DOI QR코드

DOI QR Code

Rapamycin and PF4 Induce Apoptosis by Upregulating Bax and Down-Regulating Survivin in MNU-Induced Breast Cancer

  • Published : 2014.05.15

Abstract

Background: To elucidate the role of rapamycin and PF4 on apoptosis regulation via Bax (pro-apoptosis), Bcl-2 (anti-apoptosis) and survivin activation on the growth in the 1-methyl-1-nitrosourea-induced invasive breast carcinoma model. Materials and Methods: Thirty five female Sprague Dawley rats at age 21-day old were divided into 4 groups; Group 1 (control, n=10), Group 2 (PF4, n=5), Group 3 (rapamycin, n=10) and Group 4 (rapamycin+PF4, n=10). MNU was administered intraperitionally, dosed at 70mg/kg body weight. The rats were treated when the tumors reached the size of $14.5{\pm}0.5mm$ and subsequently sacrificed after 5 days. Rapamycin and PF4 were administered as focal intralesional injections at the dose of $20{\mu}g$/lesion. The tumor tissue was then subjected to histopathological examinations for morphological appraisal and immunohistochemical assessment of the pro-apoptotic marker, Bax and anti-apoptotic markers, Bcl-2 and survivin. Results: The histopathological pattern of the untreated control cohort showed that the severity of the malignancy augments with mammary tumor growth. Tumors developing in untreated groups were more aggressive whilst those in treated groups demonstrated a transformation to a less aggressive subtype. Combined treatment resulted in a significant reduction of tumor size without phenotypic changes. Bax, the pro-apoptotic marker, was significantly expressed at higher levels in the rapamycin-treated and rapamycin+PF4-treated groups compared to controls (p<0.05). Consequently, survivin was also significantly downregulated in the rapamycin-treated and rapamycin+PF4-treated group and this was significantly different when compared to controls (p). Conclusions: In our rat model, it could be clearly shown that rapamycin specifically affects Bax and survivin signaling pathways in activation of apoptosis. We conclude that rapamycin plays a critical role in the induction of apoptosis in MNU-induced mammary carcinoma.

