Polyamines modulate the roscovitine-induced cell death switch decision autophagy vs. apoptosis in MCF-7 and MDA-MB-231 breast cancer cells

  • Authors:
    • Elif Damla Arisan
    • Yunus Akkoç
    • Kaan Gencer Akyüz
    • Ezgi Melek Kerman
    • Pinar Obakan
    • Ajda Çoker‑Gürkan
    • Narçin Palavan Ünsal
  • View Affiliations

  • Published online on: February 4, 2015     https://doi.org/10.3892/mmr.2015.3303
  • Pages: 4532-4540
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Current clinical strategies against breast cancer mainly involve the use of anti‑hormonal agents to decrease estrogen production; however, development of resistance is a major problem. The resistance phenotype depends on the modulation of cell‑cycle regulatory proteins, cyclins and cyclin‑dependent kinases. Roscovitine, a selective inhibitor of cyclin‑dependent kinases, shows high therapeutic potential by causing cell‑cycle arrest in various cancer types. Autophagy is a type of cell death characterized by the enzymatic degradation of macromolecules and organelles in double‑ or multi‑membrane autophagic vesicles. This process has important physiological functions, including the degradation of misfolded proteins and organelle turnover. Recently, the switch between autophagy and apoptosis has been proposed to constitute an important regulator of cell death in response to chemotherapeutic drugs. The process is regulated by several proteins, such as the proteins of the Atg family, essential for the initial formation of the autophagosome, and PI3K, important at the early stages of autophagic vesicle formation. Polyamines (PAs) are small aliphatic amines that play major roles in a number of eukaryotic processes, including cell proliferation. The PA levels are regulated by ornithine decarboxylase (ODC), the rate‑limiting enzyme in PA biosynthesis. In this study, we aimed to investigate the role of PAs in roscovitine‑induced autophagic/apoptotic cell death in estrogen receptor‑positive MCF‑7 and estrogen receptor‑negative MDA‑MB‑231 breast cancer cells. We show that MDA‑MB‑231 cells are more resistant to roscovitine than MCF‑7 cells. This difference was related to the regulation of autophagic key molecules in MDA‑MB‑231 cells. In addition, we found that exogenous PAs have a role in the cell death decision between roscovitine‑induced apoptosis or autophagy in MCF‑7 and MDA‑MB‑231 breast cancer cells.

Introduction

Breast cancer is one of the most commonly diagnosed cancer types in women, and the second leading cause of cancer-related deaths worldwide (1). Patients with estrogen receptor α (ERα)-positive tumors greatly benefit from existing hormonal therapies. Although anti-estrogens are being used to treat breast cancer (2,3), numerous cases show acquired resistance and irresponsiveness to endocrine therapy, which is a major clinical problem (3,4). Despite the emergence of new promising advances in therapeutics, options to treat hormone-resistant breast tumors are limited, and the mortality rate continues to increase.

It has been suggested, based on a number of findings, that deregulation of cell-cycle components such as cyclin-dependent kinases (CDKs) can contribute to endocrine resistance (5). Therefore, inhibition of CDKs by synthetic, small-molecule drugs has become an attractive therapeutic strategy. Roscovitine is a small, purine-like CDK inhibitor with increased selectivity towards CDK1, CDK2, CDK7 and CDK9 (68). Previous studies have shown that roscovitine promotes the accumulation of breast cancer cells at the G2/M phase (9,10) and potentiates the antitumor effects of other chemotherapeutic agents, by inducing apoptotic cell death (11). Besides CDKs, the progression of the cell cycle is related to polyamines (PAs), which are amine-derived cationic molecules. Several studies provided evidence for a PA-dependent G0–G1 transition and G1 phase progression in different cell lines (12,13).

Among PAs, natural putrescine (Put), spermidine (Spd) and spermine (Spm) are required for cell growth and proliferation (14). Intracellular PA levels are tightly regulated in eukaryotes by the activity of the ornithine decarboxylase (ODC), which catalyzes the conversion of ornithine to Put (15). Activation of PA biosynthesis leads to the accumulation of intracellular PAs, which is a critical event in various diseases, including breast cancer (16,17). Previous studies have shown that PAs are involved in neoplastic transformation by activating several proto-oncogenes, such as c-Myc (18,19).

Autophagy, the process responsible for the degradation of cytoplasmic proteins, macromolecules and damaged or aged organelles, is considered a type of cell death. The most significant sign of autophagy is the appearance of double-membrane enclosed vesicles in the cytoplasm, which engulf portions of the cytoplasm and/or organelles (2022).

A number of studies have shown that PAs are associated with autophagy via histone acetylation and chromatin remodeling mechanisms. Specifically, Spd was suggested to be a critical ‘tuning’ molecule in autophagy, through epigenetic alterations (2325). Spd was shown to inhibit the enzymatic activity of histone acetyl transferase (HAT) and lead to hypoacetylation of histone H3 (25). For this reason, it is considered that autophagic processes can be activated by the acetylation, by PAs, of autophagic promoter molecules. However, the molecular mechanism involved in drug-induced apoptosis or autophagy related to the regulation of PA biosynthesis has not yet been fully clarified.

