DOI QR코드

DOI QR Code

Growth Inhibition of Human Hepatoma and Bladder Carcinoma Cells by DNA Topoisomerae Inhibitor β-lapachone

DNA topoisomerase 억제제인 β-lapachone에 의한 인체 간암 및 방광암세포 증식억제에 관한 연구

  • Choi Da Yean (R&E Program, Busan Science Academy) ;
  • Lee Jae Il (R&E Program, Busan Science Academy) ;
  • Chung Hyun Sup (R&E Program, Busan Science Academy) ;
  • Seo Han Gyeol (R&E Program, Busan Science Academy) ;
  • Woo Hyun Joo (Department of Biochemistry, Dongeui University College of Oriental Medicine Busan) ;
  • Choi Yung Hyun (Department of Biochemistry, Dongeui University College of Oriental Medicine Busan)
  • 최다연 (부산과학고등학교) ;
  • 이재일 (부산과학고등학교) ;
  • 정협섭 (부산과학고등학교) ;
  • 서한결 (부산과학고등학교) ;
  • 우현주 (동의대학교 한의과대학 생화학교실) ;
  • 최영현 (동의대학교 한의과대학 생화학교실)
  • Published : 2005.06.01

Abstract

The objective of the present study was to investigate the effect of $\beta-lapachone$, a quinone obtained from the bark of the lapacho tree (Tabebuia avellanedae) in South America, on the cell growth of human hepatoma (HepG2) and bladder (T24) carcinoma cells. Exposure of cancer cells to $\beta-lapachone$ resulted in growth inhibition, morphological changes and apoptosis in a concentration-dependent manner, which could be proved by MTT assay and flow cytometry analysis. Reverse transcription-polymerase chain reaction (RT-PCR) and Western blot analyses revealed that $\beta-lapachone$ did not affect the levels of tumor suppressor p53 and cyclin-dependent kinase (Cdk) inhibitor p21 (WAFl/CIPl) expression. However, the transcriptional factor Sp-l and proliferating cell nuclear antigen (PCNA) protein levels were significantly down-regulated by $\beta-lapachone$ in both cell lines. Moreover, $\beta-lapachone$ treatment caused a dose-dependent inhibition of the expression of telomere regulatory gene products such as human telomere reverse transcriptase (hTERT) and telomerase-associated protein-l (TEP-l). Taken together, these findings suggest that $\beta-lapachone$-induced inhibition of human hepatoma and bladder carcinoma cell proliferation is associated with the induction of apoptotic cell death via modulation of several major growth regulatory gene products, and provide important new insights into the additional mechanisms of the anti-cancer activity of $\beta-lapachone$.

남미지역에서 자생하는 Tabebuia avellanedae라는 나무의 수피에서 동정된 quinone계 물질이며, DNA topoisomeras억제제로 알려진 $\beta-lapachone$의 항암작용에 관한 부가적인 자료를 얻기 위하여 인체 간암(HepG2) 및 방광암(T24)세포를 대상으로 조사한 결과 다음과 같은 결과를 얻게 되었다. MTT assay 및 flow cytometry 분석 등의 결과에서, $\beta-lapachone$의 처리에 따라 조사된 두 가지 암세포에서 $\beta-lapachone$처리 농도의존적으로 암세포의 심한 형태적 변형이 동반되면서 암세포의 증식이 억제되었으며, 생존율이 저하되었고 이는 apoptosis유발과 상관성이 있음을 알 수 있었다. $\beta-lapachone$처리에 의한 두 암세포의 증식억제는 종양억제 유전자 p53 및 Cdk inhibitor p21의 발현과는 큰 연관성이 없음을 RT-PCR 및 Western blot analysis를 통하여 확인하였다. 그러나 전사조절인자 Sp-1 및 세포증식 주요조절인자인 PCNA의 단백질 발현은 $\beta-lapachone$처리에 따라 매우 감소되었으며, telomere조절에 중요한 인자들의 선택적 발현 저하 현상도 관찰되었다. 이상의 결과들은 인체 암세포에서 $\beta-lapachone$의 항암작용을 이해하는 중요한 자료가 될 것이며, $\beta-lapachone$과 유사한 화학적 구조 및 성질을 가지는 항암제 후보물질들의 항암기전 비교 및 항암제 개발을 위한 기초 자료로서 응용될 것이다.

