DOI QR코드

DOI QR Code

Induction of apoptotic cell death in human bladder cancer cells by ethanol extract of Zanthoxylum schinifolium leaf, through ROS-dependent inactivation of the PI3K/Akt signaling pathway

  • Park, Cheol (Division of Basic Sciences, College of Liberal Studies, Dong-eui University) ;
  • Choi, Eun Ok (Anti-Aging Research Center, Dong-eui University) ;
  • Hwangbo, Hyun (Anti-Aging Research Center, Dong-eui University) ;
  • Lee, Hyesook (Anti-Aging Research Center, Dong-eui University) ;
  • Jeong, Jin-Woo (Nakdonggang National Institute of Biological Resources) ;
  • Han, Min Ho (National Marine Biodiversity Institute of Korea) ;
  • Moon, Sung-Kwon (Department of Food and Nutrition, Chung-Ang University) ;
  • Yun, Seok Joong (Department of Urology, College of Medicine, Chungbuk National University) ;
  • Kim, Wun-Jae (Department of Urology, College of Medicine, Chungbuk National University) ;
  • Kim, Gi-Young (Department of Marine Life Science, Jeju National University) ;
  • Hwang, Hye-Jin (Department of Food and Nutrition, College of Nursing, Healthcare Sciences & Human Ecology, Dong-eui University) ;
  • Choi, Yung Hyun (Anti-Aging Research Center, Dong-eui University)
  • Received : 2021.08.25
  • Accepted : 2021.11.23
  • Published : 2022.06.01

Abstract

BACKGROUND/OBJECTIVES: Zanthoxylum schinifolium is traditionally used as a spice for cooking in East Asian countries. This study was undertaken to evaluate the anti-proliferative potential of ethanol extracts of Z. schinifolium leaves (EEZS) against human bladder cancer T24 cells. MATERIALS/METHODS: Subsequent to measuring the cytotoxicity of EEZS, the anti-cancer activity was measured by assessing apoptosis induction, reactive oxygen species (ROS) generation, and mitochondrial membrane potential (MMP). In addition, we determined the underlying mechanism of EEZS-induced apoptosis through various assays, including Western blot analysis. RESULTS: EEZS treatment concentration-dependently inhibited T24 cell survival, which is associated with apoptosis induction. Exposure to EEZS induced the expression of Fas and Fas-ligand, activated caspases, and subsequently resulted to cleavage of poly (ADP-ribose) polymerase. EEZS also enhanced the expression of cytochrome c in the cytoplasm by suppressing MMP, following increase in the ratio of Bax:Bcl-2 expression and truncation of Bid. However, EEZS-mediated growth inhibition and apoptosis were significantly diminished by a pan-caspase inhibitor. Moreover, EEZS inhibited activation of the phosphoinositide 3-kinase (PI3K)/Akt pathway, and the apoptosis-inducing potential of EEZS was promoted in the presence of PI3K/Akt inhibitor. In addition, EEZS enhanced the production of ROS, whereas N-acetyl cysteine (NAC), a ROS scavenger, markedly suppressed growth inhibition and inactivation of the PI3K/Akt signaling pathway induced by EEZS. Furthermore, NAC significantly attenuated the EEZS-induced apoptosis and reduction of cell viability. CONCLUSIONS: Taken together, our results indicate that exposure to EEZS exhibits anti-cancer activity in T24 bladder cancer cells through ROS-dependent induction of apoptosis and inactivation of the PI3K/Akt signaling pathway.

Keywords

Acknowledgement

This work was supported by the National Research Foundation of Korea Grant (NRF-2020R1A2C1099910) and Korea Basic Science Institute Grant funded (NRF-2020R1A6C101A201).

