Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biomed Rep ; 20(3): 42, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38343657

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 (17ß-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.

2.
Bioimpacts ; 13(4): 313-321, 2023.
Article in English | MEDLINE | ID: mdl-37645026

ABSTRACT

Introduction: Resistance to chemotherapy and/or irradiation remains one of the key features of malignant tumors, which largely limits the efficiency of antitumor therapy. In this work, we studied the progression mechanism of breast cancer cell resistance to target drugs, including mTOR blockers, and in particular, we studied the exosome function in intercellular resistance transfer. Methods: The cell viability was assessed by the MTT assay, exosomes were purified by successive centrifugations, immunoblotting was used to evaluate protein expression, AP-1 activity was analyzed using reporter assay. Results: In experiments on the MCF-7 cell line (breast cancer) and the MCF-7/Rap subline that is resistant to rapamycin, the capability of resistant cell exosomes to trigger a similar rapamycin resistance in the parent MCF-7 cells was demonstrated. Exosome-induced resistance reproduces the changes revealed in MCF-7/Rap resistant cells, including the activation of ERK/AP-1 signaling, and it remains for a long time, for at least several months, after exosome withdrawal. We have shown that both the MCF-7 subline resistant to rapamycin and cells having exosome-triggered resistance demonstrate a stable decrease in the expression of DNMT3A, the key enzyme responsible for DNA methylation. Knockdown of DNMT3A in MCF-7 cells by siRNA leads to partial cell resistance to rapamycin; thus, the DNMT3A suppression is regarded as one of the necessary elements for the development of acquired rapamycin resistance. Conclusion: We propose that DNA demethylation followed by increased expression of key genes may be one of the factors responsible for the progression and maintenance of the resistant cell phenotype that includes exosome-induced resistance.

3.
Cancer Drug Resist ; 6(1): 103-115, 2023.
Article in English | MEDLINE | ID: mdl-37065867

ABSTRACT

Aim: The study aims to analyze the effect of long-term incubation of ERα-positive MCF7 breast cancer cells with 4-hydroxytamoxifen (HT) on their sensitivity to tubulin polymerization inhibitor docetaxel. Methods: The analysis of cell viability was performed by the MTT method. The expression of signaling proteins was analyzed by immunoblotting and flow cytometry. ERα activity was evaluated by gene reporter assay. To establish hormone-resistant subline MCF7, breast cancer cells were treated with 4-hydroxytamoxifen for 12 months. Results: The developed MCF7/HT subline has lost sensitivity to 4-hydroxytamoxifen, and the resistance index was 2. Increased Akt activity (2.2-fold) and decreased ERα expression (1.5-fold) were revealed in MCF7/HT cells. The activity of the estrogen receptor α was reduced (1.5-fold) in MCF7/HT. Evaluation of class III ß-tubulin expression (TUBB3), a marker associated with metastasis, revealed the following trends: higher expression of TUBB3 was detected in triple-negative breast cancer MDA-MB-231 cells compared to hormone-responsive MCF7 cells (P < 0.05). The lowest expression of TUBB3 was found in hormone-resistant MCF7/HT cells (MCF7/HT < MCF7 < MDA-MB-231, approximately 1:2:4). High TUBB3 expression strongly correlated with docetaxel resistance: IC50 value of docetaxel for MDA-MB-231 cells was greater than that for MCF7 cells, whereas resistant MCF7/HT cells were the most sensitive to the drug. The accumulation of cleaved PARP (a 1.6-fold increase) and Bcl-2 downregulation (1.8-fold) were more pronounced in docetaxel-treated resistant cells (P < 0.05). The expression of cyclin D1 decreased (2.8-fold) only in resistant cells after 4 nM docetaxel treatment, while this marker was unchanged in parental MCF7 breast cancer cells. Conclusion: Further development of taxane-based chemotherapy for hormone-resistant cancer looks highly promising, especially for cancers with low TUBB3 expression.

