DOI QR코드

DOI QR Code

Selenium Inhibits Metastasis of Murine Melanoma Cells through the Induction of Cell Cycle Arrest and Cell Death

  • Song, Hyun-Keun (Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine) ;
  • Hur, In-Do (Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine) ;
  • Park, Hyun-Jin (Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine) ;
  • Nam, Joo-Hyung (Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine) ;
  • Park, Ga-Bin (Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine) ;
  • Kong, Kyoung-Hye (Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine) ;
  • Hwang, Young-Mi (Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine) ;
  • Kim, Yeong-Seok (Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine) ;
  • Cho, Dae-Ho (Department of LifeScience, Sookmyung Women's University) ;
  • Lee, Wang-Jae (Department of Anatomy and Cancer Immunology, Seoul National University College of Medicine) ;
  • Hur, Dae-Young (Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine)
  • Received : 2009.11.20
  • Accepted : 2009.12.01
  • Published : 2009.12.31

Abstract

Background: Melanoma is the most fatal form of skin cancer due to its rapid metastasis. Recently, several studies reported that selenium can induce apoptosis in melanoma cells. However, the precise mechanism remains to be elucidated. In this study, we investigated the effect of selenium on cell proliferation in murine melanoma and on tumor growth and metastasis in C57BL/6 mice. Methods: Cell proliferation was measured by MTT assay in selenium-treated melanoma cells. Cell cycle distribution was analysized by staining DNA with propidum iodide (PI). mRNA and protein expression related to cell cycle arrest was measured by reverse transcription PCR and western blot. Tumor growth and metastasis was measured by in vivo model. Results: Selenium was suppressed the proliferation of melanoma cells in a dose dependent manner. The growth inhibition of melanoma by selenium was associated with an arrest of cell cycle distribution at G0/G1 stage. The mRNA and protein level of CDK2/CDK4 was suppressed by treatment with selenium in a time-dependent manner. In vivo, tumor growth was not suppressed by selenium; however tumor metastasis was suppressed by selenium in mouse model. Conclusion: These results suggest that selenium might be a potent agent to inhibit proliferative activity of melanoma cells.

