Etoposide induces apoptosis via the mitochondrial- and caspase-dependent pathways and in non-cancer stem cells in Panc-1 pancreatic cancer cells

  • Authors:
    • She-Hong Zhang
    • Qian Huang
  • View Affiliations

  • Published online on: October 1, 2013     https://doi.org/10.3892/or.2013.2767
  • Pages: 2765-2770
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pancreatic cancer is a highly aggressive malignant tumor. In the present study, we performed several methods, including CCK-8 assay, immunofluorescence technique, western blotting and flow cytometry, to determine the effects of VP16 (etoposide) on Panc-1 pancreatic cancer cells. The results demonstrated that VP16 inhibited the growth of and induced apoptosis in Panc-1 cells. Western blot analysis showed that VP16 inhibited the expression of Bcl-2 and enhanced the expression of Bax, caspases-3 and -9, cytochrome c and PARP. Notably, a strong inhibitory effect of VP16 on Panc-1 cells mainly occurred in non-CSCs. These data provide a new strategy for the therapy of pancreatic cancer.

Introduction

Pancreatic carcinoma is a refractory disease with few effective therapies (1). Only 18% of patients survive 1 year after diagnosis and the 5-year survival rate is 4%. By the time patients exhibit symptoms and the disease is diagnosed, it is already beyond the early stages (2,3). The capacity of a tumor to grow and proliferate is dependent on a small subset of cells, the cancer stem cells (CSCs), which are immature cells that can replicate or self-renew, and differentiate or grow into different types of cancer cells (4).

VP16 is an important chemotherapeutic agent that is used to treat a wide spectrum of human cancers. It has been in clinical use for more than 2 decades and remains one of the most highly prescribed anticancer drugs in the world (5). This drug is a pro-apoptosis agent which triggers cell death pathways (6).

In the present study, we examined the effect of VP16 on human pancreatic cancer cells and the change of pancreatic CSCs.

Materials and methods

Cell lines

The Panc-1 human pancreatic cancer cell line was purchased from the Shanghai Cell Bank (Shanghai, China).

Cell growth inhibition assays

The inhibitory effect of VP16 (etoposide; Qilu Pharmaceutical Co., Ltd., Shandong, China) on the growth of Panc-1 cells was evaluated by CCK-8 kit (Sigma-Aldrich, Ireland) (7). In brief, 200 μl medium/well containing 2×103 cells were seeded in 96-well microtiter plates for the CCK-8 assays. After treatment with VP16, cells in the 96-well plates were used for CCK-8 assays. The optical density was measured at 490 nm using a microplate reader (SpectraMax 340; Molecular Devices Co., Sunnyvale, CA, USA).

The cell viability ratio was calculated using the following formula: Inhibitory ratio (%) = (ODcontrol − ODtreated)/ODcontrol × 100% (8). Results from 3 independent experiments in triplicates are presented.

Cell cycle analysis

Cell cycle distribution was analyzed as previously described (9). In brief, 1×106 cells were incubated with the indicated amounts (0–300 μg/ml) of VP16 for 48 h. Cells were collected and then fixed in 70% ethanol at 4°C for 24 h. Cells were washed again with PBS and incubated with PI (10 μg/ml) with simultaneous RNase treatment at 37°C for 30 min. Cell DNA content was measured using a FACStar flow cytometer (Becton-Dickinson, San Jose, CA, USA).

Annexin V/PI analysis

Apoptosis was determined by staining cells with Annexin V-fluorescein isothiocyanate as previously described (10). In brief, 1×106 cells were incubated with the indicated concentrations (0–300 μg/ml) of VP16 for 48 h. Five microliters Annexin V-FITC and 5 μl PI were added. Cells were vortexed and incubated for 15 min in the dark. Binding buffer (200 μl) was added to each tube. Flow cytometric analysis was performed immediately after staining. All assays were independently repeated for 3 times.

Mitochondrial membrane potential (Δψm) assay

The loss of Δψm in the cells is one of the mechanisms of induction of apoptosis, which has been linked to the initiation and activation of apoptotic cascades (11). With a variety of stimuli, including the translocation of Bax from the cytosol to the mitochondria, this event occurs, which triggers the release of cytochrome c from the mitochondria to the cytosol (12). In brief, 1×106 cells were incubated with the indicated amounts (0–300 μg/ml) of VP16. Cells were incubated with JC-1 dye for 15 min at 37°C, and then resuspended in 200 μl PBS for FACS analysis.

