DOI QR코드

DOI QR Code

Effects of Rapamycin on Cell Apoptosis in MCF-7 Human Breast Cancer Cells

  • Published : 2015.01.22

Abstract

Background: Rapamycin is an effective anti-angiogenic drug. However, the mode of its action remains unclear. Therefore, in this study, we aimed to elucidate the antitumor mechanism of rapamycin, hypothetically via apoptotic promotion, using MCF-7 breast cancer cells. Materials and Methods: MCF-7 cells were plated at a density of $1{\times}10^5$ cells/well in 6-well plates. After 24h, cells were treated with a series of concentrations of rapamycin while only adding DMEM medium with PEG for the control regiment and grown at $37^{\circ}C$, 5% $CO_2$ and 95% air for 72h. Trypan blue was used to determine the cell viability and proliferation. Untreated and rapamycin-treated MCF-7 cells were also examined for morphological changes with an inverted-phase contrast microscope. Alteration in cell morphology was ascertained, along with a stage in the cell cycle and proliferation. In addition, cytotoxicity testing was performed using normal mouse breast mammary pads. Results: Our results clearly showed that rapamycin exhibited inhibitory activity on MCF-7 cell lines. The $IC_{50}$ value of rapamycin on the MCF-7 cells was determined as $0.4{\mu}g/ml$ (p<0.05). Direct observation by inverted microscopy demonstrated that the MCF-7 cells treated with rapamycin showed characteristic features of apoptosis including cell shrinkage, vascularization and autophagy. Cells underwent early apoptosis up to 24% after 72h. Analysis of the cell cycle showed an increase in the G0G1 phase cell population and a corresponding decrease in the S and G2M phase populations, from 81.5% to 91.3% and 17.3% to 7.9%, respectively. Conclusions: This study demonstrated that rapamycin may potentially act as an anti-cancer agent via the inhibition of growth with some morphological changes of the MCF-7 cancer cells, arrest cell cycle progression at G0/G1 phase and induction of apoptosis in late stage of apoptosis. Further studies are needed to further characterize the mode of action of rapamycin as an anti-cancer agent.

