Mechanisms underlying differential response to estrogen-induced apoptosis in long-term estrogen-deprived breast cancer cells

  • Authors:
    • Elizabeth E. Sweeney
    • Ping Fan
    • V. Craig Jordan
  • View Affiliations

  • Published online on: March 6, 2014     https://doi.org/10.3892/ijo.2014.2329
  • Pages: 1529-1538
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Models of long-term estrogen-deprived breast cancer cells are utilized in the laboratory to mimic clinical aromatase inhibitor-resistant breast cancer and serve as a tool to discover new therapeutic strategies. The MCF-7:5C and MCF-7:2A subclones were generated through long-term estrogen deprivation of estrogen receptor (ER)-positive MCF-7 cells, and represent anti-hormone‑resistant breast cancer. MCF-7:5C cells paradoxically undergo estrogen-induced apoptosis within seven days of estrogen (estradiol, E2) treatment; MCF-7:2A cells also experience E2-induced apoptosis but evade dramatic cell death until approximately 14 days of treatment. To discover and define the mechanisms by which MCF-7:2A cells survive two weeks of E2 treatment, systematic experiments were performed in this study. The data suggest that MCF-7:2A cells employ stronger antioxidant defense mechanisms than do MCF-7:5C cells, and that oxidative stress is ultimately required for MCF-7:2A cells to die in response to E2 treatment. Tumor necrosis factor (TNF) family member activation is also essential for E2-induced apoptosis to occur in MCF-7:2A cells; upregulation of TNFα occurs simultaneously with oxidative stress activation. Although the unfolded protein response (UPR) signaling pattern is similar to that in MCF-7:5C cells, it is not sufficient to cause cell death in MCF-7:2A cells. Additionally, increased insulin-like growth factor receptor β (IGF-1Rβ) confers a mechanism of growth and anti-apoptotic advantage in MCF-7:2A cells.

Introduction

Aromatase inhibitor-resistant breast cancer cells are modeled in vitro by long-term E2-deprived breast cancer cell lines. The MCF-7:WS8 cell line represents a clone of the estrogen receptor (ER)-positive cell line MCF-7 that is highly sensitive to E2-stimulated growth (1). The MCF-7:5C and MCF-7:2A subclones are derived from the parental MCF-7 cell line through long-term E2 deprivation (14). MCF-7:5C cells express wild-type ER at a higher level than the parental line, and are progesterone receptor (PR)-negative (3). These cells grow in the absence of E2, and do not respond to 4-hydroxytamoxifen (4-OHT) (2,3). MCF-7:2A cells can induce expression of PR and express both wild-type (66 kDa) and mutant (77 kDa) ER (4,5). The mutant ER contains a repeat of exons 6 and 7 and cannot bind E2 nor anti-estrogens; it is expressed 4- to 10-fold lower than the wild-type ER (6). The total ER level of MCF-7:2A cells is higher than in parental MCF-7 cells, and they also grow in E2-free media. 4-OHT and pure anti-E2 are able to block their growth (4,5).

In addition to the different responses to anti-E2 observed in MCF-7:5C versus MCF-7:2A cells, they also have different apoptotic responses to E2. The MCF-7:5C cells undergo apoptosis and die during the first week of E2 treatment, whereas the MCF-7:2A cells die later, after two weeks of E2 treatment (7). MCF-7:5C cell response to estrogens and anti-estrogens has been extensively studied in our lab; the data show that these cells undergo E2-induced apoptosis through mechanisms associated with endoplasmic reticulum stress (ERS) and oxidative stress (8,9). Thus far, there has been less focus on the classification and mechanisms of the MCF-7:2A response.

Network enrichment analyses done using gene arrays in timecourse experiments show overexpression of apoptotic- and stress-related pathways in the MCF-7:5C cells after 24–96 h of E2 treatment; however, these analyses show the MCF-7:2A cells expressing more genes associated with glutathione metabolism during this time period of E2 exposure (Fig. 1). This suggests that the two cell lines respond to E2 treatment using different signaling pathways. The MCF-7:5C cells respond by quickly inducing apoptosis, while the anti-oxidant pathway may be more relevant to the MCF-7:2A cells. Experiments were designed to interrogate the apoptotic, stress and antioxidant pathways in both cell lines to distinguish signaling mechanisms in response to E2.