Keywords

References

  1. Bailly, C. 2000. Topoisomerase I poisons and suppressors as anticancer drugs. Curr. Med. Chem. 7, 39-58 https://doi.org/10.2174/0929867003375489
  2. Bakshi, R. P. and T. A. Shapiro. 2003. DNA topoisomerases as targets for antiprotozoal therapy. Mini Rev. Med. Chem. 3, 597-608 https://doi.org/10.2174/1389557033487863
  3. Bellamy, W. T. 1992. Prediction of response to drug therapy of cancer. A review of in vitro assays. Drugs 44, 690-708 https://doi.org/10.2165/00003495-199244050-00002
  4. Biggs, J. R., J. E. Kudlow and A. S. Kraft. 1996. The role of the transcription factor Sp1 in regulating the expression of the WAF1/ CIP1 gene in U937 leukemic cells. J. BioI. Chem. 271, 901-906 https://doi.org/10.1074/jbc.271.2.901
  5. Capranico, G., G. Zagotto and M. Palumbo. 2004. Development of DNA topoisomerase-related therapeutics: a short perspective of new challenges. Curr. Med. Chem. Anti-Cane. Agents 4, 335-345 https://doi.org/10.2174/1568011043352885
  6. Chiarugi, V., L. Magnelli and G. Basi. 1994. Apoptosis and the cell cycle. Cell. Mol. Biol. Res. 40, 603-612
  7. Choi, Y. H., W. H. Lee, K. Y. Park and L. Zhang. 2000. p53-independent induction of p21 (WAF1/CIP1), reduction of cyclin B1 and G2/M arrest by the isoflavone genistein in human prostate carcinoma cells. Jpn. J. Cancer Res. 91, 164-173 https://doi.org/10.1111/j.1349-7006.2000.tb00928.x
  8. Choi, Y. H., H. S. Kang and M. A. Yoo. 2003. Suppression of human prostate cancer cell growth by $\beta$-lapachone via down-regulation of pRB phosphorylation and induction of Cdk inhibitor p21(WAF1/CIPl). J. Biochem. Mol. Biol. 36, 223-229 https://doi.org/10.5483/BMBRep.2003.36.2.223
  9. Choi, B. T., J. Cheong and Y. H. Choi. B-lapachone-induced apoptosis is associated with activation of caspase-3 and inactivation of NF-$\kappa$B in human colon cancer HCT-116 cells. Anticancer Drugs 14, 845-850
  10. Don, M. J., Y. H. Chang, K. K. Chen, L. K. Ho and Y. P. Chau. 2001. Induction of CDK inhibitors (p21(WAF1) and p27(Kip1)) and Bak in the $\beta$-lapachone-induced apoptosis of human prostate cancer cells. Mol. Pharmacol. 59, 784-794
  11. EI-Deiry, W. S., T. Tokino, V. E. Velculesco, D. B. Levy, R. Parsons, J. M. Trent, D. Lin, E. W. Mercer, K. W. Kinzler and B. Vogelstain. 1993. WAF1, a potential mediator of p53 tumor suppression. Cell 75, 817-825 https://doi.org/10.1016/0092-8674(93)90500-P
  12. Goijman, S. G. and A. O. Stoppani. 1985. Effects of ${\beta}$-lapachone, a peroxide-generating quinone, on macromolecule synthesis and degradation in Trypanosoma cruzi. Arch. Biochem. Biaphys. 240, 273-280 https://doi.org/10.1016/0003-9861(85)90033-5
  13. Guiraud, P., R. SteimanR, G. M. Campos-Takaki, F. SeigleMurandi and M. Simeon de Buochberg. 1994. Comparison of antibacterial and antifungal activities of lapachol and ${\beta}$-lapachone. Planta Med. 60, 373-374 https://doi.org/10.1055/s-2006-959504
  14. Homayoun, V. and B. Sam. 1996. From telomere loss to p53 induction and activation of a DNA-damage pathway at senescence: The telomere loss/DNA damage model of cell aging. Exp. Gerontol. 31, 295-301 https://doi.org/10.1016/0531-5565(95)02025-X
  15. Horikawa, J. and J. C. Barrett. 2003. Transcriptional regulation of the telomerase hTERT gene as a target for cellular and viral oncogenic mechanisms. Carcinogenesis 24, 1167-1176 https://doi.org/10.1093/carcin/bgg085
  16. Hueber, A., P. Esser, K. Heimann, N. Kociok, S. Winter and M. Weller. 1998. The topoisomerase I inhibitors, camptothecin and ${\beta}$-lapachone, induce apoptosis of human retinal pigment epithelial cells. Exp. Eye Res. 67, 525-530 https://doi.org/10.1006/exer.1998.0544
  17. Kumi-Diaka, J. 2002. Chemosensitivity of human prostate cancer cells PC3 and LNCaP to genistein isoflavone and ${\beta}$-lapachone. BioI. Cell 94, 37-44 https://doi.org/10.1016/S0248-4900(01)01175-3
  18. Li, C. J., J. Averboukh and A B. Pardee. 1993. ${\beta}$-Lapachone, a novel DNA topoisomerase I inhibitor with a mode of action different from camptothecin. J. BioI. Chem. 268, 22463-22468
  19. Li, L., S. He, J. M. Sun and J. R. Davie. 2004. Gene regulation by Sp1 and Sp3. Biochem. Cell BioI. 82, 460-471