4.
Molecules ; 26(21)2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34771077

ABSTRACT

Hormone therapy is one of the most effective breast cancer treatments, however, its application is limited by the progression of hormonal resistance, both primary or acquired. The development of hormonal resistance is caused either by an irreversible block of hormonal signalling (suppression of the activity or synthesis of hormone receptors), or by activation of oestrogen-independent signalling pathways. Recently the effect of exosome-mediated intercellular transfer of hormonal resistance was revealed, however, the molecular mechanism of this effect is still unknown. Here, the role of exosomal miRNAs (microRNAs) in the transferring of hormonal resistance in breast cancer cells has been studied. The methods used in the work include extraction, purification and RNAseq of miRNAs, transfection of miRNA mimetics, immunoblotting, reporter analysis and the MTT test. Using MCF7 breast cancer cells and MCF7/T tamoxifen-resistant sub-line, we have found that some miRNAs, suppressors of oestrogen receptor signalling, are overexpressed in the exosomes of the resistant breast cancer cells. The multiple (but not single) transfection of one of the identified miRNA, miR-181a-2, into oestrogen-dependent MCF7 cells induced the irreversible tamoxifen resistance associated with the continuous block of the oestrogen receptor signalling and the activation of PI3K/Akt pathway. We suppose that the miRNAs-ERα suppressors may act as trigger agents inducing the block of oestrogen receptor signalling and breast cancer cell transition to an aggressive oestrogen-independent state.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Estrogen Receptor alpha/antagonists & inhibitors , Exosomes/drug effects , MicroRNAs/antagonists & inhibitors , Tamoxifen/pharmacology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Exosomes/genetics , Exosomes/metabolism , Female , Humans , MCF-7 Cells , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction/drug effects
5.
Molecules ; 26(9)2021 Apr 25.
Article in English | MEDLINE | ID: mdl-33922925

ABSTRACT

Exosomes are the small vesicles that are secreted by different types of normal and tumour cells and can incorporate and transfer their cargo to the recipient cells. The main goal of the present work was to study the tumour exosomes' ability to accumulate the parent mutant DNA or RNA transcripts with their following transfer to the surrounding cells. The experiments were performed on the MCF7 breast cancer cells that are characterized by the unique coding mutation in the PIK3CA gene. Using two independent methods, Sanger sequencing and allele-specific real-time PCR, we revealed the presence of the fragments of the mutant DNA and RNA transcripts in the exosomes secreted by the MCF7 cells. Furthermore, we demonstrated the MCF7 exosomes' ability to incorporate into the heterologous MDA-MB-231 breast cancer cells supporting the possible transferring of the exosomal cargo into the recipient cells. Sanger sequencing of the DNA from MDA-MB-231 cells (originally bearing a wild type of PIK3CA) treated with MCF7 exosomes showed no detectable amount of mutant DNA or RNA; however, using allele-specific real-time PCR, we revealed a minor signal from amplification of a mutant allele, showing a slight increase of mutant DNA in the exosome-treated MDA-MB-231 cells. The results demonstrate the exosome-mediated secretion of the fragments of mutant DNA and mRNA by the cancer cells and the exosomes' ability to transfer their cargo into the heterologous cells.


Subject(s)
Breast Neoplasms/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , DNA, Neoplasm/genetics , Exosomes/genetics , Alleles , Breast Neoplasms/pathology , Female , Humans , MCF-7 Cells , Mutation/genetics , RNA, Messenger/genetics
6.
Biochimie ; 186: 51-58, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33872751