The concept of E2-induced death is important because of its clinical relevance. A clinical study published in 2009 (10) compared two doses of E2 for second-line treatment after breast cancer patients had failed aromatase inhibitor therapy. The authors showed that after long-term anti-hormone therapy, no response is lost with the lower dose of E2; overall about 30% of women responded to E2 treatment. The goal of this study is to uncover the mechanisms preventing the other 70% of patients from responding, and perhaps find ways to circumvent their resistance. To this end, MCF-7:2A cells were used as a model for E2-deprived breast tumors with the ability to evade E2-induced apoptosis in the clinic.

Materials and methods

Cell culture

All cell lines were cultured in phenol red-free RPMI-1640 media supplemented with 10% charcoal-stripped fetal bovine serum (SFS). Media and treatments were replaced every three days. Estradiol (E2) (Sigma-Aldrich, St. Louis, MO, USA), buthionine sulfoximine (BSO) (Sigma-Aldrich), and combinations were dissolved in ethanol and then in media. AG1024 (Calbiochem, San Diego, CA, USA) was dissolved in DMSO and then in media.

DNA assays

MCF-7:WS8, MCF-7:5C and MCF-7:2A cells were harvested after 7 or 14 days treatment with vehicle (0.1% ethanol), E2 (10−9 mol/l, 1 nM), BSO (10−4 mol/l, 100 μM), or E2 (1 nM) + BSO (100 μM). DNA content was measured as previously described (11).

Western blot analysis

Total MAPK (#9102), phosphorylated MAPK (#9101), total AKT (#9272), phosphorylated AKT (#4051L), total eIF2α (#9722S), phosphorylated eIF2α (#9721S), and IRE1α (#3294S) antibodies were all purchased from Cell Signaling Technology (Beverly, MA, USA). IGF-1Rβ antibody (sc-713) was purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). β-actin loading control antibody (A5441) was purchased from Sigma-Aldrich. Proteins were harvested from cells using cell lysis buffer (Cell Signaling Technology) supplemented with Protease Inhibitor Cocktail Set I and Phosphatase Inhibitor Cocktail Set II (Calbiochem). Bicinchoninic acid (BCA) assay was used to quantify total protein content (Rio-Rad Laboratories, Hercules, CA, USA). Protein (50 μg) was probed and visualized as previously described (11).

Cell cycle analysis

MCF-7:2A cells were cultured in dishes and treated with vehicle (0.1% ethanol) or E2 (10−9 mol/l, 1 nM). Cells were harvested after 24 h, fixed in 75% ethanol on ice, stained with propidium iodide and sorted using FACS flow cytometry (Becton Dickinson, San Jose, CA, USA). Results were analyzed using CellQuest software.

RT-PCR

Cells were harvested using TRIzol, and RNA was isolated using RNeasy mini kit (Qiagen, Valencia, CA, USA). RNA was reverse transcribed to cDNA using a kit (Applied Biosystems, Foster City, CA). SYBR-Green (Applied Biosystems) was used for quantitative real-time polymerase chain reaction (RT-PCR) in a 7900HT Fast Real-Time PCR system (Applied Biosystems).

Glutathione assay

Cells were harvested and de-proteinized with 5% 5-sulfosalicylic acid solution (SSA) (Sigma-Aldrich). Total glutathione [reduced glutathione (GSH) plus glutathione disulfide (GSSG)] was measured spectroscopically at 412 nm using a Glutathione Assay Kit (CS0260, Sigma-Aldrich) and the manufacturer’s instructions.

ROS assay

MCF-7:2A cells were harvested, stained with 10−6 mol/l (1 μM) CM-H2DCFDA (Invitrogen, Eugene, OR, USA), and analyzed for ROS fluorescence using flow cytometry.