Keywords

References

  1. Mancianti ML, Herlyn M: Tumor progression in melanoma: the biology of epidermal melanocytes in vitro. Carcinog Compr Surv 11;369-386, 1989
  2. Nagao N, Nakayama T, Etoh T, Saiki I, Miwa N: Tumor invasion is inhibited by phosphorylated ascorbate via enrichment of intracellular vitamin C and decreasing of oxidative stress. J Cancer Res Clin Oncol 126;511-518, 2000 https://doi.org/10.1007/s004320000120
  3. Roomi MW, Roomi N, Ivanov V, Kalinovsky T, Niedzwiecki A, Rath M: Inhibition of pulmonary metastasis of melanoma b16fo cells in C57BL/6 mice by a nutrient mixture consisting of ascorbic Acid, lysine, proline, arginine, and green tea extract. Exp Lung Res 32;517-530, 2006 https://doi.org/10.1080/01902140601098552
  4. Dua P, Ingle A, Gude RP: Suramin augments the antitumor and antimetastatic activity of pentoxifylline in B16F10 melanoma. Int J Cancer 121;1600-1608, 2007 https://doi.org/10.1002/ijc.22843
  5. Combs GF Jr, Gray WP: Chemopreventive agents: selenium. Pharmacol Ther 79;179-192, 1998 https://doi.org/10.1016/S0163-7258(98)00014-X
  6. Patrick L: Selenium biochemistry and cancer: a review of the literature. Altern Med Rev 9;239-258, 2004
  7. Han B, Wei W, Hua F, Cao T, Dong H, Yang T, Yang Y, Pan H, Xu C: Requirement for ERK activity in selenium-induced apoptosis of acute promyelocytic leukemia-derived NB4 cells. J Biochem Mol Biol 40;196-204, 2007 https://doi.org/10.5483/BMBRep.2007.40.2.196
  8. Kim EH, Sohn S, Kwon HJ, Kim SU, Kim MJ, Lee SJ, Choi KS: Selenium induces superoxide-mediated mitochondrial damage and subsequent autophagic cell death in malignant glioma cells. Cancer Res 67;6314-6324, 2007 https://doi.org/10.1158/0008-5472.CAN-06-4217
  9. Wang H, Yang X, Zhang Z, Xu H: Both calcium and ROS as common signals mediate Na(2)SeO(3)-induced apoptosis in SW480 human colonic carcinoma cells. J Inorg Biochem 97;221-230, 2003 https://doi.org/10.1016/S0162-0134(03)00284-8
  10. Kramer GF, Ames BN: Mechanisms of mutagenicity and toxicity of selenium (Na2SeO3) in salmonella typhimurium. Mutat Res 201;169-180, 1988 https://doi.org/10.1016/0027-5107(88)90123-6
  11. Kim T, Jung U, Cho DY, CHung AS: Se-methylselencysteine induces apoptosis through caspase activation in HL-60 cells. carcinogenesis 22;559-565, 2001 https://doi.org/10.1093/carcin/22.4.559
  12. Jung U, Zheng X, Yoon SO, Chung AS: Se-methylselenocysteine induces apoptosis mediated by reactive oxygen species in HL-60. Free radiac Biol Med 31;479-489, 2001 https://doi.org/10.1016/S0891-5849(01)00604-9
  13. Sherr CJ: cancer cell cycles revisited. Cancer Res 60: 3689-3695, 2000
  14. Kaeck M, Lu J, Strange R, Ip C, Ganther HE, Thompson HJ: Differential induction of growth arrest inducible genes by selenium compounds. BioChemical Pharmacology 53; 921-926, 1997 https://doi.org/10.1016/S0006-2952(97)00103-2
  15. Rudolf E, Rudolf K, Cervinka M: Selenium activates p53 and p38 pathways and induces caspase-independent cell death in cervical cancer cells. Cell biol Toxicol 24;123-241, 2008 https://doi.org/10.1007/s10565-007-9022-1
  16. Vermeulen K, Van Bockstaele DR, Berneman ZN: The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer. Cell Prolif 36;131-149, 2003 https://doi.org/10.1046/j.1365-2184.2003.00266.x
  17. Albino AP, Juan G, Traganos F, Reinhart L, Connolly J, Rose DP, Darzynkiewicz Z: Cell cycle arrest and apoptosis of melanoma cells by docosahexaenoic acid: association with decreased pRb phosphorylation. Cancer Res 60;4139-4145, 2000
  18. Tsai J, Lee JT, Wang W, Zhang J, Cho H, Mamo S, Bremer R, Gillette S, Kong J, Haass NK, Sproesser K, Li L, Smalley KS, Fong D, Zhu YL, Marimuthu A, Nguyen H, Lam B, Liu J, Cheung I, Rice J, Suzuki Y, Luu C, Settachatgul C, Shellooe R, Cantwell J, Kim SH, Schlessinger J, Zhang KY, West BL, Powell B, Habets G, Zhang C, Ibrahim PN, Hirth P, Artis DR, Herlyn M, Bollag G: Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent anti-melanoma activity. PNAS 105;3041-3046, 2008 https://doi.org/10.1073/pnas.0711741105
  19. Wang CC, Chiang YM, Sung SC, Hsu YL, Chang JK, Kuo PL: Plumbagin induces cell cycle arrest and apoptosis through reactive oxygen species/c-Jun N-terminal kinase pathways in human melanoma A375.S2 cells. Cancer Letters 259;82-98, 2008 https://doi.org/10.1016/j.canlet.2007.10.005
  20. An WW, Wang MW, Tashiro S, Onodera S, Ikejima T: Mitogen-activated protein kinase-dependent apoptosis in norcan-tharidin-treated A375-S2 cells is proceeded by the activation of protein kinase C. Chin Med J (Engl) 118; 198-203, 2005
  21. Mhaidat NM, Zhang XD, Allen J, Avery-Kiejda KA, Scott RJ, Hersey P: Temozolomide induces senescence but not apoptosis in human melanoma cells. Br J Cancer 97;1225-1233, 2007 https://doi.org/10.1038/sj.bjc.6604017
  22. Kagawa S, Fujiwara T, Hizuta A, Yasuda T, Zhang WW, Roth JA, Tanaka N: p53 expression overcomes p21WAF1/CIP1-mediated G1 arrest and induces apoptosis in human cancer cells. Oncogene 15;1903-1909, 1997 https://doi.org/10.1038/sj.onc.1201362
  23. Jiang C, Hu H, Malewiez B, Wang Z, Lu J: Selenite-induced p53 Ser-15 phosphorylation and caspase-mediated apoptosis in LNCaP human prostate cancer cells. Mol Cancer Ther 3;877-884, 2004
  24. Traynor NJ, McKenzie RC, Beckett GJ, Gibbs NK: Seleno-methionine inhibits ultraviolet radiation-induced p53 transactivation. Photodermatol Photoimmunol Photomed 22; 297-303, 2006 https://doi.org/10.1111/j.1600-0781.2006.00256.x
  25. Hahm E, Jin DH, Kang JS, Kim YI, Hong SW, Lee SK, Kim HN, Jung da J, Kim JE, Shin DH, Hwang YI, Kim YS, Hur DY, Yang Y, Cho D, Lee MS, Lee WJ: The Molecular Mechanisms of Vitamin C on Cell Cycle Regulation in B16F10 Murine Melanoma. J Cell Biochem 102;1002-1010, 2007 https://doi.org/10.1002/jcb.21336
  26. Deeds L, Teodorescu S, Chu M, Yu Q, Chen C: A p53-independent G1 cell cycle checkpoint induced by the suppression of protein kinase C alpha and theta isoforms. J Biol Chem 278;39782-39793, 2003 https://doi.org/10.1074/jbc.M306854200
  27. Peter M, Herskowitz I: Joining the complex: Cyclindependent kinase inhibitory proteins and the cell cycle. Cell 9;181-184, 1994