ABSTRACT

Energy imbalance is one of the key properties of tumour cells, which in certain cases supports fast cancer progression and resistance to therapy. The simultaneous blocking of glycolytic processes and oxidative phosphorylation pathways seems to be a promising strategy for antitumor therapies. The study aimed to evaluate the effect of glucose starvation on the antiproliferative and antiestrogenic potency of oligomycin A against hormone-dependent breast cancer cells. Cell viability was assessed by the MTT test. Estrogen receptor alpha (ERα) activity was evaluated by reporter assay. mTOR, AMPK, Akt, and S6 kinase expression was assessed by immunoblotting. Glucose starvation caused multiple increases in the antiproliferative potency of oligomycin A in the hormone-dependent breast cancer MCF-7 cells, while its effect on the sensitivity of the second hormone-dependent cancer cell line, named T47D, was weak and limited. Glycolytic inhibitors, 3-bromopyruvate and 2-deoxyglucose, greatly enhanced the antiproliferative potency of oligomycin A in MCF-7 cells. Glucose starvation leads to remarkable activation of Akt in MCF-7 cells, whereas oligomycin A enhances its effect. The mTOR, S6 kinase, and AMPK signalling pathways are significantly modulated by oligomycin A under glucose starvation. Oligomycin A demonstrates more pronounced antiestrogenic effects under glucose starvation. Thus, glucose starvation and pharmacological inhibition of glycolysis are of interest for revealing the antitumor potential of macrolide oligomycin A against hormone-dependent breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Estrogen Receptor Modulators/pharmacology , Glucose/deficiency , Oligomycins/pharmacology , Breast Neoplasms/pathology , Female , Humans , MCF-7 Cells
7.
Pharmaceuticals (Basel) ; 13(9)2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32825760

ABSTRACT

The phenomenon of the primary or acquired resistance of cancer cells to antitumor drugs is among the key problems of oncology. For breast cancer, the phenomenon of the resistance to hormonal or target therapy may be based on the numerous mechanisms including the loss or mutation of estrogen receptor, alterations of antiapoptotic pathways, overexpression of growth-related signaling proteins, etc. The perspective approaches for overcoming the resistance may be based on the usage of compounds such as inhibitors of the cell energetic metabolism. Among the latter, the antidiabetic drug metformin exerts antitumor activity via the activation of AMPK and the subsequent inhibition of mTOR signaling. The experiments were performed on the ERα-positive MCF-7 breast cancer cells, the MCF-7 sublines resistant to tamoxifen (MCF-7/T) and rapamycin (MCF-7/Rap), and on triple-negative MDA-MB-231 breast cancer cells. We have demonstrated metformin's ability to enhance the cytostatic activity of the tamoxifen and rapamycin on both parent MCF-7 cells and MCF-7-resistant derivates mediated via the suppression of mTOR signaling and growth-related transcriptional factors. The cooperative effect of metformin and tested drugs was realized in an estrogen-independent manner, and, in the case of tamoxifen, was associated with the activation of apoptotic cell death. Similarly, the stimulation of apoptosis under metformin/tamoxifen co-treatment was shown to occur in the MCF-7 cells after steroid depletion as well as in the ERα-negative MDA-MB-231 cells. We conclude that metformin co-treatment may be used for the increase and partial restoration of the cancer cell sensitivity to hormonal and target drugs. Moreover, the combination of metformin with tamoxifen induces the apoptotic death in the ERα-negative breast cancer cells opening the additional perspectives in the treatment of estrogen-independent breast tumors.

8.
Chem Biol Interact ; 330: 109243, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32861747

ABSTRACT

mTOR inhibitors are considered today to be one of the most promising anticancer drugs. Here to study the mechanism of the acquired resistance of MCF-7 breast cancer cells to mTOR inhibitors two different models of the cell resistance were used: rapamycin-resistant MCF-7/Rap subline developed under long-term rapamycin treatment, and metformin-resistant MCF-7/M subline obtained by long-term metformin treatment. We have found that both resistant sublines were characterized by common features: increased expression of mTOR-interacting Raptor protein, increased phosphorylation of Akt, and activation of growth-related transcriptional factor AP-1. Cell response to mTOR inhibitors was partially restored under treatment with PI3K inhibitor wortmannin supporting the direct connection between Akt activation and poor cell response to therapeutic drugs. Transfection of mir-181c, one of the positive regulators of Akt and mTOR, led to an increase in the cell resistance to both mTOR inhibitors, rapamycin and metformin, which correlated with Raptor overexpression and activation of Akt/AP-1 signaling. In general, the effect of Raptor overexpression in the resistant cells, as well as the ability of mir-181c to modulate the Raptor expression, can open novel perspectives in the treatment of rapalogues-resistant cancers, based on the drugs design targeting mir-181c/Raptor axis.