Protein extraction and western blot assays

The expressions of proteins were evaluated using western blot analysis, as previously described (13). In brief, cells were treated with different concentrations of VP16 (0–300 μg/ml) for 48 h. Cells were collected and suspended in 5 volumes of lysis buffer (20 mM HEPES, pH 7.9, 20% glycerol, 200 mM KCl, 0.5 mM EDTA, 0.5% NP-40, 0.5 mM DTT, 1% protease inhibitor cocktail). Lysates were collected. Supernatant samples containing 40 μg total protein were loaded on SDS-PAGE gel for electrophoresis. Membranes were blocked for 1 h with 5% milk. Membranes were then incubated overnight at 4°C with primary antibodies (caspases-3 and -9, cytochrome c, PARP, Bcl-2, Bax and β-actin) and with horseradish peroxidase-conjugated secondary antibodies for 1.5 h at room temperature. The experiment was repeated 3 times. Nuclear and Cytoplasmic Protein Extraction kit was used to extract protein of cytochrome c.

Quantification of caspase-3 activity

Caspase-3 activity was analyzed using a caspase-3 activity assay kit (Beyotime, China). Cells were treated with the indicated reagents for 24 h and cell lysates were prepared for the following experiment according to the manufacturer’s instructions.

Change of antigens of CSCs by flow cytometry

Previous studies found that malignant tumors were heterogeneous. This subpopulation of CSCs has been demonstrated to be responsible for tumor initiation, proliferation, recurrence and resistance to chemotherapy (1418). On the basis of the above findings, we hypothesized that the ratio of CSCs and non-CSCs must change during treatment of VP16. Cells were treated with VP16 (100 μg/ml) for 48 h, and then flow cytometry was performed to detect expression of CSC markers, such as CD133, CD44, CD24 and ESA antigens (BioLegend Inc., San Diego, CA, USA). Each analysis included 100,000 events. Isotype-matched mouse IgG was used as a control.

Statistical analysis

All data are expressed as means ± SD. Statistical comparisons of results were made using analysis of variance (ANOVA). Differences between groups were determined using the Student’s t-test for unpaired observations and P-values <0.05 were considered to indicate statistically significant differences.

Results

VP16 induces apoptosis of Panc-1 cells

A dose-dependent reduction in cell growth was observed. The ratio of inhibition with different concentrations of VP16 is shown in Fig. 1. These results indicated that VP16 inhibited Panc-1 cell proliferation in a dose- and time-dependent manner.

Effects of VP16 on cell cycle and apoptosis in Panc-1 cells

When we stained Panc-1 cells with Annexin V-FITC and PI, the proportion of apoptotic cells was significantly increased in a dose-dependent manner (Fig. 2).

The effect of VP16 on cell cycle distribution indicated that there was a higher number of cells in the G1 phase, in higher concentration of VP16 compared with the control (P<0.05) and, at the same time, the number of S phase cells was significantly decreased (Fig. 3).

VP16 induces loss of Δψm and subsequently enhances the release of cytochrome c in Panc-1 cells

VP16 in Panc-1 cells resulted in a dose-dependent increase in the number of JC-1 dye-positive cells from 8.67 to 92.9%. Western blot analysis revealed that VP16 caused a dose-dependent increase in the release of cytochrome c into cytosol, which confirmed the disruption of the Δψm after VP16 treatment (Fig. 4).

Involvement of caspase-3 activation

Caspases are a family of cysteine proteases that are essentially involved in the apoptotic pathway. We investigated whether caspase-3 was involved in apoptosis induced by VP16 in Panc-1 cells. The data showed that activity of caspase-3 increased significantly in a dose- and time-dependent manner in the cells treated with VP16 (Fig. 5A). In addition, activity of caspase-3 decreased in the combined treatment with VP16 and caspase-3 inhibitor (Ac-DEVD-CHO) compared with the VP16 treatment group (Fig. 5B). The results demonstrated that VP16-induced apoptosis was dependent on caspase-3.

Expression of apoptosis-related proteins in VP16-treated cells

These data showed that treatment of VP16 in Panc-1 cells resulted in a dose- and time-dependent reduction in the levels of the anti-apoptotic proteins and an increase in the level of pro-apoptotic proteins (Fig. 6).

Cells treated with VP16 highly express CD44, CD24, ESA and CD133

Results from the above studies and hypotheses of CSCs, prompted us to study the change of CSC-related markers. The data were: CD133, 0.31 vs. 1.54%; CD24, 19.51 vs. 69.47%; CD44, 21.55 vs. 93.23%; ESA, 1.51 vs. 8.38% (P<0.05, Fig. 7). The data demonstrated that most of the cells killed by VP16 were non-CSCs and that CSCs derived from Panc-1 cells played key role in chemotherapy resistance.