Statistical analysis

Values reported are means ± standard deviation (SD). Significant differences were found by Student’s t-test. P-values <0.05 were considered to indicate a statistically significant difference.

Results

MCF-7:2A initial response to E2

The MCF-7:WS8, MCF-7:5C and MCF-7:2A cell lines respond differently to 10−9 mol/l (1 nM) E2. In the presence of 1 nM E2, MCF-7:WS8 cells are stimulated to proliferate over 7 days, whereas MCF-7:5C cells are killed by this time point (Fig. 2A). MCF-7:2A cell growth is unaffected by the presence of E2 after one week, but their DNA is reduced by 50% after the second week of treatment (Fig. 2A). Interestingly, MCF-7:2A cells are initially stimulated to proliferate in response to E2. After 24 h-treatment with 1 nM E2, both the mitogen-activated protein kinase (MAPK) and serine/threonine protein kinase Akt (AKT) pathways are activated, as shown by an increase in phosphorylated MAPK (p-MAPK) and phosphorylated AKT (p-AKT) proteins, respectively (Fig. 2B). Further, MCF-7:2A cells treated with E2 for 24 h show an increase in the percentage of dividing cells compared with vehicle treatment (34.78 versus 20.17%), illustrated by S-phase in cell cycle analysis (Fig. 2C).

MCF-7:5C and MCF-7:2A UPR

To determine whether the different biological effects observed in MCF-7:5C and MCF-7:2A cells is due to different patterns of the unfolded protein response (UPR), proteins associated with the UPR were measured over a 72 h timecourse. Two markers of the UPR, phosphorylated eIF2α (p-eIF2α) and IRE1α, were visualized by western blot analysis in MCF-7:5C and MCF-7:2A cells in the presence of vehicle and 1 nM E2 (Fig. 3). p-eIF2α is directly downstream of protein kinase RNA-like endoplasmic reticulum kinase (PERK), a sensor which initiates UPR. Both cell lines show an increase in the protein expression of p-eIF2α and IRE1α by 72 h of E2 treatment, indicating activated UPR. Though MCF-7:2A cells show a slightly higher basal p-eIF2α level, no differences in UPR activation can be seen between the two cell lines.

MCF-7:5C and MCF-7:2A estrogen-induced apoptosis

To determine whether MCF-7:2A cells experience apoptosis through the same mechanism as MCF-7:5C cells, RT-PCR was used to quantify mRNA levels of apoptosis-related genes. MCF-7:5C cells noticeably upregulate LTA (4.19±1.92 fold change), LTB (5.39±1.82), TNFα (9.40±3.86), and BCL2L11 (6.06±0.87) after 72 h of E2 treatment, while MCF-7:2A cells show no major changes during this time period (Fig. 4A). MCF-7:2A cells were then treated with E2 for a longer time period to measure apoptosis-related genes during the time when they appear to die. MCF-7:2A cells increase both TNFα (33.55±12.09 fold change) and BCL2L11 (3.71±0.35 fold change) after 12 days of 1 nM E2 treatment (Fig. 4B). The upregulated apoptosis-related genes correspond to the time when cell death is most apparent in both cell lines, during week one in MCF-7:5C cells, and during week two in MCF-7:2A cells.

MCF-7:5C and MCF-7:2A oxidative stress

Heme oxygenase 1 (HMOX1) was used as an indicator to illustrate when MCF-7:5C and MCF-7:2A cells experience oxidative stress. After 72 h of 1 nM E2 treatment, HMOX1 mRNA was increased 4.61-fold in MCF-7:5C cells (Fig. 5A), suggesting this cell line undergoes oxidative stress at this time point. MCF-7:2A cells did not generate an upregulation of HMOX1 mRNA until 12 days of 1 nM E2 treatment when it increased 10.03-fold (Fig. 5B), suggesting an earlier protective mechanism inherent in these cells to prevent oxidative stress longer than MCF-7:5C cells.