Keywords

References

  1. Asanuma H, Torigoe T, Kamiguchi K, et al (2005). Survivin expression is regulated by coexpression of human epidermal growth factor receptor 2 and epidermal growth factor receptor via phosphatidylinositol 3-kinase/AKT signaling pathway in breast cancer cells. Cancer Res, 65, 11018-25. https://doi.org/10.1158/0008-5472.CAN-05-0491
  2. Ballou LM, Lin RZ (2008). Rapamycin and mTOR kinase inhibitors. J Chem Biol, 1, 27-36. https://doi.org/10.1007/s12154-008-0003-5
  3. Burz C, Berindan-Neagoe I, Balacescu O, Irimie A (2009). Apoptosis in cancer: key molecular signaling pathways and therapy targets. Acta oncologica, 48, 811-21. https://doi.org/10.1080/02841860902974175
  4. GLOBOCAN 2008 (IARC), S. o. C. I. (2008). Section of Cancer Information (29/8/2013)
  5. Gullino P, Pettigrew HM, Grantham F (1975). N-nitrosomethylurea as mammary gland carcinogen in rats. J Natl Cancer Inst, 54, 401-14.
  6. Hahm H, Davidson N (1998). Apoptosis in the mammary gland and breast cancer. Endocrine-related cancer, 5, 199-211. https://doi.org/10.1677/erc.0.0050199
  7. Hassan Z K, Elamin M H, Omer S A, et al (2013). Oleuropein induces apoptosis via the p53 pathway in breast cancer cells. Asian Pac J Cancer Prev, 14, 6739-42. https://doi.org/10.7314/APJCP.2013.14.11.6739
  8. Hengartner M O (2000). The biochemistry of apoptosis. Nature, 407, 770-76. https://doi.org/10.1038/35037710
  9. Huang K, Huang Y, Diao Y (2012). Wogonin induces apoptosis and down-regulates survivin in human breast cancer MCF-7 cells by modulating PI3K-AKT pathway. Int Immunopharmacol, 12, 334-41. https://doi.org/10.1016/j.intimp.2011.12.004
  10. Huynh H (2010). Molecularly targeted therapy in hepatocellular carcinoma. Biochemical Pharmacol, 80, 550-60. https://doi.org/10.1016/j.bcp.2010.03.034
  11. Jaafar H, Abdullah S, Murtey M D, Idris F M (2012). Expression of Bax and Bcl-2 in tumour cells and blood vessels of breast cancer and their association with angiogenesis and hormonal receptors. Asian Pac J Cancer Prev, 13, 3857-62. https://doi.org/10.7314/APJCP.2012.13.8.3857
  12. Jaafar H, Idris F M, Nafi S N M (2009). The association between phenotype and size of breast tumors induced by 1-methyl-1-nitrosourea (MNU) injection in rats. Med Sci Monit, 15.
  13. Jha K, Shukla M, Pandey M (2012). Survivin expression and targeting in breast cancer. Surgical Oncology, 21, 125-31. https://doi.org/10.1016/j.suronc.2011.01.001
  14. Jurgensmeier J M, Xie Z, Deveraux Q, et al (1998). Bax directly induces release of cytochrome c from isolated mitochondria. Proc Natl Acad Sci USA, 95, 4997-5002. https://doi.org/10.1073/pnas.95.9.4997
  15. Lu J, Tan M, Huang WC, et al (2009). Mitotic deregulation by survivin in ErbB2-overexpressing breast cancer cells contributes to Taxol resistance. Clin Cancer Res, 15, 1326-34. https://doi.org/10.1158/1078-0432.CCR-08-0954
  16. Maione T E, Gray G S, Hunt A J, Sharpe R J (1991). Inhibition of tumor growth in mice by an analogue of platelet factor 4 that lacks affinity for heparin and retains potent angiostatic activity. Cancer Res, 51, 2077-83.
  17. Namba R, Young L J, Abbey C K, et al (2006). Rapamycin inhibits growth of premalignant and malignant mammary lesions in a mouse model of ductal carcinoma in situ. Clinical Cancer Res, 12, 2613-21. https://doi.org/10.1158/1078-0432.CCR-05-2170
  18. Noh WC, Mondesire W H, Peng J, et al (2004). Determinants of rapamycin sensitivity in breast cancer cells. Clinical Cancer Res, 10, 1013-23. https://doi.org/10.1158/1078-0432.CCR-03-0043
  19. Purcell J W, Davis J, Reddy M, et al (2010). Activity of the kinesin spindle protein inhibitor ispinesib (SB-715992) in models of breast cancer. Clinical Cancer Res, 16, 566-76. https://doi.org/10.1158/1078-0432.CCR-09-1498
  20. Reed J (1996). Balancing cell life and death: bax, apoptosis, and breast cancer. J Clin Invest, 97, 2403. https://doi.org/10.1172/JCI118684
  21. Russo J, Russo I H (1995). The etiopathogenesis of breast cancer prevention. Cancer letters, 90, 81-9. https://doi.org/10.1016/0304-3835(94)03681-8
  22. Russo J, Russo I H (2000). Atlas and histologic classification of tumors of the rat mammary gland. J Mammary Gland Biol Neoplasia, 5, 187-200. https://doi.org/10.1023/A:1026443305758
  23. Samkari A, Cooper Z A, Holloway M P, Liu J, Altura R A (2012). Rapamycin induces the anti-apoptotic protein survivin in neuroblastoma. Int J Biochem Mol Biol, 3, 28.
  24. Shirouzu Y, Ryschich E, Salnikova O, et al (2010). Rapamycin inhibits proliferation and migration of hepatoma cells. J Surg Res, 159, 705-13. https://doi.org/10.1016/j.jss.2008.07.035
  25. Shor B, Gibbons J J, Abraham R T, Yu K (2009). Targeting mTOR globally in cancer: thinking beyond rapamycin. Cell Cycle, 8, 3831-7. https://doi.org/10.4161/cc.8.23.10070
  26. Signore M, Ricci-Vitiani L, De Maria R (2013). Targeting apoptosis pathways in cancer stem cells. Cancer letters, 332, 374-82. https://doi.org/10.1016/j.canlet.2011.01.013
  27. Tan M L, Ooi J P, Ismail N, Moad A I H, Muhammad T S T (2009). Programmed cell death pathways and current antitumor targets. Pharmaceutical Res, 26, 1547-60. https://doi.org/10.1007/s11095-009-9895-1
  28. Tanaka K, Iwamoto S, Gon G, et al (2000). Expression of survivin and its relationship to loss of apoptosis in breast carcinomas. Clinical Cancer Res, 6, 127-34.
  29. Thompson H J, McGinley J N, Rothhammer K, Singh M (1995). Rapid induction of mammary intraductal proliferations, ductal carcinoma in situ and carcinomas by the injection of sexually immature female rats with 1-methyl-1-nitrosourea. Carcinogenesis, 16, 2407-12. https://doi.org/10.1093/carcin/16.10.2407
  30. Verdecia M A, Huang Hk, Dutil E, et al (2000). Structure of the human anti-apoptotic protein survivin reveals a dimeric arrangement. Nature Structural Molec Biol, 7, 602-8. https://doi.org/10.1038/76838
  31. Wan L Z, Hu N, Liu XJ, et al (2014). Fangchinoline Inhibits cell proliferation via Akt/GSK-3beta/cyclin D1 signaling and induces apoptosis in MDA-MB-231 breast cancer cells. Asian Pac J Cancer Prev, 15, 769-73. https://doi.org/10.7314/APJCP.2014.15.2.769
  32. Yip C H, Taib N A, Mohamed I (2006). Epidemiology of breast cancer in Malaysia. Asian Pac J Cancer Prev, 7, 369-74.
  33. Zhou J, Luo YH, Wang JR, Lu BB, Tian Y (2013). Gambogenic acid induction of apoptosis in a breast cancer cell line. Asian Pac J Cancer Prev, 14, 7601-05. https://doi.org/10.7314/APJCP.2013.14.12.7601
  34. Zhu N, Gu L, Findley H W, Li F, Zhou M (2004). An alternatively spliced survivin variant is positively regulated by p53 and sensitizes leukemia cells to chemotherapy. Oncogene, 23, 7545-51. https://doi.org/10.1038/sj.onc.1208038

Cited by

  1. Correlation of Tumour Response with Starting Tumour Size and Dose of Tamoxifen in an N-Methyl-N-Nitrosourea (NMU)-Induced Rat Mammary Cancer Model vol.15, pp.16, 2014, https://doi.org/10.7314/APJCP.2014.15.16.6721
  2. Lack of Relation of Survivin Gene Expression with Survival and Surgical Prognostic Factors in Endometrial Carcinoma Patients vol.15, pp.16, 2014, https://doi.org/10.7314/APJCP.2014.15.16.6905
  3. Upregulation of HIF-1α by Hypoxia Protect Neuroblastoma Cells from Apoptosis by Promoting Survivin Expression vol.15, pp.19, 2014, https://doi.org/10.7314/APJCP.2014.15.19.8251
  4. Effects of Rapamycin on Cell Apoptosis in MCF-7 Human Breast Cancer Cells vol.15, pp.24, 2014, https://doi.org/10.7314/APJCP.2014.15.24.10659