Subject(s)
Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Proto-Oncogene Proteins c-akt/metabolism , Regulatory-Associated Protein of mTOR/metabolism , Sirolimus/pharmacology , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Humans , MCF-7 Cells , MicroRNAs/genetics , MicroRNAs/pharmacology , Signal Transduction , Up-Regulation/drug effects
9.
Molecules ; 23(4)2018 04 04.
Article in English | MEDLINE | ID: mdl-29617321

ABSTRACT

Exosomes are small vesicles which are produced by the cells and released into the surrounding space. They can transfer biomolecules into recipient cells. The main goal of the work was to study the exosome involvement in the cell transfer of hormonal resistance. The experiments were performed on in vitro cultured estrogen-dependent MCF-7 breast cancer cells and MCF-7 sublines resistant to SERM tamoxifen and/or biguanide metformin, which exerts its anti-proliferative effect, at least in a part, via the suppression of estrogen machinery. The exosomes were purified by differential ultracentrifugation, cell response to tamoxifen was determined by MTT test, and the level and activity of signaling proteins were determined by Western blot and reporter analysis. We found that the treatment of the parent MCF-7 cells with exosomes from the resistant cells within 14 days lead to the partial resistance of the MCF-7 cells to antiestrogen drugs. The primary resistant cells and the cells with the exosome-induced resistance were characterized with these common features: decrease in ERα activity and parallel activation of Akt and AP-1, NF-κB, and SNAIL1 transcriptional factors. In general, we evaluate the established results as the evidence of the possible exosome involvement in the transferring of the hormone/metformin resistance in breast cancer cells.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor Modulators/pharmacology , Exosomes/metabolism , Breast Neoplasms/metabolism , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Snail Family Transcription Factors/metabolism
10.
IUBMB Life ; 68(4): 281-92, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26892736

ABSTRACT

Metformin, a biguanide antidiabetic drug, is used to decrease hyperglycemia in patients with type 2 diabetes. Recently, the epidemiological studies revealed the potential of metformin as an anti-tumor drug for several types of cancer, including breast cancer. Anti-tumor metformin action was found to be mediated, at least in part, via activation of adenosine monophosphate-activated protein kinase (AMPK)-intracellular energy sensor, which inhibits the mammalian target of rapamycin (mTOR) and some other signaling pathways. Nevertheless, some patients can be non-sensitive or resistant to metformin action. Here we analyzed the mechanism of the formation of metformin-resistant phenotype in breast cancer cells and its role in estrogen receptor (ER) regulation. The experiments were performed on the ER-positive MCF-7 breast cancer cells and metformin-resistant MCF-7 subline (MCF-7/M) developed due to long-term metformin treatment. The transcriptional activity of NF-κB and ER was measured by the luciferase reporter gene analysis. The protein expression was determined by immunoblotting (Snail1, (phospho)AMPK, (phospho)IκBα, (phospho)mTOR, cyclin D1, (phospho)Akt and ERα) and immunohistochemical analysis (E-cadherin). We have found that: 1) metformin treatment of MCF-7 cells is accompanied with the stimulation of AMPK and inhibition of growth-related proteins including IκBα, NF-κB, cyclin D1 and ERα; 2) long-term metformin treatment lead to the appearance and progression of cross-resistance to metformin and tamoxifen; the resistant cells are characterized with the unaffected AMPK activity, but the irreversible ER suppression and constitutive activation of Akt/Snail1 signaling; 3) Akt/Snail1 signaling is involved into progression of metformin resistance. The results presented may be considered as the first evidence of the progression of cross-resistance to metformin and tamoxifen in breast cancer cells. Importantly, the acquired resistance to both drugs is based on the constitutive activation of Akt/Snail1/E-cadherin signaling that opens new perspectives to overcome the metformin/tamoxifen resistance of breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Tamoxifen/pharmacology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
11.
Exp Cell Res ; 319(20): 3150-9, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-23973669