Glutathione is a potent antioxidant and was quantified in MCF-7:5C and MCF-7:2A cells to illustrate a potential protective mechanism in MCF-7:2A cells against oxidative stress (Fig. 6A). In fact, MCF-7:2A cells have significantly more basal glutathione than do MCF-7:WS8 and MCF-7:5C cells (Fig. 6A). Buthionine sulfoximine (BSO) is a synthetic amino acid that blocks glutathione synthesis by inhibiting γ-glutamylcysteine synthetase. BSO (100 μM) dramatically decreases glutathione levels in both MCF-7:5C and MCF-7:2A cells (Fig. 6B). To ask the question of whether glutathione is protecting MCF-7:2A cells from oxidative stress and E2-induced apoptosis, HMOX1 was measured following treatment with vehicle, 1 nM E2 alone, 100 μM BSO alone, and 1 nM E2 + 100 μM BSO after 24, 48 and 72 h (Fig. 6C). MCF-7:2A cells show increased HMOX1 mRNA at 72 h after treatment with 100 μM BSO and 1 nM E2 + 100 μM BSO (3.57±0.36 and 2.60±0.70 fold change, respectively), suggesting a protective role of glutathione in these cells. Reactive oxygen species (ROS) increased 634% over vehicle in MCF-7:2A cells after 12 days of the combination treatment (Fig. 6D). Furthermore, 1 nM E2 + 100 μM BSO treatment caused a significant decrease in DNA after 14 days treatment (Fig. 6E), suggesting that oxidative stress is a key factor in determining E2-induced MCF-7:2A cell death.

MCF-7:5C and MCF-7:2A IGFR

Insulin-like growth factor receptor β (IGF-1Rβ) upregulation is another mechanism through which MCF-7:2A cells could receive anti-apoptotic advantage over MCF-7:5C cells. MCF-7:2A cells exhibit 2.71-fold greater basal IGF-1Rβ mRNA than MCF-7:5C cells (Fig. 7A). This is consistent at the protein level as shown by western blot analysis, where MCF-7:2A cells exhibit more IGF-1Rβ protein expression than MCF-7:5C cells (Fig. 7B). When treated with an IGF-1Rβ inhibitor (10 μM AG1024) for 7 days, MCF-7:2A cells show significantly decreased DNA content when compared to vehicle and 1 nM E2 treatments (Fig. 7C). Combination treatment of 1 nM E2 + 10 μM AG1024 decreased DNA content significantly more than either treatment alone (Fig. 7C), suggesting an integral role of IGF-1Rβ in MCF-7:2A cells evading E2-induced apoptosis. To interrogate this further, growth pathway proteins were measured in response to 10 μM AG1024 treatment. MAPK and AKT pathways are both blocked by the IGF-1Rβ inhibitor after 72 h as shown by decreased p-MAPK and p-AKT levels when compared to vehicle-treated MCF-7:2A cells (Fig. 7D).

Discussion

This study investigated the mechanisms through which MCF-7:2A cells evade E2-induced apoptosis in vitro as a means to understand resistant breast cancer cells after long-term anti-hormone therapy in the clinic. After failure on an aromatase inhibitor, approximately 30% of breast cancer patients will respond to treatment with E2 (10); their nascent or remaining breast tumors will become cytostatic or disappear with physiological levels of E2. Further, E2 replacement therapy (ERT) has been shown to reduce the risk of breast cancer in hysterectomized post-menopausal women (12), perhaps due to E2-deprived breast cancer cells undergoing E2-induced apoptosis before resulting in clinically apparent disease. This study sought to discriminate between E2-deprived breast tumors that will quickly respond to treatment with E2 versus those that will respond more slowly and less dramatically. We modeled these different scenarios with MCF-7:5C and MCF-7:2A cell lines, respectively.