Discussion

In the present study, we investigated the molecular mechanisms of VP16-induced apoptosis in Panc-1 cells and the change of markers of CSCs. VP16 inhibited the growth of cells in a dose- and time-dependent manner. In addition, VP16 markedly induced apoptosis in Panc-1 cells in the same manner, which was confirmed by Annexin V and PI staining. Based on the above assays, we observed a strong growth inhibitory effect of VP16 in Panc-1 cells; we then determined the possible mechanism of anti-proliferative activity of VP16. Our data demonstrated that treatment with VP16 in Panc-1 cells induced G1 phase arrest of cell cycle, indicating that one of the mechanisms was inhibition of cell cycle progression. Loss of mitochondrial membrane potential was suicidal to cells as they became bioenergetically deficient (30) and that led to release of cytochrome c into the cytosol. As the level of cytochrome c increased in the cytosol, it interacted with Apaf-1 and ATP formed a complex with pro-caspase-9, leading to activation of caspases-9 and -3 which led to the cleavage of PARP (19,20).

Mitochondria depolarization, considered an irreversible step in the apoptosis process, could trigger a cascade of caspases. The mitochondrion was a major subcellular compartment where the Bcl-2 family members interacted with each other or exerted function independently (21).

The Bcl-2 family of proteins include proteins that could either inhibit (Bcl-2, Bcl-xL, etc.) or induce (Bax, Bak, Bad, etc.) apoptosis. The anti-apoptotic proteins prevented cytochrome c release by forming heterodimer complexes with pro-apoptotic Bcl-2 family proteins (22).

The pro-apoptotic Bcl-2 proteins including Bax and Bak facilitate release of apoptogenic molecules from mitochondria to the cytosol and accelerate apoptotic cell death (2325).

To evaluate the role of caspases, the master executioner during apoptotic cell death, in VP16-induced cell death, the activities of caspase-3 were assessed using the caspase-3 colorimetric assay kits. The data suggested that caspase-3 inhibitor could significantly inhibit the activities of caspase-3 in the VP16-induced cell death, indicating that apoptosis in VP16-treated Panc-1 cells occurred in a caspase-dependent manner.

VP16 induces apoptosis in various cancer cell lines and performs antitumor activity (35). In the present study, we observed a strong inhibitory effect of VP16 on Panc-1 cells in vitro, indicating chemotherapy drugs could kill cancer cells. However, pancreatic cancer patients continue to have a dismal prognosis with an average overall median survival of 4–6 months. The overall 5-year survival is <5% (1). The preliminary data suggested that tumor-cell repopulation describes the continuing proliferation of surviving tumor stem cells that can occur during a course of radiotherapy or chemotherapy (2629).

These findings led to our investigation of the role of CSCs during cytotoxic cancer therapy. Our results suggested that CSCs were more resistant to VP16 and most CSCs could escape cytotoxic cancer therapy. The relationship between CSCs and repopulation during the process of radiotherapy and chemotherapy will be our next research project.

Acknowledgements

The authors thank the participants for taking part in the study and for the laboratory assistance.

References

1 

Jemal A, Siegel R, Ward E, et al: Cancer statistics, 2007. CA Cancer J Clin. 57:43–66. 2007. View Article : Google Scholar

2 

Niederhuber JE, Brennan MF and Menck HR: The National Cancer Data Base report on pancreatic cancer. Cancer. 76:1671–1677. 1995. View Article : Google Scholar : PubMed/NCBI

3 

Jemal A, Thomas A, Murray T, et al: Cancer statistics, 2002. CA Cancer J Clin. 52:23–47. 2002. View Article : Google Scholar

4 

Bjerkvig R, Tysnes BB, Aboody KS, et al: Opinion: the origin of the cancer stem cell: current controversies and new insights. Nat Rev Cancer. 5:899–904. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Baldwin EL and Osheroff N: Etoposide, topoisomerase II and cancer. Curr Med Chem Anticancer Agents. 5:363–372. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Meresse P, Dechaux E, Monneret C, et al: Etoposide: discovery and medicinal chemistry. Curr Med Chem. 11:2443–2466. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Chen Y, Zhang J, Wang H, et al: miRNA-135a promotes breast cancer cell migration and invasion by targeting HOXA10. BMC Cancer. 12:1112012. View Article : Google Scholar : PubMed/NCBI

8 

Yingkun N, Lvsong Z and Huimin Y: Shikonin inhibits the proliferation and induces the apoptosis of human HepG2 cells. Can J Physiol Pharmacol. 88:1138–1146. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Kuo SY, Hsieh TJ, Wang YD, et al: Cytotoxic constituents from the leaves of Cinnamomum subavenium. Chem Pharm Bull. 56:97–101. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Madan E, Prasad S, Roy P, et al: Regulation of apoptosis by resveratrol through JAK/STAT and mitochondria mediated pathway in human epidermoid carcinoma A431 cells. Biochem Biophys Res Commun. 377:1232–1237. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Hu X, Wu N, Xia P, et al: Correlation between low-level expression of the tumor suppressor gene TAp73 and the chemoresistance of human glioma stem cells. Cancer Chemother Pharmacol. 69:1205–1212. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Yen YP, Tsai KS, Chen YW, et al: Arsenic induces apoptosis in myoblasts through a reactive oxygen species-induced endoplasmic reticulum stress and mitochondrial dysfunction pathway. Arch Toxicol. 86:923–933. 2012. View Article : Google Scholar

