• 검색 결과가 없습니다.

Fusion between cervical cancer cells, HeLa cells, led to vast cell death, even though parental cells had low levels of p53. In the process, the fused cells showed mitotic defect such as formation of spindle multipolarity due to increased centrosomes, resulting in asymmetric division. In addition, when the fate of daughter cells was traced, multinucleated cells showed tendency to die. However, surviving fused cells, were characterized by upregulation of survivin, reflecting increased survivin protein stability. Moreover, in fused cells, survivin became preferentially localized to the cytosol, where it is known to exert its anti-apoptotic function. Knockdown of survivin decreased survival to a greater extent in fused cells than in unfused cells, suggesting that fused cells became more dependent on survivin. Therefore, above findings indicate that, after cancer cell fusion, a subpopulation of fused cells with a higher level of cytosolic survivin are able to avoid apoptotic crisis and survive to proliferate continuously, a process that might contribute to human cancer progression. Fused cells occuring depolyploidization had similar amounts of DNA and proliferated but showed genetic instability, which is far superior to cell migration or chemoresistance, which could have detrimental effects on cancer therapy. This suggests that fused cells made from isotypic parental cells may lead to more severe tumor heterogeneity, a difficult problem in cancer treatment, and that cell fusion may result in genome diversity.

49

REFFERENCE

1. Andreassen PR, Lohez OD, Lacroix FB, Margolis RL (2001) Tetraploid State Induces p53-dependent Arrest of Nontransformed Mammalian Cells in G1. Molecular Biology of the Cell 12:1315-1328

2. Aylon Y, Oren M (2011) p53: guardian of ploidy. Molecular oncology 5:315-323

3. Bai H, Ge S, Lu J, Qian G, Xu R (2013) Hypoxia inducible factor-1alpha-mediated activation of survivin in cervical cancer cells. J Obstet Gynaecol Res 39:555-563 4. Bayani J, Squire JA (2005) Comparative genomic hybridization. Current protocols in cell

biology Chapter 22:Unit 22.26

5. Behera B, Mishra D, Roy B, Devi KS, Narayan R, Das J, Ghosh SK, Maiti TK (2014) Abrus precatorius agglutinin-derived peptides induce ROS-dependent mitochondrial apoptosis through JNK and Akt/P38/P53 pathways in HeLa cells. Chem Biol Interact 222:97-105

6. Castedo M, Coquelle A, Vitale I, Vivet S, Mouhamad S, Viaud S, Zitvogel L, Kroemer G (2006) Selective resistance of tetraploid cancer cells against DNA damage-induced apoptosis. Annals of the New York Academy of Sciences 1090:35-49

7. Chen X, Duan N, Zhang C, Zhang W (2016) Survivin and Tumorigenesis: Molecular Mechanisms and Therapeutic Strategies. Journal of Cancer 7:314-323

8. Cheung CH, Huang CC, Tsai FY, Lee JY, Cheng SM, Chang YC, Huang YC, Chen SH, Chang JY (2013) Survivin - biology and potential as a therapeutic target in oncology.

OncoTargets and therapy 6:1453-1462

9. Duelli DM, Hearn S, Myers MP, Lazebnik Y (2005) A primate virus generates transformed human cells by fusion. J Cell Biol 171:493-503

10. Foley EA, Kapoor TM (2013) Microtubule attachment and spindle assembly checkpoint signalling at the kinetochore. Nature reviews Molecular cell biology 14:25-37

11. Fortugno P, Beltrami E, Plescia J, Fontana J, Pradhan D, Marchisio PC, Sessa WC, Altieri DC (2003) Regulation of survivin function by Hsp90. Proceedings of the National

50

Academy of Sciences of the United States of America 100:13791-13796

12. Fukuta K, Kohri K, Fukuda H, Watanabe M, Sugimura T, Nakagama H (2008) Induction of multinucleated cells and apoptosis in the PC-3 prostate cancer cell line by low concentrations of polyethylene glycol 1000. Cancer science 99:1055-1062

13. Ganem NJ, Pellman D (2007) Limiting the proliferation of polyploid cells. Cell 131:437-440

14. Ganem NJ, Storchova Z, Pellman D (2007) Tetraploidy, aneuploidy and cancer. Current opinion in genetics & development 17:157-162

15. Guo XX, Li Y, Sun C, Jiang D, Lin YJ, Jin FX, Lee SK, Jin YH (2014) p53-dependent Fas expression is critical for Ginsenoside Rh2 triggered caspase-8 activation in HeLa cells. Protein Cell 5:224-234

16. Halliwell B (2007) Biochemistry of oxidative stress. Biochemical Society transactions 35:1147-1150

17. Ho CC, Hau PM, Marxer M, Poon RY (2010) The requirement of p53 for maintaining chromosomal stability during tetraploidization. Oncotarget 1:583-595

18. Horn GP, Vongpunsawad S, Kornmann E, Fritz B, Dittmer DP, Cattaneo R, Dobbelstein M (2005) Enhanced cytotoxicity without internuclear spread of adenovirus upon cell fusion by measles virus glycoproteins. J Virol 79:1911-1917

19. Kerbel RS, Lagarde AE, Dennis JW, Donaghue TP (1983) Spontaneous fusion in vivo between normal host and tumor cells: possible contribution to tumor progression and metastasis studied with a lectin-resistant mutant tumor. Mol Cell Biol 3:523-538

20. Khodjakov A, Pines J (2010) Centromere tension: a divisive issue. Nature cell biology 12:919-923

21. Levine DS, Sanchez CA, Rabinovitch PS, Reid BJ (1991) Formation of the tetraploid intermediate is associated with the development of cells with more than four centrioles in the elastase-simian virus 40 tumor antigen transgenic mouse model of pancreatic cancer.