We have found that the UPR, associated with endoplasmic reticulum stress (ERS), is a fundamental element in E2-induced MCF-7:5C cell apoptosis (8). In this setting, E2 triggers UPR and rapidly causes apoptosis within one week of treatment. Two main sensors of the UPR, IRE1α and PERK are activated in both cell lines similarly. PERK activation is confirmed by elevated p-eIF2α, since eIF2α is phosphorylated by activated PERK. In MCF-7:2A cells, the same sensors are activated as in MCF-7:5C cells (Fig. 3), but significant cell death is not apparent at the same timepoint (Fig. 2A). Despite similar signaling patterns, the biological responses between the two cell lines differ. Our data suggested that another mechanism was preventing cell death after E2-induced UPR in MCF-7:2A cells.

Oxidative stress is a critical pathway for MCF-7:2A cells to undergo E2-induced apoptosis. MCF-7:2A cells inherently exhibit stronger survival and antioxidant mechanisms than MCF-7:5C cells (Figs. 46). This relationship is consistent with previously published data showing that MCF-7 cells with higher levels of glutathione peroxidase-1 (GSHPx-1) can survive better under oxidative stress conditions, such as hydrogen peroxide treatment (13), and that MCF-7 cells can increase antioxidant enzymes (i.e. manganese superoxide dismutase, MnSOD) to prevent TNF-mediated apoptosis (14). Activation of E2-induced apoptosis in MCF-7:2A cells also seems to require TNF family member upregulation (Fig. 4A and B). Oxidative stress occurs concurrently with upregulation of apoptosis-related genes in the TNF family. Whether increased TNFα causes oxidative stress or oxidative stress causes increased TNFα is not yet documented in this setting.

Additionally, B cell lymphoma 2 (BCL2) plays a role in preventing cell death caused by oxidative stress (15). In fact, MCF-7:2A cells exhibit 3.76-fold and 3.02-fold higher basal BCL2 and B cell lymphoma extra large (BCL-xL, BCL2L1) mRNA levels than MCF-7:5C cells, respectively (Table I), providing support for the idea of a stronger survival signal. Other data from our lab shows that MCF-7:2A cells exhibit 6.19-fold higher glutathione peroxidase 2 gene (GPX2) over MCF-7:5C cells (Table II), illustrating more evidence in favor of increased protection from E2-induced oxidative stress and apoptosis in this context.

Table I.

Basal apoptosis gene expression in MCF-7:2A cells versus MCF-7:5C.

Table I.

Basal apoptosis gene expression in MCF-7:2A cells versus MCF-7:5C.

Gene symbolFold change
AIFM25.7601
AKT12.5203
ANXA157.2949
ANXA42.7965
APAF12.839
ATF52.5303
BAG12.7188
BCL23.7598
BCL2L13.0192
BDNF5.8519
BIK6.2803
BIRC733.6437
CARD92.7968
CASP72.5278
CD272.7439
CD53.884
CD708.1739
CRYAB2.967
CUL33.2377
DAPK12.6145
DAPK26.023
EDAR5.7874
ERCC32.7634
ERN25.1671
GRM46.4268
HTT4.3186
HIP15.7736
HSPA1B2.5548
HSPB17.5902
IGF1R3.4421
IL1A31.2667
INHA2.5996
LGALS1430.9062
MAL3.0587
MALT13.2679
NLRC42.84
NOL32.9365
PLAGL13.3963
PLAGL23.0314
PPP1R13B2.7465
PPP2R1B4.5273
PRKCA2.503
PRODH3.8158
PTH5.7472
PYCARD3.1633
RARG2.968
SEMA4D2.9335
SFN3.2245
SIPA13.777
SOCS24.3464
STK17B3.8901
TBX53.3289
TNFRSF10D2.5864
TNFRSF184.0067
TNFRSF1976.9083
TNFRSF6B2.7982
TNFRSF83.103
TNFSF144.5599
TP6315.4118
TRAF22.5655
UNC13B3.0047
VHL3.1063
ZAK2.8369

[i] RT-PCR gene arrays of apoptosis-related genes were performed using MCF-7:5C and MCF-7:2A cells. Fold change represents gene expression of basal MCF-7:2A levels over basal MCF-7:5C levels. Only genes overexpressed in MCF-7:2A cells are shown. Particularly noteworthy in this study are BCL2 and BCL2L1.

Table II.