13 

Cha JD and Kim JY: Essential oil from Cryptomeria japonica induces apoptosis in human oral epidermoid carcinoma cells via mitochondrial stress and activation of caspases. Molecules. 17:3890–3901. 2012.PubMed/NCBI

14 

Reya T, Morrison SJ, Clarke MF, et al: Stem cells, cancer, and cancer stem cells. Nature. 414:105–111. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Singh SK, Hawkins C, Clarke ID, et al: Identification of human brain tumour initiating cells. Nature. 432:396–401. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Collins AT, Berry PA, Hyde C, et al: Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 65:10946–10951. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Li C, Heidt DG, Dalerba P, et al: Identification of pancreatic cancer stem cells. Cancer Res. 67:1030–1037. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Fukuda K, Saikawa Y, Ohashi M, et al: Tumor initiating potential of side population cells in human gastric cancer. Int J Oncol. 34:1201–1207. 2009.PubMed/NCBI

19 

Gross A, McDonnell JM and Korsmeyer SJ: BCL-2 family members and the mitochondria in apoptosis. Genes Dev. 13:1899–1911. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Wolf BB and Green DR: Suicidal tendencies: apoptotic cell death by caspase family proteinases. J Biol Chem. 274:20049–20052. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Kluck RM, Bossy-Wetzel E, Green DR, et al: The release of cytochrome c from mitochondria: a primary site for Bcl-2 regulation of apoptosis. Science. 275:1132–1136. 1997. View Article : Google Scholar : PubMed/NCBI

22 

Huppertz B, Kadyrov M and Kingdom JC: Apoptosis and its role in the trophoblast. Am J Obstet Gynecol. 195:29–39. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Tsujimoto Y and Shimizu S: Bcl-2 family: life-or-death switch. FEBS Lett. 466:6–10. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Hockenbery D, Nuñez G, Milliman C, et al: Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature. 348:334–336. 1990. View Article : Google Scholar : PubMed/NCBI

25 

Qiu B, Wang Y, Tao J, et al: Expression and correlation of Bcl-2 with pathological grades in human glioma stem cells. Oncol Rep. 28:155–160. 2012.PubMed/NCBI

26 

Davis AJ, Chapman W, Hedley DW, et al: Assessment of tumor cell repopulation after chemotherapy for advanced ovarian cancer: pilot study. Cytometry A. 51:1–6. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Stephens TC and Peacock JH: Tumour volume response, initial cell kill and cellular repopulation in B16 melanoma treated with cyclophosphamide and 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea. Br J Cancer. 36:313–321. 1977.PubMed/NCBI

28 

Wu L and Tannock IF: Repopulation in murine breast tumours during and after sequential treatments with cyclophosphamide and 5-fluorouracil. Cancer Res. 63:2134–2138. 2003.PubMed/NCBI

29 

Wang L, Huang X, Zheng X, et al: Enrichment of prostate cancer stem-like cells from human prostate cancer cell lines by culture in serum-free medium and chemoradiotherapy. Int J Biol Sci. 9:472–479. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Jin F, Zhao L, Guo YJ, et al: Influence of Etoposide on anti-apoptotic and multidrug resistance-associated protein genes in CD133 positive U251 glioblastoma stem-like cells. Brain Res. 1336:103–111. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December 2013
Volume 30 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang S and Zhang S: Etoposide induces apoptosis via the mitochondrial- and caspase-dependent pathways and in non-cancer stem cells in Panc-1 pancreatic cancer cells. Oncol Rep 30: 2765-2770, 2013
APA
Zhang, S., & Zhang, S. (2013). Etoposide induces apoptosis via the mitochondrial- and caspase-dependent pathways and in non-cancer stem cells in Panc-1 pancreatic cancer cells. Oncology Reports, 30, 2765-2770. https://doi.org/10.3892/or.2013.2767
MLA
Zhang, S., Huang, Q."Etoposide induces apoptosis via the mitochondrial- and caspase-dependent pathways and in non-cancer stem cells in Panc-1 pancreatic cancer cells". Oncology Reports 30.6 (2013): 2765-2770.
Chicago
Zhang, S., Huang, Q."Etoposide induces apoptosis via the mitochondrial- and caspase-dependent pathways and in non-cancer stem cells in Panc-1 pancreatic cancer cells". Oncology Reports 30, no. 6 (2013): 2765-2770. https://doi.org/10.3892/or.2013.2767