Inhibitory Effect of Snake Venom on Colon Cancer Cell Growth Through Induction of Death Receptor Dependent Apoptosis

사독(蛇毒)이 세포자멸사와 관계있는 Death Receptor를 통한 인간 대장암 세포 성장억제에 미치는 영향

  • Oh, Myung-Jin (Dept. of Acupuncture & Moxibustion, College of Oriental Medicine, Kyungwon University) ;
  • Song, Ho-Sueb (Dept. of Acupuncture & Moxibustion, College of Oriental Medicine, Kyungwon University)
  • 오명진 (경원대학교 한의과대학 침구학교실) ;
  • 송호섭 (경원대학교 한의과대학 침구학교실)
  • Received : 2012.01.17
  • Accepted : 2012.01.26
  • Published : 2012.02.20

Abstract

목적 : 이 연구는 $Vipera$ $lebetina$ $turanica$ 사독(蛇毒)이 인간 대장암 세포주인 HCT116 세포에서 세포주기진행, death receptor 의존적 세포자멸사 경로 관련단백질 발현 및 NK-${\kappa}B$와 STAT3 활성에 미치는 영향을 규명함으로써 대장암 세포 성장에 대한 억제와 그 기전에 대하여 살펴보고자 하였다. 방법 : 사독을 처리한 후 HCT116의 세포주기를 분석하기 위해서 FACS analysis를 시행하였고, apoptosis 평가에는 TUNEL assay를 시행하였으며 death receptor 의존적 세포자멸사 경로 관련단백질 및 NF-${\kappa}B$와 STAT3 활성 변동 관찰에는 RT-PCR 및 western blot analysis를 시행하였다. 결과 : 1. 0.1, 0.5 및 $1{\mu}g/m{\ell}$ 등의 사독을 처리한 결과 농도 의존적으로 HCT116 대장암 세포활성의 억제가 나타났다. 2. 0.1, 0.5 및 $1{\mu}g/m{\ell}$ 등의 사독을 처리한 결과 농도의존적으로 세포자멸사 활성세포의 증가가 나타났고, SVT $1{\mu}g/m{\ell}$에서는 60-70%의 대장암세포 억제 효과가 나타났다. 3. 0.1, 0.5 및 $1{\mu}g/m{\ell}$ 등의 사독을 처리한 결과 약한 G1 arrest와 강한 G2/M arrest가 나타났고, G0/G1 또는 G2/M 관련 cyclin D, E 및 B1의 증가가 나타났다. 4. 0.1, 0.5 및 $1{\mu}g/m{\ell}$ 등의 사독을 처리한 결과 death receptor4, 5의 발현증가와 그에 따른 세포자멸사 촉진 Bax, PARP, caspase-3, -8, -9 발현 증가 및 세포자멸사 억제의 Bcl-2의 발현 감소 등이 나타났다. 6. 0.1, 0.5 및 $1{\mu}g/m{\ell}$ 등의 사독을 처리한 결과 NF-${\kappa}B$와 STAT3의 활성변동은 관찰되지 않았다. 결론 : 이상의 연구에서 사독은 death receptor 의존적인 세포자멸사를 촉진하여 대장암의 화학치료 내성을 극복할 수 있는 하나의 대안이 될 것으로 생각되지만 보다 심화된 연구가 필요할 것으로 사료된다.