References

  1. Kiraz Y, Adan A, Kartal Yandim M, Baran Y. Major apoptotic mechanisms and genes involved in apoptosis. Tumour Biol 2016;37:8471-86. https://doi.org/10.1007/s13277-016-5035-9
  2. Pfeffer CM, Singh ATK. Apoptosis: a target for anticancer therapy. Int J Mol Sci 2018;19:448. https://doi.org/10.3390/ijms19020448
  3. Hassan M, Watari H, AbuAlmaaty A, Ohba Y, Sakuragi N. Apoptosis and molecular targeting therapy in cancer. Biomed Res Int 2014;2014:150845. https://doi.org/10.1155/2014/150845
  4. Birkinshaw RW, Czabotar PE. The BCL-2 family of proteins and mitochondrial outer membrane permeabilisation. Semin Cell Dev Biol 2017;72:152-62. https://doi.org/10.1016/j.semcdb.2017.04.001
  5. Edlich F. BCL-2 proteins and apoptosis: recent insights and unknowns. Biochem Biophys Res Commun 2018;500:26-34. https://doi.org/10.1016/j.bbrc.2017.06.190
  6. Faes S, Dormond O. PI3K and AKT: unfaithful partners in cancer. Int J Mol Sci 2015;16:21138-52. https://doi.org/10.3390/ijms160921138
  7. Lien EC, Dibble CC, Toker A. PI3K signaling in cancer: beyond AKT. Curr Opin Cell Biol 2017;45:62-71. https://doi.org/10.1016/j.ceb.2017.02.007
  8. Mayer IA, Arteaga CL. The PI3K/AKT pathway as a target for cancer treatment. Annu Rev Med 2016;67:11-28. https://doi.org/10.1146/annurev-med-062913-051343
  9. Badrinath N, Yoo SY. Mitochondria in cancer: in the aspects of tumorigenesis and targeted therapy. Carcinogenesis 2018;39:1419-30. https://doi.org/10.1093/carcin/bgy148
  10. Moloney JN, Cotter TG. ROS signalling in the biology of cancer. Semin Cell Dev Biol 2018;80:50-64. https://doi.org/10.1016/j.semcdb.2017.05.023
  11. Mi Y, Xiao C, Du Q, Wu W, Qi G, Liu X. Momordin Ic couples apoptosis with autophagy in human hepatoblastoma cancer cells by reactive oxygen species (ROS)-mediated PI3K/Akt and MAPK signaling pathways. Free Radic Biol Med 2016;90:230-42. https://doi.org/10.1016/j.freeradbiomed.2015.11.022
  12. Karimian A, Mir SM, Parsian H, Refieyan S, Mirza-Aghazadeh-Attari M, Yousefi B, Majidinia M. Crosstalk between phosphoinositide 3-kinase/Akt signaling pathway with DNA damage response and oxidative stress in cancer. J Cell Biochem 2019;120:10248-72. https://doi.org/10.1002/jcb.28309
  13. Song X, Wang Z, Liang H, Zhang W, Ye Y, Li H, Hu Y, Zhang Y, Weng H, Lu J, et al. Dioscin induces gallbladder cancer apoptosis by inhibiting ROS-mediated PI3K/AKT signalling. Int J Biol Sci 2017;13:782-93. https://doi.org/10.7150/ijbs.18732
  14. Sun X, Duan Z. Research progress of medicinal plants of Zanthoxylum linn. Yao Xue Xue Bao 1996;31:231-40.
  15. Hu L, Wang K, Wang Z, Liu J, Wang K, Zhang J, Luo Z, Xue Y, Zhang Y, Zhang Y. A new megastigmane sesquiterpenoid from Zanthoxylum schinifolium Sieb. et Zucc. Molecules 2016;21:383. https://doi.org/10.3390/molecules21030383
  16. Liu ZL, Chu SS, Jiang GH. Feeding deterrents from Zanthoxylum schinifolium against two stored-product insects. J Agric Food Chem 2009;57:10130-3. https://doi.org/10.1021/jf9012983