ABSTRACT

The tolerance of cancer cells to hypoxia depends on the combination of different factors--from increase of glycolysis (Warburg Effect) to activation of intracellular growth/apoptotic pathways. Less is known about the influence of epithelial-mesenchymal transition (EMT) and EMT-associated pathways on the cell sensitivity to hypoxia. The aim of this study was to explore the role of Snail signaling, one of the key EMT pathways, in the mediating of hypoxia response and regulation of cell sensitivity to hypoxia, using as a model in vitro cultured breast cancer cells. Earlier we have shown that estrogen-independent HBL-100 breast cancer cells differ from estrogen-dependent MCF-7 cells with increased expression of Snail1, and demonstrated Snail1 involvement into formation of hormone-resistant phenotype. Because Snail1 belongs to hypoxia-activated proteins, here we studied the influence of Snail1 signaling on the cell tolerance to hypoxia. We found that Snail1-enriched HBL-100 cells were less sensitive to hypoxia-induced growth suppression if compared with MCF-7 line (31% MCF-7 vs. 71% HBL-100 cell viability after 1% O2 atmosphere for 3 days). Snail1 knock-down enhanced the hypoxia-induced inhibition of cell proliferation giving the direct evidence of Snail1 involvement into cell protection from hypoxia attack. The protective effect of Snail1 was shown to be mediated, at least in a part, via beta-catenin which positively regulated expression of HIF-1-dependent genes. Finally, we found that cell tolerance to hypoxia was accompanied with the failure in the phosphorylation of AMPK - the key energy sensor, and demonstrated an inverse relationship between AMPK and Snail/beta-catenin signaling. Totally, our data show that Snail1 and beta-catenin, besides association with loss of hormone dependence, protect cancer cells from hypoxia and may serve as an important target in the treatment of breast cancer. Moreover, we suggest that the level of these proteins as well the level of AMPK phosphorylation may be considered as predictors of the tumor sensitivity to anti-angiogenic drugs.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Hypoxia/metabolism , Signal Transduction , Transcription Factors/metabolism , beta Catenin/metabolism , Cells, Cultured , Female , Humans , MCF-7 Cells , Snail Family Transcription Factors
12.
J Cell Biochem ; 113(6): 2147-55, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22307688

ABSTRACT

The loss of hormonal dependency of breast tumor cells is often accompanied with the appearance of epithelial-mesenchymal transition (EMT) features and increase in cell metastasis and invasiveness. The central role in the EMT belongs to transcription factors Snail responded for the decrease in E-cadherin expression and cell contacts, stimulation of cell mobility and invasiveness. Aim was to study the relationships between estrogen receptor machinery and Snail1 signaling, and mechanism of Snail1 regulation in hormone-resistant breast cancer cells. The experiments were performed on the estrogen-dependent MCF-7 breast cancer cells, estrogen-hyposensitive MCF-7/LS subline generated through long-term cultivation of the parental cells in steroid-free medium, and ER-negative estrogen-resistant HBL-100 cells. Snail1, estrogen receptor, p65 NF-κB, E-cadherin levels were analyzed by Western blot. We found that decrease in the estrogen dependency is correlated with increase in Snail1 expression and activity, we demonstrated the Snail1 involvement in the negative regulation of ER, and showed that Snail1 inhibition partially restores the sensitivity of the estrogen-hyposensitive cells to antiestrogen tamoxifen. Furthermore, NF-κB was found to serve as a positive regulator of Snail1 in breast cancer cells, and simultaneous inhibition of NF-κB and Snail1 resulted in additional increase in cell response to tamoxifen. In general, the results obtained demonstrate the phenomenon of Snail1 activation in the hormone-resistant breast cancer cells, and show that Snail1 and NF-κB may serve as an important targets in the treatment of breast cancer, both estrogen-dependent and estrogen-independent tumors.