Cited by

  1. Surface decoration of selenium nanoparticles by mushroom polysaccharides–protein complexes to achieve enhanced cellular uptake and antiproliferative activity vol.22, pp.19, 2012, https://doi.org/10.1039/c2jm16828f
  2. Is Selenium a Potential Treatment for Cancer Metastasis? vol.5, pp.4, 2009, https://doi.org/10.3390/nu5041149
  3. Supplementation with Selenium Can Influence Nausea, Fatigue, Physical, Renal, and Liver Function of Children and Adolescents with Cancer vol.18, pp.1, 2009, https://doi.org/10.1089/jmf.2014.0030
  4. Selenium and Human Health: Witnessing a Copernican Revolution? vol.33, pp.3, 2009, https://doi.org/10.1080/10590501.2015.1055163
  5. Dietary Selenium Supplementation Modulates Growth of Brain Metastatic Tumors and Changes the Expression of Adhesion Molecules in Brain Microvessels vol.172, pp.None, 2009, https://doi.org/10.1007/s12011-015-0595-x
  6. AMPK interacts with β-catenin in the regulation of hepatocellular carcinoma cell proliferation and survival with selenium treatment vol.35, pp.3, 2009, https://doi.org/10.3892/or.2015.4519
  7. A Selenium Containing Inhibitor for the Treatment of Hepatocellular Cancer vol.9, pp.2, 2009, https://doi.org/10.3390/ph9020018
  8. Nutraceuticals and their role in tumor angiogenesis vol.408, pp.2, 2009, https://doi.org/10.1016/j.yexcr.2021.112859