Keywords

References

  1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics. CA Cancer J Clin. 2009 ; 59(4) : 225-49. https://doi.org/10.3322/caac.20006
  2. Danaei G, Van der Hoorn S, Lopez AD, Murray CJL, Ezzati M. The comparative risk assessment collaborating group(cancers), causes of cancer in the world: Comparative risk assessment of nine behavioural and environmental risk factors. Lancet. 2005 ; 366(9499) : 1784-93. https://doi.org/10.1016/S0140-6736(05)67725-2
  3. Scheele J, Altendorf-Hofmann A. Resection of colorectal liver metastases. Langenbeck's Arch Surg. 1999 ; 384(4) : 313-27. https://doi.org/10.1007/s004230050209
  4. Goldberg RM, Sargent DJ, Morton RF, Fuchs CS, Ramanathan RK, Williamson SK, Findlay BP, Pitot HC, Alberts SR. A randomized controlled trial of fluorouracil plus leucovorin, irinotecan and oxaliplatin combinations in patients with previously untreated metastatic colorectal cancer. J Clin Oncol. 2004 ; 22(1) : 23-30. https://doi.org/10.1200/JCO.2004.09.046
  5. Kamb A, Wee S, Lengauer C. Why is cancer drug discovery so difficult? Nature Rev Drug Discovery. 2007 ; 6(2) : 115-20. https://doi.org/10.1038/nrd2155
  6. Dalerba P, Cho RW, Clarke MF. Cancer Stem Cells: Models and Concepts. Annual Rev Med. 2007 ; 58 : 267-84. https://doi.org/10.1146/annurev.med.58.062105.204854
  7. Strasser A, O'Connor L, Dixit VM. Apoptosis signaling. Ann Rev Biochem. 2001 ; 69 : 217-45.
  8. Salvase GS, Abrams JM. Caspase activation— stepping on the gas or releasing the brakes Lessons from humans and flies. Oncogene. 2004 ; 23(16) : 2774-84. https://doi.org/10.1038/sj.onc.1207522
  9. Debatin KM, Krammer PH. Death receptors in chemotherapy and cancer. Oncogene. 2004 ; 23(16) : 2950-66. https://doi.org/10.1038/sj.onc.1207558
  10. Balan KV, Demetzos C, Prince J, Dimas K, Cladaras M, Han Z, Wyche JH, Pantazis P. Induction of apoptosis in human colon cancer HCT116 cells treated with an extract of the plant product, Chios mastic gum. In Vivo. 2005 ; 19(1) : 93-102.
  11. Balan KV, Prince J, Han Z, Dimas K, Cladaras M, Wyche JH, Sitaras NM, Pantazis P. Antiproliferative activity and induction of apoptosis in human colon cancer cells treated in vitro with constituents of a product derived from Pistacia lentiscus L. var. chia. Phytomedicine. 2007 ; 14(4) : 263-72. https://doi.org/10.1016/j.phymed.2006.03.009
  12. Efimova EV, Liang H, Pitroda SP, Labay E, Darga TE, Levina V, Lokshin A, Roizman B, Weichselbaum RR, Khodarev NN. Radioresistance of Stat1 overexpressing tumour cells is associated with suppressed apoptotic response to cytotoxic agents and increased IL6-IL8 signalling. Int J Radiat Biol. 2009 ; 85(5) : 421-31. https://doi.org/10.1080/09553000902838566
  13. Maduro JH, Noordhuis MG, ten Hoor KA, Pras E, Arts HJ, Eijsink JJ, Hollema H, Mom CH, de Jong S, de Vries EG, de Bock GH, van der Zee AG. The prognostic value of TRAIL and its death receptors in cervical cancer. Int J Radiat Oncol Biol Phys. 2009 ; 75(1) : 203-11. https://doi.org/10.1016/j.ijrobp.2009.03.071
  14. Siigur E, Aaspollu A, Siigur J. Sequence diversity of Vipera lebetina SVT gland serine proteinase homologs-result of alternativesplicing or genome alteration. Gene. 2001 ; 263 : 199-203. https://doi.org/10.1016/S0378-1119(00)00571-0
  15. Alves RM, Antonucci GA, Paiva HH, Cintra AC, Franco JJ, Mendonca-Franqueiro EP, Dorta DJ, Giglio JR, Rosa JC, Fuly AL, Dias-Baruffi M, Soares AM, Sampaio SV. Evidence of caspase-mediated apoptosis induced by l-amino acid oxidase isolated from Bothrops atrox snake venom. Comp Biochem Physiol A Mol Integr Physiol. 2008 ; 151(4) : 542-50. https://doi.org/10.1016/j.cbpa.2008.07.007