In the present study, we aimed to reveal the potential role of PAs in roscovitine-induced apoptosis and/or autophagy in MCF-7 and MDA-MB-231 breast cancer cells.

Materials and methods

Drugs and antibodies

Roscovitine was purchased from Sigma-Aldrich (St. Louis, MO, USA), was dissolved in dimethyl sulfoxide (DMSO) to make a 10 mM stock solution, and was stored at −20°C. Spd, Spm (each at 10 mM) and 3-aminoguanidine were purchased from Sigma-Aldrich. 3-Aminoguanidine was used as an amine oxidase blocker in the Spd and Spm treatment experiments.

Antibodies targeting beclin-1 (dilution, 1:1,000), Atg5 (1:1,000), Atg12 (1:1,000), LC3A/B (1:1,000), β-actin (1:1,000), β-tubulin (1:1,000), pro-caspase-9 (1:1,000), cleaved-caspase-9 (1:1,000), caspase-7 (1:1,000) and horseradish peroxidase (HRP)-conjugated secondary IgG (1:3,000) were purchased from Cell Signaling Technology (Danvers, MA, USA).

Cell cultures

The breast cancer cell lines MCF-7 (HTB 22) and MDA-MB-231 (HTB 26) were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). The cells were maintained in Gibco® Dulbecco’s modified Eagle’s medium (DMEM; Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% fetal bovine serum (Pan-Biotech GmbH, Aidenbach, Germany) and 100 units or 100 mg/ml penicillin or streptomycin, and were grown in humidified air with 5% CO2 at 37°C, in a Heracell® 150i incubator (Thermo Fisher Scientific).

Cell viability assay

The effect of roscovitine on cell viability in the presence or absence of PAs was determined by the colorimetric assay 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Roche Diagnostics, Indianapolis, IN, USA). Cells were plated in 96-well plates at a density of 1×105 cells/well, were allowed to attach overnight, and were treated for 24 h with various concentrations of roscovitine in the presence or absence of PAs. After 24 h of treatment, 10 μl of the MTT reagent (5 mg/ml) were added to the cell culture medium, and cells were incubated for 4 h. Following medium removal, 200 μl of DMSO were added to dissolve the formazan crystals, which are produced by the activated mitochondria. The absorbance of the suspensions was measured at 595 nm on a microplate reader (Bio-Rad, Hercules, CA, USA).

Fluorescence staining

Cells (5×104) were seeded into 12-well plates, allowed to attach overnight and then treated with appropriate concentrations of drugs for 24 h. In order to assess the mitochondrial membrane potential (MMP), cells were washed once with 1X phosphate-buffered saline (PBS) and stained with 0,4 mM 3,3′-dihexyloxacarbocyanine iodide (DiOC6). The absorbance of samples (Abs 488/525) was measured on a Fluoroskan Ascent Microplate fluorometer (Thermo Fisher Scientific, Beverly, MA, USA), with excitation and emission settings of 488 and 525 nm, respectively; the Abs 488/525 of the samples was compared to that of the control.

For monodansylcadaverine (MDC) staining, cells (6×104) were seeded into 6-well plates on coverslips, allowed to attach overnight and then treated with the appropriate drug concentrations for 24 h. Cells were washed once with 1X PBS and stained with 50 μM MDC in order to visualize the autophagic vesicles. Next, they were observed under a fluorescence microscope (Olympus, Tokyo, Japan).

Cell death enzyme-linked immunosorbent assay (ELISA) assay

The cytoplasmic histone-associated DNA-fragments (mono- and oligonucleosomes) were measured with the Cell Death Detection ELISA PLUS kit (Roche Diagnostics) according to the manufacturer’s instructions. Cells (1×104) were seeded into 96-well plates and treated with the desired drug concentrations for 24 h. The cell lysates were placed in a streptavidin-coated microplate. A mixture of anti-histone biotin and anti-DNA peroxidase (POD) was added, and samples were kept at room temperature for 2 h. Following washing of the unbound antibodies, the colorimetric assay was performed with 2,2′-azino-bis(3-ethylbenzthiazoline-6-sulphonic acid as a substrate, and the absorbance was measured at 405 nm.

Immunoblot analysis

Cells were treated with the appropriate concentrations of each drug for 24 h. The MCF-7 and MDA-MB-231 cells were lysed with ProteoJET Mammalian cell lysis reagent (Fermentas, Thermo Fisher Scientific, Waltham, MA, USA) containing total protease inhibitor cocktail (Roche Diagnostics GmbH, Mannheim, Germany).