Top 10 overexpressed and underexpressed oxidative stress-related genes in MCF-7:2A versus MCF-7:5C.

Table II.

Top 10 overexpressed and underexpressed oxidative stress-related genes in MCF-7:2A versus MCF-7:5C.

Gene nameGene symbolCategoryFold change
Glutathione peroxidase 2 GPX2Glutathione peroxidases, oxidative stress responsive genes6.19
Keratin 1KRT1Oxidative stress responsive genes2.71
Heme oxygenase 1HMOX1Oxidative stress responsive genes2.66
Thioredoxin reductase 1TXNRD1Oxidative stress responsive genes, other antioxidants2.24
Peroxiredoxin 1PRDX1Peroxiredoxins (TPx)2.22
24-Dehydrocholesterol reductaseDHCR24Oxidative stress responsive genes2.21
Aldehyde oxidaseAOX1Other genes involved in ROS metabolism2.20
Forkhead box M1FOXM1Oxidative stress responsive genes1.83
ThioredoxinTXNOxidative stress responsive genes1.71
Prostaglandin-endoperoxide synthase 1PTGS1Other peroxidases1.71
Copper chaperone for superoxide dismutaseCCSOther genes involved in superoxide metabolism−1.65
Ring finger protein 7RNF7Oxidative stress responsive genes−1.65
Neutrophil cytosolic factor 2NCF2Other genes involved in superoxide metabolism−1.81
NADPH oxidase, EF-hand calcium binding domain 5NOX5Other genes involved in superoxide metabolism−1.83
Scavenger receptor class A, member 3SCARA3Oxidative stress responsive genes−1.98
Superoxide dismutase 3, extracellularSOD3Superoxide dismutases, other antioxidants−2.55
Cytochrome b-245, beta polypeptideCYBBOther peroxidases−3.19
Selenoprotein P, plasma, 1SEPP1Oxidative stress responsive genes−4.94
Apolipoprotein EAPOEOxidative stress responsive genes, other antioxidants−8.55
Chemokine (C-C motif) ligand 5CCL5Oxidative stress responsive genes−50.23

[i] Global gene expression analyses were performed, and oxidative stress-related genes were ranked by fold change of MCF-7:2A expression over MCF-7:5C expression. Notably, GPX2 shows the highest fold change.

Increased IGFR promotes anti-hormone resistance in breast cancer, likely through growth factor receptor crosstalk and aberrant ER, MAPK, and AKT signal transduction pathway activation (1618). Our data correlate with these findings in that higher IGF-1Rβ mRNA and protein expression confer a growth advantage and apoptotic resistance in MCF-7:2A cells despite treatment with E2 (Fig. 7). This suggests an IGF-1Rβ signaling pathway that can circumvent normal ER signaling in long-term estrogen-deprived breast cancer cells. Studies using hepato-cellular carcinoma cells (HCC) have demonstrated that IGF-1R overexpression can potentially cause increased glutathione transferase (GST) and protection from oxidative stress (19). Although this mechanism is shown in liver cancer cells, it may apply to our models of breast cancer as well. Perhaps the higher level of IGF-1Rβ in MCF-7:2A cells generates the increased glutathione levels necessary to escape cell death in the presence of E2.

The evidence thus far shows that TNF family member gene expression, protection against oxidative stress, and growth factor signaling are major mechanisms underlying the different biological responses to E2 seen in MCF-7:2A cells versus MCF-7:5C cells. Despite similar UPR signaling patterns, MCF-7:2A cells resist ERS-induced death longer and stronger than MCF-7:5C cells. Additional studies may provide further insight into the connection between IGF-1Rβ and glutathione in MCF-7:2A cells, and how this relationship functions in the presence and absence of a stressor such as E2. In order to effectively treat breast cancer patients who have undergone exhaustive anti-hormone treatment, and to explain why ERT can prevent breast cancer in some post-menopausal women, the examination of breast cancer cell models of E2 deprivation is proving invaluable. By understanding mechanisms that prevent apoptosis in these breast cancer cells, we can translate key findings into clinical practice.