  17. Tsai IL, Lin WY, Teng CM, Ishikawa T, Doong SL, Huang MW, Chen YC, Chen IS. Coumarins and antiplatelet constituents from the root bark of Zanthoxylum schinifolium. Planta Med 2000;66:618-23. https://doi.org/10.1055/s-2000-8648
  18. Cao LH, Lee YJ, Kang DG, Kim JS, Lee HS. Effect of Zanthoxylum schinifolium on TNF-alpha-induced vascular inflammation in human umbilical vein endothelial cells. Vascul Pharmacol 2009;50:200-7. https://doi.org/10.1016/j.vph.2009.01.008
  19. Choi EO, Park C, Shin SS, Cho EJ, Kim BW, Hwang JA, Hwang HJ, Choi YH. Zanthoxylum schinifolium leaf ethanol extract inhibits adipocyte differentiation through inactivation of the extracellular signal regulated kinase and phosphoinositide 3-kinase/Akt signaling pathways in 3T3-L1 pre-adipocytes. Mol Med Rep 2015;12:1314-20. https://doi.org/10.3892/mmr.2015.3463
  20. Nguyen PH, Zhao BT, Kim O, Lee JH, Choi JS, Min BS, Woo MH. Anti-inflammatory terpenylated coumarins from the leaves of Zanthoxylum schinifolium with α-glucosidase inhibitory activity. J Nat Med 2016;70:276-81. https://doi.org/10.1007/s11418-015-0957-x
  21. Kim KK, Kim TW, Kang YH, Kim DJ, Choe M. Lipid-lowering effects of Zanthoxylum schinifolium Siebold & Zucc. seed oil (ZSO) in hyperlipidemic rats and lipolytic effects in 3T3-L1 adipocytes. Food Sci Biotechnol 2016;25:1427-36. https://doi.org/10.1007/s10068-016-0222-4
  22. Lee HY, Park YM, Lee YH, Kang YG, Lee HM, Park DS, Yang HJ, Kim MJ, Lee YR. Immunostimulatory effect of Zanthoxylum schinifolium-based complex oil prepared by supercritical fluid extraction in splenocytes and cyclophosphamide-induced immunosuppressed rats. Evid Based Complement Alternat Med 2018;2018:8107326.
  23. Lee SW, Lim JM, Mohan H, Seralathan KK, Park YJ, Lee JH, Oh BT. Enhanced bioactivity of Zanthoxylum schinifolium fermented extract: Anti-inflammatory, anti-bacterial, and anti-melanogenic activity. J Biosci Bioeng 2020;129:638-45. https://doi.org/10.1016/j.jbiosc.2019.12.003
  24. Paik SY, Koh KH, Beak SM, Paek SH, Kim JA. The essential oils from Zanthoxylum schinifolium pericarp induce apoptosis of HepG2 human hepatoma cells through increased production of reactive oxygen species. Biol Pharm Bull 2005;28:802-7. https://doi.org/10.1248/bpb.28.802
  25. Jun DY, Kim JS, Park HS, Han CR, Fang Z, Woo MH, Rhee IK, Kim YH. Apoptogenic activity of auraptene of Zanthoxylum schinifolium toward human acute leukemia Jurkat T cells is associated with ER stress-mediated caspase-8 activation that stimulates mitochondria-dependent or -independent caspase cascade. Carcinogenesis 2007;28:1303-13. https://doi.org/10.1093/carcin/bgm028
  26. Min BK, Hyun DG, Jeong SY, Kim YH, Ma ES, Woo MH. A new cytotoxic coumarin, 7-[(E)-3',7'-dimethyl-6'-oxo-2',7'-octadienyl] oxy coumarin, from the leaves of Zanthoxylum schinifolium. Arch Pharm Res 2011;34:723-6. https://doi.org/10.1007/s12272-011-0504-6