Following lysis, cell debris was removed by centrifugation for 15 min at 18,500 × g, and protein concentration was determined with the Bradford method (Quick Start™ Bradford Protein Assay kit; Bio-Rad Laboratories, Hercules, CA, USA). Total protein lysates (30 μg) were separated by 15% SDS-polyacrylamide gel electrophoresis (PAGE) and transferred onto polyvinylidene difluoride membranes (Roche Diagnostics). The membranes were then blocked with 5% non-fat milk, prepared in a 1% Tris-buffered saline and Tween-20 (TBST) solution. Following incubation of the membranes with the appropriate primary antibody at 4°C overnight, the membranes were washed with TBST. The membranes were then incubated with the appropriate HRP-conjugated secondary antibody overnight at 4°C, an enhanced chemiluminescence (ECL) reagent (Lumi-Light Western Blotting substrate; Roche Diagnostics GmbH) was used to visualize the antigens. Finally, the membranes were exposed to Kodak X-ray film (Kodak, Rochester, NY, USA) in a dark room.

Statistical analysis

Differences between samples were statistically evaluated using an Office Excel calculation file (Microsoft, New York, NY, USA). The results from the MTT, cell death ELISA and MMP assay were expressed as mean ± standard deviation. Student’s t-tests were applied toassess the significance of comparisons. Differences were regarded as statistically significant at P<0.05.

Results

Roscovitine-induced cytotoxicity is altered by polyamine treatment

In order to understand the effect of roscovitine on MCF-7 and MDA-MB-231 breast cancer cells, the MTT cell viability assay was performed following treatment with various concentrations of the drug (0–100 μM) for 24 h. The cell viability was decreased by 35 and 25% following treatment with 20 μM roscovitine in MCF-7 and MDA-MB-231 breast cancer cell lines, respectively (Fig. 1A). This concentration was selected for the following experiments.

To evaluate the combined effect of Spd or Spm (each 10 μM) with roscovitine, each cell line was exposed to drugs for 24 h. Although Spd treatment caused moderate cytotoxicity (23% reduction in cell viability in MCF-7 and 21% in MDA-MB-231 cells vs. control, respectively), Spm treatment was less effective (7% in MCF-7 and 4% in MDA-MB-231 cells) (Fig. 1B and C). Co-treatment with Spd or Spm and roscovitine enhanced the roscovitine-induced cytotoxicity in both breast cancer cell lines. In addition, the promoting effects of Spd on cytotoxicity were significant in both cell lines, particularly in the MDA-MB-231 cells (P<0.0002).

Roscovitine-induced mitochondria-mediated apoptosis via caspase activation

We determined the apoptotic potential of roscovitine in the presence or absence of Spd/Spm in the cells. Although neither Spd nor Spm exerted significant apoptotic effects, roscovitine induced apoptosis by 4- and 2.5-fold in MCF-7 and MDA-MB-231 breast cancer cells, respectively, as compared to untreated control cells. Spm alone slightly induced apoptosis in MDA-MB-231 cells by 1.5-fold compared to control cells. Co-treatment with Spd or Spm and roscovitine enhanced the cell viability reduction in both cell lines; it also prevented drug-induced apoptosis by decreasing the DNA fragmentation ratio (Fig. 2).

We performed DiOC6 staining to visualize the MMP loss on a fluorometer and thus, investigate the role of PAs in roscovitine-induced apoptosis. Although roscovitine decreased MMP in both cell lines, co-treatment with Spd did not affect the roscovitine-induced MMP reduction (Fig. 3). By contrast, Spm protected the MCF-7, but not the MDA-MB-231 cells, from roscovitine-induced mitochondria-mediated apoptosis, although theses changes were not significant.

To further investigate drug-induced caspase activation, we determined the level of cleaved fragments of caspase-9 and -7 by immunoblotting. While caspase-9 cleavage, which is the initial step for caspase activation in mitochondria-mediated apoptosis, appeared increased, the level of the full-length caspase-7, the executioner caspase for apoptosis, was decreased after roscovitine treatment for 24 h in breast cancer cells. Although exposure of MCF-7 cells to Spd or Spm for 24 h did not appear to activate caspase-9, treatment with each of these PAs led to a decrease in the caspase-7 level. In addition, combined treatment with Spd or Spm and roscovitine further decreased the expression level of the full-length caspase-7 in MCF-7 cells. Roscovitine induced the cleavage of caspase-9 and -7 in MDA-MB-231 breast cancer cell lines. In addition, PAs enhanced the roscovitine-induced caspase-9 and -7 activation by decreasing the level of the full-length fragments of these caspases in MDA-MB-231 cells (Fig. 4).

Roscovitine induces autophagic modulation

In order to evaluate the role of roscovitine on autophagic cell death in the MCF-7 and MDA-MB-231 breast cancer cell lines, we performed immunoblotting assays at different time-points. We examined the expression profile of beclin-1, which is referred to as the initial key molecule for autophagy, following roscovitine treatment within 24 h. Interestingly, while beclin-1 appeared to be time-dependently upregulated from 0 to 1 h in MCF-7 cells, its expression was stable in MDA-MB-231 cells for up to 4 h.