Acknowledgements

This study was supported by the Department of Defense Breast Program (award number W81XWH-06-1-0590) Center of Excellence, the Susan G. Komen for the Cure Foundation (award number SAC100009), and the Lombardi Comprehensive Cancer Center Support Grant (core grant NIH P30 CA051008). The views and opinions of the authors do not reflect those of the US Army or the Department of Defense. We would like to acknowledge Heather Cunliffe, PhD at Translational Genomics (Phoenix, AZ) for her work on the network enrichment analysis in Fig. 1.

References

1. 

Sweeney EE, McDaniel RE, Maximov PY, Fan P and Jordan VC: Models and mechanisms of acquired resistance in breast cancer: Significant clinical progress despite limitations. Hom Mol Biol Clin Investig. 9:143–163. 2012.PubMed/NCBI

2. 

Jiang SY, Wolf DM, Yingling JM, Chang C and Jordan VC: An estrogen receptor positive MCF-7 clone that is resistant to anti-estrogens and estradiol. Mol Cell Endocrinol. 90:77–86. 1992. View Article : Google Scholar : PubMed/NCBI

3. 

Lewis JS, Osipo C, Meeke K and Jordan VC: Estrogen-induced apoptosis in a breast cancer model resistant to long-term estrogen withdrawal. J Steroid Biochem Mol Biol. 94:131–141. 2005. View Article : Google Scholar : PubMed/NCBI

4. 

Pink JJ, Jiang SY, Fritsch M and Jordan VC: An estrogen-independent MCF-7 breast cancer cell line which contains a novel 80-kilodalton estrogen receptor-related protein. Cancer Res. 55:2583–2590. 1995.

5. 

Pink JJ and Jordan VC: Models of estrogen receptor regulation by estrogens and antiestrogens in breast cancer cell lines. Cancer Res. 56:2321–2330. 1996.PubMed/NCBI

6. 

Pink JJ, Wu SQ, Wolf DM, Bilimoria MM and Jordan VC: A novel 80 kDa human estrogen receptor containing a duplication of exons 6 and 7. Nucleic Acids Res. 24:962–969. 1996. View Article : Google Scholar : PubMed/NCBI

7. 

Lewis JS, Meeke K, Osipo C, Ross EA, Kidawi N, Li T, Bell E, Changel NS and Jordan VC: Intrinsic mechanism of estradiol-induced apoptosis in breast cancer cells resistant to estrogen deprivation. J Natl Cancer Inst. 97:1746–1759. 2005. View Article : Google Scholar

8. 

Ariazi EA, Cunliffe HE, Lewis-Wambi JS, Slif ker MJ, Willis AL, Ramos P, Tapia C, Kim HR, Yerrum S, Sharma CG, Nicolas E, Balagurunathan Y, Ross EA and Jordan VC: Estrogen induces apoptosis in estrogen deprivation-resistant breast cancer through stress responses as identified by global gene expression across time. Proc Natl Acad Sci USA. 108:18879–18886. 2011. View Article : Google Scholar

9. 

Fan P, Griffith OL, Agboke FA, Anur P, Zou X, McDaniel RE, Creswell K, Kim SH, Katzenellenbogen JA, Gray JW and Jordan VC: c-Src modulates estrogen-induced stress and apoptosis in estrogen-deprived breast cancer cells. Cancer Res. 73:4510–4520. 2013. View Article : Google Scholar : PubMed/NCBI

10. 

Ellis MJ, Gao F, Dehdashti F, Jeffe DB, Marcom PK, Carey LA, Dickler MN, Silverman P, Fleming GF, Kommareddy A, Jamalabadi-Majidi S, Crowder R and Siegel BA: Lower-dose vs high-dose oral estradiol therapy of hormone receptor positive, aromatase inhibitor resistant advanced breast cancer: A Phase 2 randomized study. JAMA. 302:774–780. 2009. View Article : Google Scholar

11. 