Keywords

References

  1. Aranovich A, Liu Q, Collins T, et al (2012). Differences in the mechanisms of proapoptotic BH3 proteins binding to Bcl-XL and Bcl-2 quantified in live MCF-7 cells. Molecular Cell, 45, 754-63. https://doi.org/10.1016/j.molcel.2012.01.030
  2. Bateman NW., Sun M, Hood BL, et al (2010). Defining central themes in breast cancer biology by differential proteomics: conserved regulation of cell spreading and focal adhesion kinase. J Proteome Res, 9, 5311-24. https://doi.org/10.1021/pr100580e
  3. Boffa DJ, LuanF, Thomas D, et al (2004). Rapamycin inhibits the growth and metastatic progression of non-small cell lung cancer. Clin Cancer Res, 10, 293-300. https://doi.org/10.1158/1078-0432.CCR-0629-3
  4. Gelmann EP, Thompson EW, Sommers CL (1992). Invasive and metastatic properties of MCF-7 cells and rasH-transfected MCF-7 cell lines. Int J Cancer, 50, 665-9. https://doi.org/10.1002/ijc.2910500431
  5. Geoerger B, Kerr K, Tang CB, et al (2001). Antitumor activity of the rapamycin analog CCI-779 in human primitive neuroectodermal tumor/medulloblastoma models as single agent and in combination chemotherapy. Cancer Res, 61, 1527-32.
  6. Gibbons JJ, Abraham RT, Yu K (2009). Mammalian target of rapamycin: discovery of rapamycin reveals a signaling pathway important for normal and cancer cell growth. Semin Oncol, 3, 3-17.
  7. Grunwald V, DeGraffenried L, Russel D, et al (2002). Inhibitors of mTOR reverse doxorubicin resistance conferred by PTEN status in prostate cancer cells. Cancer Res, 62, 6141-5.
  8. Guba M, von Breitenbuch P, Steinbauer M, et al (2002). Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nature Med, 8, 128-35. https://doi.org/10.1038/nm0202-128
  9. Hamelers IH, Van Schaik R, Sussenbach JS, Steenbergh PH (2003). $17{\beta}$-Estradiol responsiveness of MCF-7 laboratory strains is dependent on an autocrine signal activating the IGF type I receptor. Cancer Cell Int, 3, 10. https://doi.org/10.1186/1475-2867-3-10
  10. Hashemolhosseini S, Nagamine Y, Morley SJ, et al (1998). Rapamycin inhibition of the G1 to S transition is mediated by effects on cyclin D1 mRNA and protein stability. J Biol Chem, 273, 14424-9. https://doi.org/10.1074/jbc.273.23.14424
  11. Hidalgo M, Rowinsky EK (2000). The rapamycin-sensitive signal transduction pathway as a target for cancer therapy. Oncogene, 19, 6680-6. https://doi.org/10.1038/sj.onc.1204091
  12. Hosoi H, Dilling MB, Shikata T, et al (1999). Rapamycin causes poorly reversible inhibition of mTOR and induces p53-independent apoptosis in human rhabdomyosarcoma cells. Cancer Res, 59, 886-94.
  13. Huang S, Liu LN, Hosoi H, et al (2001). p53/p21CIP1 cooperate in enforcing rapamycin-induced G1 arrest and determine the cellular response to rapamycin. Cancer Res, 61, 3373-81.
  14. Idris FM, Mansor WNAW, Irfan M, Jalal A, Jaafar H (2014). Rapamycin and PF4 induce apoptosis by upregulating Bax and down-regulating survivin in MNU-induced breast cancer. Asian Pac J Cancer Prev, 15, 3939-44. https://doi.org/10.7314/APJCP.2014.15.9.3939
  15. Kawamata S, Sakaida H, Hori T, et al (1998). The upregulation of p27Kip1 by rapamycin results in G1 arrest in exponentially growing T-cell lines. Blood, 91, 561-9.
  16. Khemapech N, Pitchaiprasert S, Triratanachat S (2012). Prevalence and clinical significance of mammalian target of rapamycin phosphorylation (p-mTOR) and vascular endothelial growth factor (VEGF) in clear cell carcinoma of the ovary. Asian Pac J Cancer Prev, 13, 6357-62. https://doi.org/10.7314/APJCP.2012.13.12.6357
  17. Lai D, Ho KC, Hao Y, Yang X (2011). Taxol resistance in breast cancer cells is mediated by the hippo pathway component TAZ and its downstream transcriptional targets Cyr61 and CTGF. Cancer Res, 71, 2728-38. https://doi.org/10.1158/0008-5472.CAN-10-2711
  18. Law BK (2005). Rapamycin: an anti-cancer immunosuppressant? Crit Rev Oncol Hematol, 56, 47-60. https://doi.org/10.1016/j.critrevonc.2004.09.009
  19. Mahalati K, Kahan BD (2001). Clinical pharmacokinetics of sirolimus. Clin Pharmacokinetics, 40, 573-85. https://doi.org/10.2165/00003088-200140080-00002
  20. Noh WC, Mondesire WH, Peng J, et al (2004). Determinants of rapamycin sensitivity in breast cancer cells. Clin Cancer Res, 10, 1013-23. https://doi.org/10.1158/1078-0432.CCR-03-0043
  21. Rathmell WK, Wright TM, Rini BI (2005). Molecularly targeted therapy in renal cell carcinoma. Expert Rev Anticancer Ther, 5, 1031-40. https://doi.org/10.1586/14737140.5.6.1031
  22. Rizzieri DA, Feldman, E., DiPersio, J. F, et al (2008). A phase 2 clinical trial of deforolimus (AP23573, MK-8669), a novel mammalian target of rapamycin inhibitor, in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res, 14, 2756-62. https://doi.org/10.1158/1078-0432.CCR-07-1372
  23. Sykes SM, Lane SW, Bullinger L, et al (2011). AKT/FOXO signaling enforces reversible differentiation blockade in myeloid leukemias. Cell, 146, 697-708. https://doi.org/10.1016/j.cell.2011.07.032
  24. Soule H, Vazquez J, Long A, Albert S, Brennan M (1973). A human cell line from a pleural effusion derived from a breast carcinoma. J National Cancer Inst, 51, 1409-16.
  25. Thimmaiah KN, Easton JB, Houghton PJ (2010). Protection from rapamycin-induced apoptosis by insulin-like growth factor-I is partially dependent on protein kinase C signaling. Cancer Res, 70, 2000-9. https://doi.org/10.1158/0008-5472.CAN-09-3693
  26. Wang Y, Wang X, Zhao H, Liang B, Du Q (2012). Clusterin confers resistance to TNF-alpha-induced apoptosis in breast cancer cells through NF-kappaB activation and Bcl-2 overexpression. J Chemotherapy, 24, 348-57. https://doi.org/10.1179/1973947812Y.0000000049
  27. Wiedmann MW, Caca K (2005). Molecularly targeted therapy for gastrointestinal cancer. Current Cancer Drug Targets, 5, 171-93. https://doi.org/10.2174/1568009053765771
  28. Yaacob NS, Nasir R, Norazmi MN (2013). Influence of $17{\beta}$-estradiol on 15-deoxy-Δ12, 14 prostaglandin J2-induced apoptosis in MCF-7 and MDA-MD-231 cells. Asian Pac J Cancer Prev, 14, 6761-7. https://doi.org/10.7314/APJCP.2013.14.11.6761
  29. Yu K, Toral-Barza L, Discafani C, et al (2001). mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocrine-Related Cancer, 8, 249-58. https://doi.org/10.1677/erc.0.0080249

Cited by

  1. Morusin suppresses breast cancer cell growth in vitro and in vivo through C/EBPβ and PPARγ mediated lipoapoptosis vol.34, pp.1, 2015, https://doi.org/10.1186/s13046-015-0252-4
  2. The Effect of Rapamycin on Oxidative Stress in MCF-7 and MDA MB-231 Human Breast Cancer Cell Lines vol.11, pp.3, 2016, https://doi.org/10.17795/jjnpp-38177
  3. Allyl-isatin suppresses cell viability, induces cell cycle arrest, and promotes cell apoptosis in hepatocellular carcinoma HepG2 cells vol.30, pp.3, 2016, https://doi.org/10.1111/fcp.12193
  4. Rapamycin enhances the anti-angiogenesis and anti-proliferation ability of YM155 in oral squamous cell carcinoma vol.39, pp.6, 2017, https://doi.org/10.1177/1010428317706213
  5. Monitoring Early Breast Cancer Response to Neoadjuvant Therapy Using H-Scan Ultrasound Imaging: Preliminary Preclinical Results pp.02784297, 2018, https://doi.org/10.1002/jum.14806