To indirectly assess the autophagosome complex formation at different time periods of drug treatment, the expression profile of Atg5 and Atg12, which are critical molecules for the elongation of the autophagosomal membrane, were also determined by immunoblotting. The basal expression levels of Atg5 and Atg12 were found to be higher in MCF-7 compared to MDA-MB-231 cells. In general, while roscovitine decreased the expression of Atg5 within 24 h, the Atg5 expression level was increased after 1 h of drug treatment in the MDA-MB-231 breast cancer cell line (Fig. 5A). Another key marker of autophagosomal formation is LC3A/B, which integrates to double membranes; the level of this protein was increased after 8 h of roscovitine treatment in MCF-7 cells. However, the expression level of LC3A/B was decreased after 24 h of drug treatment. When we examined the autophagic effect of roscovitine on LC3A/B expression in MDA-MB-231 cells, we observed a rapid upregulation within 30 min and an overall higher basal level compared to MCF-7 cells. In addition, LC3A/B expression was higher in MDA-MB-231 cells after 24 h of drug treatment compared to MCF-7 cells. When the MCF-7 cells were treated with roscovitine for 72 h, the expression of the autophagic key markers LC3A/B, Atg5 and Atg12 was decreased from the first 24 h. However, the protein levels of these markers appeared increased again after 72 h of drug treatment in MCF-7 cells. By contrast, the expression levels of LC3A/B, Atg5 and Atg12 appeared increased after 48 h of drug treatment in the MDA-MB-231 cell line (Fig. 5B). Based on these results, we conclude that MCF-7 cells are more sensitive to roscovitine-induced autophagy than MDA-MB-231 cells.

Polyamines modulate roscovitine induced autophagy

In order to further explore the role of polyamines in drug-induced autophagy, cells were treated with roscovitine in the presence of Spd or Spm for 24 h. Spd was not an autophagy inducer but Spm was a good candidate to induce autophagy by upregu-lating beclin-1 and Atg5 in MCF-7 and MDA-MB-231 cells (Fig. 6A). Moreover, cleaved fragments of LC3A/B were observed following Spm treatment in MDA-MB-231 cells. After treatment with Spd alone, beclin-1 and Atg5 expression levels decreased. By contrast, after treatment with Spm alone, the beclin-1 and Atg5 expression levels increased. Co-treatment with roscovitine and Spm showed opposite effects on autophagic marker expression in the two breast cancer cell lines compared to co-treatment with roscovitine and Spd (Fig. 6A). These results were confirmed in MCF-7 cells by MDC staining, which allows to detect the autophagic vacuoles (Fig. 6B).

Discussion

The majority of malignancies are associated with the loss of functional cell-cycle control, which results in impaired apoptosis and unlimited growth. An emerging anticancer approach is to control the aberrant cell cycle machinery by evaluating key molecules for drug design. As shown in previous studies, CDK inhibitors exert their apoptotic effect by causing cell-cycle arrest (10,26,27). Roscovitine is a promising CDK inhibitor with high apoptotic potential in malignant cells. It competitively binds to the ATP binding site of CDKs and prevents cyclin-CDK complex formation (2831). Furthermore, roscovitine is the first orally bioavailable CDK inhibitor in clinical trials for B-cell malignancies and lung cancer (31,32). A previous study indicated that roscovitine induces apoptosis in breast cancer cells by causing cell-cycle arrest at the G2/M phase (9). PAs are key regulators of cellular processes such as transcription, translation and proliferation (33). PA metabolic enzymes have been proposed as targets for antineoplastic therapy in breast cancer, since their high intracellular level was found associated with rapid cel-cycle turnover in these cells compared to healthy breast tissue cells (14,3437). In the present study, we demonstrated that roscovitine decreases cell viability in a dose-dependent manner in the MCF-7 and MDA-MB-231 breast cancer cell lines (Fig. 1A). We also determined that the combination of Spd or Spm with roscovitine can enhance drug-induced cytotoxicity in both breast cancer cell lines (Fig. 1B and C). MCF-7 and MDA-MB-231 cells have a different expression status for ERα, which regulates the transcription of genes such as CDK2, a target of roscovitine. CDK2 has been also shown to enhance the ligand-independent ERα activation (3840), which indicates that this protein can play a critical role in the responsiveness against the hormone ablation therapy (5,41,42). Similar to previous findings (9,10), roscovitine inhibited the proliferation rates to different degrees in ERα-positive and -negative breast cancer cell lines in our study.

Exposure of cancer cells to PAs may affect the modulation of cell responses to drug treatment in a cell-dependent manner. While Spd treatment protected Erhlich ascite tumor cells against apoptosis triggered by acetoxychavicol acetate (43), Spm was shown to synergistically act with bovine serum oxidase in docetaxel-induced apoptosis in MCF-7 cells (44). According to a previous study by our group, roscovitine-induced apoptotic cell death may be altered when PA biosynthesis is inhibited in HCT116 colon carcinoma cells (45).

Although increased accumulation of intracellular PAs is associated with disease progression and rapid cell-cycle turnover, due to high PA catabolic activity, Spm may induce apoptosis by activating cellular caspases (4648).