Proceedings of the National Academy of Sciences of the United States of America 88:6427-6431

22. Li F, Yang J, Ramnath N, Javle MM, Tan D (2005) Nuclear or cytoplasmic expression of survivin: what is the significance? Int J Cancer 114:509-512

23. Lu X, Kang Y (2009) Cell fusion as a hidden force in tumor progression. Cancer Res

51 69:8536-8539

24. Mortensen K, Lichtenberg J, Thomsen PD, Larsson LI (2004) Spontaneous fusion between cancer cells and endothelial cells. Cell Mol Life Sci 61:2125-2131

25. Nam HJ, Lee IJ, Jang S, Bae CD, Kwak SJ, Lee JH (2014) p90 ribosomal S6 kinase 1 (RSK1) isoenzyme specifically regulates cytokinesis progression. Cell Signal 26:208-219 26. Nigg EA, Stearns T (2011) The centrosome cycle: Centriole biogenesis, duplication and

inherent asymmetries. Nature cell biology 13:1154-1160

27. Scheffner M, Werness BA, Huibregtse JM, Levine AJ, Howley PM (1990) The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of p53. Cell 63:1129-1136

28. Vitale I, Senovilla L, Jemaa M, Michaud M, Galluzzi L, Kepp O, Nanty L, Criollo A, Rello-Varona S, Manic G, Metivier D, Vivet S, Tajeddine N, Joza N, Valent A, Castedo M, Kroemer G (2010) Multipolar mitosis of tetraploid cells: inhibition by p53 and dependency on Mos. The EMBO journal 29:1272-1284

29. Vogel C, Kienitz A, Hofmann I, Muller R, Bastians H (2004) Crosstalk of the mitotic spindle assembly checkpoint with p53 to prevent polyploidy. Oncogene 23:6845-6853 30. Vousden KH, Lu X (2002) Live or let die: the cell's response to p53. Nature reviews

Cancer 2:594-604

31. Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V, Ross AJ, Roth KA, MacGregor GR, Thompson CB, Korsmeyer SJ (2001) Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science 292:727-730

32. Yu J, Wang Z, Kinzler KW, Vogelstein B, Zhang L (2003) PUMA mediates the apoptotic response to p53 in colorectal cancer cells. Proceedings of the National Academy of Sciences of the United States of America 100:1931-1936

33. Zhao J, Tenev T, Martins LM, Downward J, Lemoine NR (2000) The ubiquitin-proteasome pathway regulates survivin degradation in a cell cycle-dependent manner.

Journal of cell science 113 Pt 23:4363-4371

52

53

증식을 보이는 세포주에서도 염색체 불안정성이 유도된 것을 알 수 있었다. 특히, 세포사멸에서 살아남은 세포군은 안정적으로 증식을 한다는 것이다. 융합된 세포군 에서 항 세포사멸 기능을 갖는 survivin 단백질의 안정성 증가와 세포질에 존재하는

survivin의 증가를 알 수 있었다. survivin의 결핍은 비 융합세포와 비교 시 융합 된 세포의 생존율 감소를 초래하는 것으로 보아 survivin에 더 의존적임을 알 수 있었 다. 따라서 위의 결과는 암세포간의 융합 후에 세포질에 존재하는 survivin이 더 높 은 수준으로 증가한 융합 세포군이 세포사멸의 위기를 피하여 지속적으로 증식하므 로 암 진행에 기여할 수 있음을 시사한다. 암 진행과 관련하여 융합 세포는 비 융 합 세포보다 전체적인 세포 이동 능력이 뛰어 났으며 각 세포주에 따라 세포 이동 능이 다양하게 나타났다. 또한, 저농도의 cisplatin을 가한 경우, 융합 세포는 융합되 지 않은 세포보다 저항성이 높았다. 암세포의 융합은 p53 의존성 세포 사멸을 유도 하는 염색체 불안정성을 초래한다. 그러나 암세포간의 융합에서 세포질 내 survivin 이 더 증가된 세포군이 생존에 유리하고 유전적 다양성을 갖는 것을 알 수 있었고 이러한 유전적 다양성이 아마도 항암치료의 저항성 및 이동 능력의 획득과 같은 더 진보된 특성을 갖게 함을 알 수 있었다.

세포 융합, 세포 사멸, survivin, 비대칭적 분열, 유전체 불안정성

관련 문서