Fan P, McDaniel RM, Kim HR, Clagett D, Haddad B and Jordan VC: Modulating therapeutic effects of the c-Src inhibitor via oestrogen receptor and human epidermal growth factor receptor 2 in breast cancer cell lines. Eur J Cancer. 48:3488–3498. 2012. View Article : Google Scholar

12. 

Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA, Black H, Bonds D, Brunner R, Brzyski R, Caan B, Chlebowski R, Curb D, Gass M, Hays J, Heiss G, Hendrix S, Howard BV, Hsia J, Hubbell A, Jackson R, Johnson KC, Judd H, Kotchen JM, Kuller L, LaCroix AZ, Lane D, Langer RD, Lasser N, Lewis CE, Manson J, Margolis K, Ockene J, O’Sullivan MJ, Phillips L, Prentice RL, Ritenbaugh C, Robbins J, Rossouw JE, Sarto G, Stefanick ML, Van Horn L, Wactawski-Wende J, Wallace R and Wassertheil-Smoller S; Women’s Health Initiative Steering Committee: Effects of conjugated equine estrogen in post-menopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. J Am Med Assoc. 291:1701–1712. 2004.

13. 

Doroshow JH: Glutathione peroxidase and oxidative stress. Toxicol Lett. 82–83:395–398. 1995.

14. 

Siemankowski LM, Morreale J and Briehl MM: Antioxidant defenses in the TNF-treated MCF-7 cells: selective increase in MnSOD. Free Radic Biol Med. 26:919–924. 1999. View Article : Google Scholar : PubMed/NCBI

15. 

Lee YJ, Chen JC, Amoscato AA, Bennouna J, Spitz DR, Suntharalingam M and Rhee JG: Protective role of Bcl2 in metabolic oxidative stress-induced cell death. J Cell Sci. 114:677–684. 2001.PubMed/NCBI

16. 

Brodie A, Macedo L and Sabnis G: Aromatase resistance mechanisms in model systems in vivo. J Steroid Biochem Mol Biol. 118:283–287. 2010. View Article : Google Scholar : PubMed/NCBI

17. 

Fox EM, Miller TW, Balko JM, Kuba MG, Sanchez V, Smith RA, Liu S, Gonzalez-Angulo AM, Mills GB, Ye F, Shyr Y, Manning HC, Buck E and Arteaga CL: A kinome-wide screen identifies the insulin/IGF-I receptor pathway as a mechanism of escape from hormone dependence in breast cancer. Cancer Res. 71:6773–6784. 2011. View Article : Google Scholar : PubMed/NCBI

18. 

Osborne CK and Schiff R: Mechanisms of endocrine resistance in breast cancer. Annu Rev Med. 62:233–247. 2011. View Article : Google Scholar : PubMed/NCBI

19. 

Lee JY, Han CY, Yang JW, Smith C, Kim SK, Lee EY, Kim SG and Kang KW: Induction of glutathione transferase in insulin-like growth factor type I receptor-overexpressed hepatoma cells. Mol Pharmacol. 72:1082–1093. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2014
Volume 44 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sweeney EE, Fan P and Jordan VC: Mechanisms underlying differential response to estrogen-induced apoptosis in long-term estrogen-deprived breast cancer cells. Int J Oncol 44: 1529-1538, 2014
APA
Sweeney, E.E., Fan, P., & Jordan, V.C. (2014). Mechanisms underlying differential response to estrogen-induced apoptosis in long-term estrogen-deprived breast cancer cells. International Journal of Oncology, 44, 1529-1538. https://doi.org/10.3892/ijo.2014.2329
MLA
Sweeney, E. E., Fan, P., Jordan, V. C."Mechanisms underlying differential response to estrogen-induced apoptosis in long-term estrogen-deprived breast cancer cells". International Journal of Oncology 44.5 (2014): 1529-1538.
Chicago
Sweeney, E. E., Fan, P., Jordan, V. C."Mechanisms underlying differential response to estrogen-induced apoptosis in long-term estrogen-deprived breast cancer cells". International Journal of Oncology 44, no. 5 (2014): 1529-1538. https://doi.org/10.3892/ijo.2014.2329