Cell death in vertebrates mostly proceeds via the mitochondrial pathway in a caspase-dependent manner (49,50). CDK inhibitors were shown to exert their apoptotic effect through inducing MMP loss and activating caspases in cancer cells (51,52). Similar to these observations, we determined that exposure of cells to roscovitine for 24 h induces the modulation of MMP in the MCF-7 and MDA-MB-231 breast cancer cell lines. However, combined treatment of Spd or Spm with roscovitine caused different effects in both cell lines. Spm protected cells against roscovitine-induced mitochondria-mediated apoptosis in MCF-7, but not in MDA-MB-231 breast cancer cells (Fig. 3). According to the results of the present study, there was a difference in the two breast cancer cell lines treated with roscovitine and PAs, with regards to cell death response. The MCF-7 and MDA-MB-231 cell lines have different genetic backgrounds, particularly with regards to ER status, which is associated with cellular growth and the fate of cells. Therefore, it may be hypothesized that the difference in cell death response between these cell lines is due to key targets within the ER. Furthermore, hormone signaling may promote different cell signaling pathways to induce either apoptosis or autophagy (2,16). In addition, PAs also have a role in cell growth, and the treatment of the cells with PAs resulted in an altered cell response to roscovitine treatment. Therefore, MCF-7 and MDA-MB-231 cells may act differently upon drug exposure. In association with these data, we showed that in the MCF-7 breast cancer cell line, roscovitine treatment for 24 h results in the cleavage of pro-caspase-9 and -7, which is referred to in the literature as the initial eevent during induction of apoptosis. Upon treatment with roscovitine, additional Spm exposure affected the activation of both caspases in the MCF-7 and MDA-MB-231 breast cancer cell lines. However, treatment with roscovitine only did not exert the same effect on the MDA-MB-231 cells (Fig. 4).

In the second part of the present study, we investigated the role of the CDK inhibitor on autophagy and the potential role of PAs on autophagic regulation in MCF-7 and MDA-MB-231 breast cancer cell lines. The therapeutic efficiencies of drug candidates for cancer treatment were investigated in recent studies by examination of their potential to activate both apoptosis and autophagy, and by studying their interactions (53,54). Therefore, elucidation of the molecular mechanism common to apoptosis and autophagy, as well as of the crosstalk between these two processes is of high importance. Inhibition of autophagy has been shown to enhance the induction of apoptosis (55,56). Under cellular stress conditions, such as in the presence of DNA-damaging agents, autophagy is inhibited and the intrinsic pathway of apoptosis is triggered in MCF-7 cells, but the induction of autophagy can delay apoptosis (57).

In association with these findings, we found that 24-h treatment with roscovitine modulates the mechanism underlying autophagy in MDA-MB-231, but not in MCF-7 cells (Fig. 5A). Longer exposure of both cell lines to roscovitine confirmed that the autophagic process is more prominent in MDA-MB-231 cells compared to MCF-7 cells. Therefore, we conclude that MCF-7 cells are more sensitive to roscovitine-induced autophagy than MDA-MB-231 cells (Fig. 5B).

In general, autophagy delays cell death and prolongs the lifespan in various experimental aging models (5860). Recent studies showed that PAs, and in particular Spd, induce autophagy and cause increased lifespan. For instance, naphthalimide-PA conjugates trigerred autophagy by modulating the mTOR signaling cascade. Exposure of HepG2 cells to the naphthalimide-PA conjugates induced autophagic vesicle formation (61,62). In a similar way, Spd treatment can induce LC3 formation in HeLa cells (63). Therefore, Spd-induced autophagy may be therapeutically useful for cancer treatment. Indeed, increased levels of highly and positively-charged PAs have been found to correlate with chromatin condensation, and to modulate HAT and HDAC activities in murine skin tumors (24,25). However, in yeast, Spd treatment has been shown to trigger the global hypoacetylation of histone H3 and selectively acetylate the promoter region of the atg7 gene, which led to the upregulation of autophagic genes (23,63). According to our findings, Spm may be proposed as an autophagic agent in MCF-7 and MDA-MB-231 cells (Fig. 6).

Therefore, we conclude that roscovitine is a mediator of apoptosis in the ERα+ MCF-7 breast cancer cells, and that apoptosis is delayed by the induction of autophagy in ERα MDA-MB-231 cells. In addition, PAs play critical roles in roscovitine-induced autophagy in a cell type-dependent manner.