  16. Park MH, Son DJ, Kwak DH, Song HS, Oh KW, Yoo HS, Lee YM, Song MJ, Hong JT. Snake venom toxin Inhibits Cell Growth through Induction of Apoptosis in Neuroblastoma Cells. Arch Pharm Res. 2009 ; 32(11) : 1545-54, https://doi.org/10.1007/s12272-009-2106-0
  17. Son DJ, Park MH, Chae SJ, Moon SO, Lee JW, Song HS, Moon DC, Kang SS, Kwon YE, Hong JT. Inhibitory effect of SVT toxin from Vipera lebetina turanica on hormone-refractory human prostate cancer cell growth: induction of apoptosis through inactivation of nuclear factor kappaB. Mol Cancer Ther. 2007 ; 6(2) : 675-83.
  18. O'Donovan TR, O'Sullivan GC, McKenna SL. Induction of autophagy by drugresistant esophageal cancer cells promotes their survival and recovery following treatment with chemotherapeutics. Autophagy. 2011 ; 7(5) : 509-24. https://doi.org/10.4161/auto.7.5.15066
  19. Kang YJ, Kim IY, Kim EH, Yoon MJ, Kim SU, Kwon TK, Choi KS. Paxilline enhances TRAIL-mediated apoptosis of glioma cells via modulation of c-FLIP, survivin and DR5. Exp Mol Med. 2011 ; 43(1) : 24-34. https://doi.org/10.3858/emm.2011.43.1.003
  20. Li J, Yu W, Tiwary R, Park SK, Xiong A, Sanders BG, Kline K. ${\alpha}$-TEA-induced death receptor dependent apoptosis involves activation of acid sphingomyelinase and elevated ceramideenriched cell surface membranes. Cancer Cell Int. 2010 ; 10 : 40. https://doi.org/10.1186/1475-2867-10-40
  21. Zhu H, Liu XW, Ding WJ, Xu DQ, Zhao YC, Lu W, He QJ, Yang B. Up-regulation of death receptor 4 and 5 by celastrol enhances the anti-cancer activity of TRAIL/Apo-2L. Cancer Lett. 2010 ; 297(2) : 155-64. https://doi.org/10.1016/j.canlet.2010.04.030
  22. Kim EJ, Park SY, Lee JY, Park JH. Fucoidan present in brown algae induces apoptosis of human colon cancer cells. BMC Gastroenterol. 2010 ; 10 : 96. https://doi.org/10.1186/1471-230X-10-96
  23. Tang Y, Li X, Liu Z, Simoneau AR, Xie J, Zi X. Flavokawain B, a kava chalcone, induces apoptosis via up-regulation of death-receptor 5 and Bimexpression in androgen receptor negative, hormonal refractory prostate cancer cell lines and reduces tumor growth. Int J Cancer. 2010 ; 127(8) : 1758-68. https://doi.org/10.1002/ijc.25210
  24. Sun SY. Understanding the role of the death receptor 5/FADD/caspase-8 death signaling in cancer metastasis. Mol Cell Pharmacol. 2011 ; 3(1) : 31-4.
  25. Elrod HA, Sun SY. Modulation of death receptors by cancer therapeutic agents. Cancer Biol Ther. 2008 ; 7(2) : 163-73. https://doi.org/10.4161/cbt.7.2.5335
  26. Grana X, Reddy EP. Cell cycle control in mammalian cells: role of cyclins, cyclin dependent kinases(CDKs), growth suppressor genes and cyclin-dependent kinase inhibitors (CKIs). Oncogene. 1995 ; 11(2) : 211-9.
  27. Morgan DO. Effects of pulmonary gas embolism on circulation and respiration in the dog. VI. Influence of body position on the effects of pulmonary gas embolism. Nature. 1995 ; 374 : 131-4. https://doi.org/10.1038/374131a0
  28. Stewart ZA, Westfall MD, Pietenpol JA. Cellcycle dysregulation and anticancer therapy. Trends Pharmacol Sci. 2003 ; 24(3) : 139-45. https://doi.org/10.1016/S0165-6147(03)00026-9
  29. Zhao J, Dynlacht B, Imai T, Hori T, Harlow E. Expression of NPAT, a novel substrate of cyclin E-CDK2, promotes S-phase entry. Genes Dev. 1998 ; 12(4) : 456-61. https://doi.org/10.1101/gad.12.4.456