Subject(s)
Breast Neoplasms/metabolism , NF-kappa B/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis , Cadherins/biosynthesis , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , NF-kappa B/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction , Snail Family Transcription Factors , Tamoxifen/pharmacology , Transcription Factor RelA/biosynthesis , Transcription Factors/genetics
13.
Biosci Rep ; 31(2): 137-43, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20662767

ABSTRACT

Recently, it was shown that the resistance of breast cancer cells to growth-stimulating oestrogen action may be accompanied with the paradoxical tumour sensitization to oestrogen apoptotic action. In the present paper, we studied the influence of oestrogens on the sensitivity of resistant breast tumours to cytostatic drugs, and to evaluate the role of NF-κB (nuclear factor κB) signalling in the regulation of the apoptotic response of the resistant cells. The experiments were carried out on the oestrogen-dependent MCF-7 breast cancer cells and resistant MCF-7/LS subline generated through long-term cultivation of the parental cells in the absence of oestrogen. The cell treatment with the combination of oestradiol and Dox (doxorubicin) was found to enhance the apoptotic action of Dox in MCF-7/LS cells but not in the parent cells. MCF-7/LS cells were characterized by the increased level of ROS (reactive oxygen species) and decreased NF-κB activity. Oestradiol in combination with Dox leads to significant NF-κB stimulation and its accumulation in the nucleus of MCF-7/LS cells. The knockdown of NF-κB with siRNA (small interfering RNA) increased the apoptotic response of the MCF-7/LS cells to both Dox and oestradiol demonstrating the important role of NF-κB in the protection of the MCF-7/LS cells against apoptosis. In general, the results obtained show that: (i) oestradiol enhances the apoptotic action of Dox in the resistant breast cancer cells; and (ii) suppression of NF-κB signalling amplifies the apoptotic response of the resistant cells to both oestrogen and Dox, demonstrating that NF-κB may serve as a potential target in the therapy of the resistant breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Estradiol/pharmacology , Estrogens/pharmacology , Neoplasms, Hormone-Dependent/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Doxorubicin/therapeutic use , Estrogens/therapeutic use , Female , Humans , NF-kappa B/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species , Signal Transduction
14.
Steroids ; 74(6): 535-42, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19428442

ABSTRACT

The progression of cancer is associated with tumor's ability to outgrow the existing vasculature resulting in chronic hypoxic pressure, however the molecular mechanism of cancer cell response to chronic hypoxia is poorly understood. In this study we have analyzed the reorganization of estrogen receptor (ER) signaling in breast cancer cells under chronic hypoxia and examined the role of interrelations between ER and NF-kB signaling in cell adaptation to hypoxia. Using long-term culturing of MCF-7 breast cancer cells in hypoxia-mimetic conditions (cobalt chloride) we have established a hypoxia-tolerant subline characterized by HIF-1 hyperexpression that retained the tolerance to hypoxia even when the cells were returned to normoxic conditions. The hypoxia-tolerant cells were characterized by non-affected ER signaling, irreversible suppression of NF-kB activity, and increased sensitivity to cytokine-induced apoptosis. Estradiol treatment suppressed the NF-kB activity in both parent and hypoxia-tolerant MCF-7 cells. In contrast to MCF-7 cells, the exposure of estrogen-independent MCF-7/T2 subline to chronic hypoxia was not accompanied by noticeable changes in NF-kB activity or cell sensitivity to cytokines. Taken together, the results presented demonstrate the importance of interrelations between ER and NF-kB signaling in the response of estrogen-dependent breast cancer cells to chronic hypoxia.


Subject(s)
NF-kappa B/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/physiology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogens/pharmacology , Female , Humans , Luciferases/genetics , Luciferases/metabolism , NF-kappa B/genetics , Receptors, Estrogen/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , Signal Transduction/drug effects , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Protein p53/metabolism
15.
Anticancer Res ; 27(4A): 1777-82, 2007.
Article in English | MEDLINE | ID: mdl-17649772

ABSTRACT

BACKGROUND: The phosphatidylinositol 3-kinase (PI3K)/Akt signalling pathway plays a major role in the regulation of breast cancer growth and survival, but the clinical value of its components in human tumours is unclear. PATIENTS AND METHODS: PI3K was analysed using Western blotting with monoclonal antibodies to the p85 subunit in tumour and adjacent mammary gland samples from 33 breast cancer patients. Activated Akt1 (pAkt1) expression was quantified in 46 sample pairs by a direct sandwich ELISA assay. RESULTS: Tumour PI3K expression was increased in 79% of the investigated sample pairs and was not associated with the main clinico-pathological features. Only 49% of breast cancers had increased pAkt1, but the frequency of its elevation was positively associated with tumour size and histological grade, and controversially related to estrogen and progesterone receptor status. CONCLUSION: Increased PI3K but not pAkt1, expression appears to be a widespread feature of human breast cancer indicating the different roles of the two components of one signalling system.