References

1 

Jemal A, Siegel R, Ward E, Hao Y, Xu J and Thun MJ: Cancer statistics, 2009. CA Cancer J Clin. 59:225–249. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Lerner LJ and Jordan VC: Development of antiestrogens and their use in breast cancer: eighth Cain memorial award lecture. Cancer Res. 50:4177–4189. 1990.PubMed/NCBI

3 

Jaiyesimi IA, Buzdar AU, Decker DA and Hortobagyi GN: Use of tamoxifen for breast cancer: twenty-eight years later. J Clin Oncol. 13:513–529. 1995.PubMed/NCBI

4 

Buzdar AU and Hortobagyi G: Update on endocrine therapy for breast cancer. Clin Cancer Res. 4:527–534. 1998.PubMed/NCBI

5 

Nair BC and Vadlamudi RK: Regulation of hormonal therapy resistance by cell cycle machinery. Gene Ther Mol Biol. 12:3952008.PubMed/NCBI

6 

Al-Minawi AZ, Saleh-Gohari N and Helleday T: The ERCC1/XPF endonuclease is required for efficient single-strand annealing and gene conversion in mammalian cells. Nucleic Acids Res. 36:1–9. 2008. View Article : Google Scholar :

7 

Hunt T: You never know: Cdk inhibitors as anti-cancer drugs. Cell Cycle. 7:3789–3790. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Aldoss IT, Tashi T and Ganti AK: Seliciclib in malignancies. Expert Opin Investig Drugs. 18:1957–1965. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Wojciechowski J, Horky M, Gueorguieva M and Wesierska-Gadek J: Rapid onset of nucleolar disintegration preceding cell cycle arrest in roscovitine-induced apoptosis of human MCF-7 breast cancer cells. Int J Cancer. 106:486–495. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Wesierska-Gadek J, Gueorguieva M and Horky M: Roscovitine-induced up-regulation of p53AIP1 protein precedes the onset of apoptosis in human MCF-7 breast cancer cells. Mol Cancer Ther. 4:113–124. 2005.PubMed/NCBI

11 

Appleyard MV, O’Neill MA, Murray KE, et al: Seliciclib (CYC202, R-roscovitine) enhances the antitumor effect of doxorubicin in vivo in a breast cancer xenograft model. Int J Cancer. 124:465–472. 2009. View Article : Google Scholar

12 

Charollais RH and Mester J: Resumption of cell cycle in BALB/c-3T3 fibroblasts arrested by polyamine depletion: relation with ‘competence’ gene expression. J Cell Physiol. 137:559–564. 1988. View Article : Google Scholar : PubMed/NCBI

13 

Harada JJ and Morris DR: Cell cycle parameters of Chinese hamster ovary cells during exponential, polyamine-limited growth. Mol Cell Biol. 1:594–599. 1981.PubMed/NCBI

14 

Pegg AE: Polyamine metabolism and its importance in neoplastic growth and a target for chemotherapy. Cancer Res. 48:759–774. 1988.PubMed/NCBI

15 

Deng W, Jiang X, Mei Y, et al: Role of ornithine decarboxylase in breast cancer. Acta Biochim Biophys Sin (Shanghai). 40:235–243. 2008. View Article : Google Scholar

16 

Hoggard N and Green CD: Polyamines and growth regulation of cultured human breast cancer cells by 17 beta-oestradiol. Mol Cell Endocrinol. 46:71–78. 1986. View Article : Google Scholar : PubMed/NCBI

17 

Manni A: Polyamine involvement in breast cancer phenotype. In Vivo. 16:493–500. 2002.PubMed/NCBI

18 

Bello-Fernandez C, Packham G and Cleveland JL: The ornithine decarboxylase gene is a transcriptional target of c-Myc. Proc Natl Acad Sci USA. 90:7804–7808. 1993. View Article : Google Scholar : PubMed/NCBI

19 

Celano P, Baylin SB, Giardiello FM, Nelkin BD and Casero RA Jr: Effect of polyamine depletion on c-myc expression in human colon carcinoma cells. J Biol Chem. 263:5491–5494. 1988.PubMed/NCBI

20 

Kondo Y, Kanzawa T, Sawaya R and Kondo S: The role of autophagy in cancer development and response to therapy. Nat Rev Cancer. 5:726–734. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Gozuacik D and Kimchi A: Autophagy and cell death. Curr Top Dev Biol. 78:217–245. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Maiuri MC, Zalckvar E, Kimchi A and Kroemer G: Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol. 8:741–752. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Madeo F, Eisenberg T, Büttner S, Ruckenstuhl C and Kroemer G: Spermidine: a novel autophagy inducer and longevity elixir. Autophagy. 6:160–162. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Hobbs CA and Gilmour SK: High levels of intracellular polyamines promote histone acetyltransferase activity resulting in chromatin hyperacetylation. J Cell Biochem. 77:345–360. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Hobbs CA, Paul BA and Gilmour SK: Deregulation of polyamine biosynthesis alters intrinsic histone acetyltransferase and deacetylase activities in murine skin and tumors. Cancer Res. 62:67–74. 2002.PubMed/NCBI

26 

Wesierska-Gadek J, Wandl S, Kramer MP, Pickem C, Krystof V and Hajek SB: Roscovitine up-regulates p53 protein and induces apoptosis in human HeLaS(3) cervix carcinoma cells. J Cell Biochem. 105:1161–1171. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Wesierska-Gadek J, Gueorguieva M, Wojciechowski J and Horky M: Cell cycle arrest induced in human breast cancer cells by cyclin-dependent kinase inhibitors: a comparison of the effects exerted by roscovitine and olomoucine. Pol J Pharmacol. 56:635–641. 2004.PubMed/NCBI

