breast cancer cells induced by DNA damage agents. Biomedical Reports, 20, 42. https://doi.org/10.3892/br.2024.1727
MLA
Scherbakov, A. M., Sorokin, D. V., Razuvaeva, V. E., Shchegolev, Y. Y., Andreeva, O. E., Salnikova, D. I., Fetisov, T. I., Vlasova, O. A., Kirsanov, K. I., Gudkova, M. V., Krasil'nikov, M. A."Irreversible inhibition of estrogen receptor α signaling and the emergence of hormonal resistance in MCF7 breast cancer cells induced by DNA damage agents". Biomedical Reports 20.3 (2024): 42.
Chicago
Scherbakov, A. M., Sorokin, D. V., Razuvaeva, V. E., Shchegolev, Y. Y., Andreeva, O. E., Salnikova, D. I., Fetisov, T. I., Vlasova, O. A., Kirsanov, K. I., Gudkova, M. V., Krasil'nikov, M. A."Irreversible inhibition of estrogen receptor α signaling and the emergence of hormonal resistance in MCF7 breast cancer cells induced by DNA damage agents". Biomedical Reports 20, no. 3 (2024): 42. https://doi.org/10.3892/br.2024.1727