  30. Senderowicz AM, Sausville EA. Preclinical and clinical development of cyclin-dependent kinase modulators. J Natl Cancer Inst. 2000 ; 92(5) : 376-87. https://doi.org/10.1093/jnci/92.5.376
  31. Jackson JR, Gilmartin A, Imburgia C, Winkler JD, Marshall LA, Roshak A. An indolocarbazole inhibitor of human checkpoint kinase (Chk1) abrogates cell-cycle arrest caused by DNA damage. Cancer Res. 2000 ; 60(3) : 566-72.
  32. Hirose Y, Berger MS, Pieper RO. Abrogation of the Chk1 mediated G2 checkpoint pathway potentiates Temozolomide-induced toxicity in a p53-independent manner in human glioblastoma cells. Cancer Res. 2001 ; 61(15) : 5843-9.
  33. Sausville EA, Arbuck SG, Messmann R, Headlee D, Bauer KS, Lush RM, Murgo A, Figg WD, Lahusen T, Jaken S, Jing X, Roberge M, Fuse E, Kuwabara T, Senderowicz AM. Phase I trial of 72 hour continuous infusion UCN-01 in patients with refractory neoplasms. J Clin Oncol. 2001 ; 19(8) : 2319-33. https://doi.org/10.1200/JCO.2001.19.8.2319
  34. Carlson B, Lahusen T, Singh S, Loaiza-Perez A, Worland PJ, Pestell R, Albanese C, Sausville EA, Senderowicz AM. Down-regulation of cyclin D1 by transcriptional repression in MCF-7 human breast cancer cells induced by flavopiridol. Cancer Res. 1999 ; 59(18) : 4634-41.
  35. Bible KC, Kaufmann SH. Cytotoxic synergy between flavopiridol(NSC 649890, L86-8275) and various antineoplastic agents: the importance of sequence of administration. Cancer Res. 1997 ; 57(16) : 3375-80.
  36. Shapiro GI, Supko JG, Patterson A, Lynch C, Lucca J, Zacarola PF, Muzikansky A, Wright JJ, Lynch TJ Jr, Rollins BJ. A Phase II trial of the cyclin-dependent kinase inhibitor flavopiridol in patients with previously untreated stage IV non-small cell lung cancer. Clin Cancer Res. 2001 ; 7(6) : 1590-9.
  37. Tyagi AK, Singh RP, Agarwal C, Chan DC, Agarwal R. Silibinin strongly synergizes human prostate cancer DU145 cells to doxorubicininduced growth inhibition, G2-M arrest, and apoptosis. Clin Cancer Res. 2002 ; 8(11) : 3512-9.
  38. Sauter ER, Yeo UC, von Stemm A, Zhu W, Litwin S, Tichansky DS, Pistritto G, Nesbit M, Pinkel D, Herlyn M, Bastian BC. Cyclin D1 is a candidate oncogene in cutaneous melanoma. Cancer Res. 2002 ; 62(11) : 3200-6.
  39. Shan XL, Zhou XY, Yang J, Wang YL, Deng YH, Zhang MX. Inhibitory effect and mechanism of cucurbitacin E on the proliferation of ovarian cancer cells and its mechanism. Chin J Cancer. 2010 ; 29(1) : 20-4. https://doi.org/10.5732/cjc.009.10223
  40. Colomiere M, Ward AC, Riley C, Trenerry MK, Cameron-Smith D, Findlay J, Ackland L, Ahmed N. Cross talk of signals between EGFR and IL-6R through JAK2/STAT3 mediate epithelial-mesenchymal transition in ovarian carcinomas. Br J Cancer. 2009 ; 100(1) : 134-44. https://doi.org/10.1038/sj.bjc.6604794
  41. Sun M, Liu C, Nadiminty N, Lou W, Zhu Y, Yang J, Evans CP, Zhou Q, Gao AC. Inhibition of Stat3 activation by sanguinarine suppresses prostate cancer cell growth and invasion. Prostate. 2011 ; 72(1) : 82-9.
  42. Park MH, Choi MS, Kwak DH, Oh KW, Yoon DY, Han SB, Song HS, Song MJ, Hong JT. Anti-cancer effect of bee venom in prostate cancer cells through activation of caspase pathway via inactivation of NF-${\kappa}B$. Prostate. 2010 ; 61 : 801-12.
  43. Saydmohammed M, Joseph D, Syed V. Curcumin suppresses constitutive activation of STAT-3 by up-regulating protein inhibitor of activated STAT-3(PIAS-3) in ovarian and endometrial cancer cells. J Cell Biochem. 2010 ; 110(2) : 447-56.