28 

Meijer L, Borgne A, Mulner O, et al: Biochemical and cellular effects of roscovitine, a potent and selective inhibitor of the cyclin-dependent kinases cdc2, cdk2 and cdk5. Eur J Biochem. 243:527–536. 1997. View Article : Google Scholar : PubMed/NCBI

29 

Fischer PM and Gianella-Borradori A: CDK inhibitors in clinical development for the treatment of cancer. Expert Opin Investig Drugs. 12:955–970. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Hahntow IN, Schneller F, Oelsner M, et al: Cyclin-dependent kinase inhibitor Roscovitine induces apoptosis in chronic lymphocytic leukemia cells. Leukemia. 18:747–755. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Decker T, Hipp S, Hahntow I, Schneller F and Peschel C: Expression of cyclin E in resting and activated B-chronic lymphocytic leukaemia cells: cyclin E/cdk2 as a potential therapeutic target. Br J Haematol. 125:141–148. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Benson C, White J, De Bono J, et al: A phase I trial of the selective oral cyclin-dependent kinase inhibitor seliciclib (CYC202; R-Roscovitine), administered twice daily for 7 days every 21 days. Br J Cancer. 96:29–37. 2007. View Article : Google Scholar

33 

Zaletok S, Alexandrova N, Berdynskykh N, et al: Role of polyamines in the function of nuclear transcription factor NF-kappaB in breast cancer cells. Exp Oncol. 26:221–225. 2004.PubMed/NCBI

34 

Wang Y and Casero RA Jr: Mammalian polyamine catabolism: a therapeutic target, a pathological problem, or both? J Biochem. 139:17–25. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Persson L and Rosengren E: Increased formation of N1-acetylspermidine in human breast cancer. Cancer Lett. 45:83–86. 1989. View Article : Google Scholar : PubMed/NCBI

36 

Cañizares F, Salinas J, de las Heras M, et al: Prognostic value of ornithine decarboxylase and polyamines in human breast cancer: correlation with clinicopathologic parameters. Clin Cancer Res. 5:2035–2041. 1999.PubMed/NCBI

37 

Casero RA Jr and Marton LJ: Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nat Rev Drug Discov. 6:373–390. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Ali S and Coombes RC: Endocrine-responsive breast cancer and strategies for combating resistance. Nat Rev Cancer. 2:101–112. 2002. View Article : Google Scholar

39 

Loi S, Haibe-Kains B, Desmedt C, et al: Definition of clinically distinct molecular subtypes in estrogen receptor-positive breast carcinomas through genomic grade. J Clin Oncol. 25:1239–1246. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Sutherland RL and Musgrove EA: CDK inhibitors as potential breast cancer therapeutics: new evidence for enhanced efficacy in ER+ disease. Breast Cancer Res. 11:1122009. View Article : Google Scholar

41 

Rogatsky I, Trowbridge JM and Garabedian MJ: Potentiation of human estrogen receptor alpha transcriptional activation through phosphorylation of serines 104 and 106 by the cyclin A-CDK2 complex. J Biol Chem. 274:22296–22302. 1999. View Article : Google Scholar : PubMed/NCBI

42 

Trowbridge JM, Rogatsky I and Garabedian MJ: Regulation of estrogen receptor transcriptional enhancement by the cyclin A/Cdk2 complex. Proc Natl Acad Sci USA. 94:10132–10137. 1997. View Article : Google Scholar : PubMed/NCBI

43 

Moffatt J, Hashimoto M, Kojima A, et al: Apoptosis induced by 1′-acetoxychavicol acetate in Ehrlich ascites tumor cells is associated with modulation of polyamine metabolism and caspase-3 activation. Carcinogenesis. 21:2151–2157. 2000. View Article : Google Scholar

44 

Marra M, Lombardi A, Agostinelli E, et al: Bovine serum amine oxidase and spm potentiate docetaxel and interferon-alpha effects in inducing apoptosis on human cancer cells through the generation of oxidative stress. Biochim Biophys Acta. 1783:2269–2278. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Arisan ED, Coker A and Palavan-Ünsal N: Polyamine depletion enhances the roscovitine-induced apoptosis through the activation of mitochondria in HCT116 colon carcinoma cells. Amino Acids. 42:655–665. 2012. View Article : Google Scholar

46 

Xie X, Tome ME and Gerner EW: Loss of intracellular putrescine pool-size regulation induces apoptosis. Exp Cell Res. 230:386–392. 1997. View Article : Google Scholar : PubMed/NCBI

47 

Stefanelli C, Bonavita F, Stanic I, et al: Spermine causes caspase activation in leukaemia cells. FEBS Lett. 437:233–236. 1998. View Article : Google Scholar : PubMed/NCBI