Subject(s)
Breast Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Adult , Blotting, Western , Breast Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Middle Aged
16.
Breast Cancer Res Treat ; 100(1): 1-11, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16990991

ABSTRACT

Paradoxical induction of apoptosis by estrogen has been described previously for estrogen-deprived and antiestrogen-resistant breast cancer cells. In this study we analyzed the possible interrelations between cell sensitization to estrogen apoptotic action and cell ability to (anti)estrogen-independent growth. Using tamoxifen-resistant sublines derived from the parent MCF-7 breast cancer cells by long-term tamoxifen treatment we demonstrated that resistant cells are characterized by increased level of EGF receptor and unexpected increase of VEGF receptor 2 (Flk-1/KDR) and its specific ligand, VEGF-A. The importance of the VEGF signaling in the autocrine regulation of cell growth was indicated by the ability of VEGF inhibitor, soluble fragment of Flt-1/Fc chimera, to suppress the phosphorylation of MAP kinases as well as to inhibit the estrogen-independent growth of MCF-7 cells. Sensitization of tamoxifen-resistant cells to estrogen-induced apoptosis required the additional continuous cultivation in steroid-depleted medium and did not depend on the activity of both EGF and VEGF pathways. Finally, we showed that treatment of the cells with 17beta-estradiol (10(-9) M) resulted in a marked increase in p53 level both in the resistant cells undergoing apoptosis and in the parent MCF-7 cells insensitive to apoptotic estrogen action. These data provide an important support for the existence of a disbalance between pro- and anti-apoptotic machinery in the resistant breast cancer cells that forms independently of the acquired ability to estrogen-independent growth.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Selective Estrogen Receptor Modulators , Tamoxifen , Apoptosis , Breast Neoplasms/pathology , Cell Line, Tumor/metabolism , ErbB Receptors/metabolism , Female , Flow Cytometry , Humans , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
17.
J Steroid Biochem Mol Biol ; 82(4-5): 369-76, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12589944

ABSTRACT

STAT proteins act as signal transducers and activators of transcription in cells treated with cytokines or growth factors. Here we analyzed the possible cooperation between STAT3 and phosphatidylinositol-3 kinase (PI-3 kinase) and its involvement in antiproliferative signals induced by glucocorticoid hormones. Treatment of melanoma cells with dexamethasone (DEX) resulted in coexpression of STAT3 activation and increase in the PI-3 kinase protein level. Using plasmids-containing JAK2 and STAT3 constructs, we demonstrated that activation of JAK/STAT signaling led to up regulation of PI-3 kinase and enhancement of DEX's ability to increase PI-3 kinase levels in target cells. Prolonged DEX treatment of melanoma cells resulted in constitutive increases in both STAT3 and PI-3 kinase protein levels that were correlated with increased melanoma resistance to antiproliferative hormone action. Similarly, forced expression of both STAT3 and PI-3 kinase in melanoma cells led to enhanced resistance to hormone treatment. Forced expression of PI-3 kinase led to increase in STAT3 activity in a JAK-dependent manner, indicating the existence of a feedback regulatory cascade between the JAK/STAT3 and PI-3 kinase pathways. We suggest that protection of melanoma cells from antiproliferative effects of glucocorticoid hormones may be mediated, at least in part, by the constitutive activation of the STAT3/PI-3 kinase signaling pathway.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Melanoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins , Signal Transduction/drug effects , Trans-Activators/metabolism , Blotting, Western , Cell Division/drug effects , Drug Resistance, Neoplasm , Enzyme Activation , Humans , Janus Kinase 2 , Luciferases/metabolism , Melanoma/pathology , Phosphorylation , Plasmids , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...