  20. Liu, T. J., S. Y. Lin and Y. P. Chau. 2002. Inhibition of poly (ADP-ribose) polymerase activation attenuates $\beta$-lapachoneinduced necrotic cell death in human osteosarcoma cells. Toxicol. Appl. Pharmacol. 182, 116-125 https://doi.org/10.1006/taap.2002.9438
  21. Lopes, J. N., F. S. Cruz, R. Docampo, M. S. Vasconcellos, M. C. Sampaio, A. V. Pinto and B. Gilbert. 1978. In vitro and in vivo evaluation of the toxicity of 1,4-naphthoquinone and 1,2- naphthoquinone derivatives against Trypanosoma cruzi. Ann. Trap. Med. Parasitol. 72, 523-531
  22. Maga, G. and U. Hubscher. 2003. Proliferating cell nuclear antigen (PCNA): a dancer with many partners. J. Cell Sci. 116, 3051-3060 https://doi.org/10.1242/jcs.00653
  23. Mathieu, N., J. Pirzio, M. A. Freulet-Marriere, C. Desmaze and J. Sabatier. 2004. Telomeres and chromosomal instability. Cell Mol. Life Sci. 61, 641-656 https://doi.org/10.1007/s00018-003-3296-0
  24. Pardee, A. B., Y. Z. Li and C. J. Li. 2002. Cancer therapy with ${\beta}$-lapachone. Curr. Cancer. Drug Targets 2, 227-242 https://doi.org/10.2174/1568009023333854
  25. Planchon, S. M., S. M. Wuerzberger-Davis, J. J. Pink, K. A. Robertson, W. G. Bommann and D. A. Boothman. 1999. Bcl-2 protects against ${\beta}$-lapachone-mediated caspase 3 activation and apoptosis in human myeloid leukemia (HL-60) cells. Oneal. Rep. 6, 485-492
  26. Planchon, S. M., J. J. Pink, C. Tagliarino, W. G. Bornmann, M.E. Varnes and D. A. Boothman. 2001. ${\beta}$-lapachone-induced apoptosis in human prostate cancer cells: involvement of NQO1/xip3. Exp. Cell. Res. 267, 95-106 https://doi.org/10.1006/excr.2001.5234
  27. Rezler, E. M., D. J. Bearss and L. H. Hurley. 2002. Telomeres and telomerases as drug targets. Curr. Opin. Pharmacol. 2, 415-423 https://doi.org/10.1016/S1471-4892(02)00182-0
  28. Rickers, A., N. Peters, V. Badock, R. Beyaert, P. Vandenabeele, B. Dorken and K. Bommert. 1999. Cleavage of transcription factor SPl by caspases during anti-lgM-induced B-cell apoptosis. Eur. J. Biochem. 261, 269-274 https://doi.org/10.1046/j.1432-1327.1999.00273.x
  29. Schaffner-Sabba, K., K. H. Schmidt-Ruppin, W. Wehrli, A. R. Schuerch and J.W. Wasley. 1984. ${\beta}$-lapachone: synthesis of derivatives and activities in tumor models. J. Med. Chem. 27, 990-994 https://doi.org/10.1021/jm00374a010
  30. She, Q. B., A. M. Bode, W.Y. Ma, N.Y. Chen and Z. Dong. 2001. Resveratrol-induced activation of p53 and apoptosis is mediated by extracellular-signal-regulated protein kinases and p38 kinase. Cancer Res. 61, 1604-1610
  31. Sherr, C.J. 2000. The Pezcoller lecture: cancer cell cycles revisited. Cancer Res. 60, 3689-3695
  32. Song, J., H. Ugai, H. Nakata-Tsutsui, S. Kishikawa, E. Suzuki, T. Murata and K.K. Yokoyama. 2003. Transcriptional regulation by zinc-finger proteins Sp1 and MAZ involves interactions with the same cis-elements. Int. J. Mol. Med. 11, 547-553
  33. Surh, Y.J., K.S. Chun, H.H. Cha, S.S. Han, Y.S. Keum, K.K. Park and S.S. Lee. 2001. Molecular mechanisms underlying chemopreventive activities of anti-inflanunatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-$\kappa$B activation. Mutat. Res. 480-481, 243-268
  34. Wang, C.H., Y.P. Tsao, H.J. Chen, H.L. Chen, H.W. Wang and S.L. Chen. 2000. Transcriptional repression of p21(Waf1/Cip1/Sdil) gene by c-jun through Sp1 site. Biochem. Biophys. Res. Commun. 270, 303-310 https://doi.org/10.1006/bbrc.2000.2422
  35. Weinberg, R.A. 1995. The retinoblastoma protein and cell cycle control. Cell 81, 323-330 https://doi.org/10.1016/0092-8674(95)90385-2
  36. Weller, M., S. Winter, C. Schmidt, P. Esser, A. Fontana, J. Dichgans and P. Groscurth. 1997. Topoisomerase-I inhibitors for human malignant glioma: differential modulation of p53, p21, bax and bcl-2 expression and of CD95-mediated apoptosis by camptothecin and ${\beta}$-lapachone. Int. J. Cancer. 73, 707-714 https://doi.org/10.1002/(SICI)1097-0215(19971127)73:5<707::AID-IJC16>3.0.CO;2-2
  37. Xiong, Y., G. Hannon, H. Zhang, D. Casso, R. Kobayashi and D. Beach. 1993. p21 is a universal inhibitor of cyclin kinases. Nature 366, 701-704 https://doi.org/10.1038/366701a0