48 

Stefanelli C, Stanic I, Zini M, et al: Polyamines directly induce release of cytochrome c from heart mitochondria. Biochem J. 347:875–880. 2000. View Article : Google Scholar : PubMed/NCBI

49 

Ouyang L, Shi Z, Zhao S, et al: Programmed cell death pathways in cancer: a review of apoptosis, autophagy and programmed necrosis. Cell Prolif. 45:487–498. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Green DR and Kroemer G: The pathophysiology of mitochondrial cell death. Science. 305:626–629. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Yenugonda VM, Deb TB, Grindrod SC, et al: Fluorescent cyclin-dependent kinase inhibitors block the proliferation of human breast cancer cells. Bioorg Med Chem. 19:2714–2725. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Ringer L, Sirajuddin P, Yenugonda VM, et al: VMY-1-103, a dansylated analog of purvalanol B, induces caspase-3-dependent apoptosis in LNCaP prostate cancer cells. Cancer Biol Ther. 10:320–325. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Liu B, Wu JM, Li J, et al: Polygonatum cyrtonema lectin induces murine fibrosarcoma L929 cell apoptosis and autophagy via blocking Ras-Raf and PI3K-Akt signaling pathways. Biochimie. 92:1934–1938. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Cheng Y, Qiu F, Huang J, Tashiro S, Onodera S and Ikejima T: Apoptosis-suppressing and autophagy-promoting effects of calpain on oridonin-induced L929 cell death. Arch Biochem Biophys. 475:148–155. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Lambert LA, Qiao N, Hunt KK, et al: Autophagy: a novel mechanism of synergistic cytotoxicity between doxorubicin and roscovitine in a sarcoma model. Cancer Res. 68:7966–7974. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Amaravadi RK, Yu D, Lum JJ, et al: Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest. 117:326–336. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Boya P, González-Polo RA, Casares N, et al: Inhibition of macroautophagy triggers apoptosis. Mol Cell Biol. 25:1025–1040. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Abedin MJ, Wang D, McDonnell MA, Lehmann U and Kelekar A: Autophagy delays apoptotic death in breast cancer cells following DNA damage. Cell Death Differ. 14:500–510. 2007. View Article : Google Scholar

59 

Jia K and Levine B: Autophagy is required for dietary restriction-mediated life span extension in C. elegans. Autophagy. 3:597–599. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Meléndez A, Tallóczy Z, Seaman M, Eskelinen EL, Hall DH and Levine B: Autophagy genes are essential for dauer development and life-span extension in C. elegans. Science. 301:1387–1391. 2003. View Article : Google Scholar : PubMed/NCBI

61 

Tavernarakis N, Pasparaki A, Tasdemir E, Maiuri MC and Kroemer G: The effects of p53 on whole organism longevity are mediated by autophagy. Autophagy. 4:870–873. 2008. View Article : Google Scholar : PubMed/NCBI

62 

Tian ZY, Xie SQ, Mei ZH, Zhao J, Gao WY and Wang CJ: Conjugation of substituted naphthalimides to polyamines as cytotoxic agents targeting the Akt/mTOR signal pathway. Org Biomol Chem. 7:4651–4660. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Eisenberg T, Knauer H, Schauer A, et al: Induction of autophagy by spermidine promotes longevity. Nat Cell Biol. 11:1305–1314. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2015
Volume 11 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Arisan ED, Akkoç Y, Akyüz KG, Kerman EM, Obakan P, Çoker‑Gürkan A and Ünsal NP: Polyamines modulate the roscovitine-induced cell death switch decision autophagy vs. apoptosis in MCF-7 and MDA-MB-231 breast cancer cells. Mol Med Rep 11: 4532-4540, 2015
APA
Arisan, E.D., Akkoç, Y., Akyüz, K.G., Kerman, E.M., Obakan, P., Çoker‑Gürkan, A., & Ünsal, N.P. (2015). Polyamines modulate the roscovitine-induced cell death switch decision autophagy vs. apoptosis in MCF-7 and MDA-MB-231 breast cancer cells. Molecular Medicine Reports, 11, 4532-4540. https://doi.org/10.3892/mmr.2015.3303
MLA
Arisan, E. D., Akkoç, Y., Akyüz, K. G., Kerman, E. M., Obakan, P., Çoker‑Gürkan, A., Ünsal, N. P."Polyamines modulate the roscovitine-induced cell death switch decision autophagy vs. apoptosis in MCF-7 and MDA-MB-231 breast cancer cells". Molecular Medicine Reports 11.6 (2015): 4532-4540.
Chicago
Arisan, E. D., Akkoç, Y., Akyüz, K. G., Kerman, E. M., Obakan, P., Çoker‑Gürkan, A., Ünsal, N. P."Polyamines modulate the roscovitine-induced cell death switch decision autophagy vs. apoptosis in MCF-7 and MDA-MB-231 breast cancer cells". Molecular Medicine Reports 11, no. 6 (2015): 4532-4540. https://doi.org/10.3892/mmr.2015.3303