  27. Li W, Sun YN, Yan XT, Yang SY, Kim EJ, Kang HK, Kim YH. Coumarins and lignans from Zanthoxylum schinifolium and their anticancer activities. J Agric Food Chem 2013;61:10730-40. https://doi.org/10.1021/jf403479c
  28. Choi YH. Trans-cinnamaldehyde protects C2C12 myoblasts from DNA damage, mitochondrial dysfunction and apoptosis caused by oxidative stress through inhibiting ROS production. Genes Genomics 2021;43:303-12. https://doi.org/10.1007/s13258-020-00987-9
  29. Pham TNA, Le B, Yang SH. Anticancer activity of the potential Pyropia yezoensis galactan fractionated in human prostate cancer cells. Biotechnol Bioprocess Eng 2021;26:63-70. https://doi.org/10.1007/s12257-020-0157-8
  30. Park S, Kim M, Hong Y, Lee H, Tran Q, Kim C, Kwon SH, Park J, Park J, Kim SH. Myristoylated TMEM39AS41, a cell-permeable peptide, causes lung cancer cell death. Toxicol Res 2020;36:123-30. https://doi.org/10.1007/s43188-020-00038-1
  31. Liang Y, Kong D, Zhang Y, Li S, Li Y, Ramamoorthy S, Ma J. Fisetin inhibits cell proliferation and induces apoptosis via JAK/STAT3 signaling pathways in human thyroid TPC 1 cancer cells. Biotechnol Bioprocess Eng 2020;25:197-205. https://doi.org/10.1007/s12257-019-0326-9
  32. Hwangbo H, Kim SY, Lee H, Park SH, Hong SH, Park C, Kim GY, Leem SH, Hyun JW, Cheong J, et al. Auranofin enhances sulforaphane-mediated apoptosis in hepatocellular carcinoma Hep3B cells through inactivation of the PI3K/Akt signaling pathway. Biomol Ther (Seoul) 2020;28:443-55. https://doi.org/10.4062/biomolther.2020.122
  33. Bae CS, Lee CM, Ahn T. Encapsulation of apoptotic proteins in lipid nanoparticles to induce death of cancer cells. Biotechnol Bioprocess Eng 2020;25:264-71. https://doi.org/10.1007/s12257-019-0409-7
  34. Yin XM. Bid, a BH3-only multi-functional molecule, is at the cross road of life and death. Gene 2006;369:7-19. https://doi.org/10.1016/j.gene.2005.10.038
  35. Billen LP, Shamas-Din A, Andrews DW. Bid: a Bax-like BH3 protein. Oncogene 2008;27 Suppl 1:S93-104. https://doi.org/10.1038/onc.2009.47
  36. Kantari C, Walczak H. Caspase-8 and Bid: caught in the act between death receptors and mitochondria. Biochim Biophys Acta 2011;1813:558-63. https://doi.org/10.1016/j.bbamcr.2011.01.026
  37. Houede N, Pourquier P. Targeting the genetic alterations of the PI3K-AKT-mTOR pathway: its potential use in the treatment of bladder cancers. Pharmacol Ther 2015;145:1-18. https://doi.org/10.1016/j.pharmthera.2014.06.004
  38. Sathe A, Nawroth R. Targeting the PI3K/AKT/mTOR pathway in bladder cancer. Methods Mol Biol 2018;1655:335-50. https://doi.org/10.1007/978-1-4939-7234-0_23
  39. Song G, Ouyang G, Bao S. The activation of Akt/PKB signaling pathway and cell survival. J Cell Mol Med 2005;9:59-71. https://doi.org/10.1111/j.1582-4934.2005.tb00337.x
  40. Manning BD, Cantley LC. AKT/PKB signaling: navigating downstream. Cell 2007;129:1261-74. https://doi.org/10.1016/j.cell.2007.06.009
  41. Galadari S, Rahman A, Pallichankandy S, Thayyullathil F. Reactive oxygen species and cancer paradox: to promote or to suppress? Free Radic Biol Med 2017;104:144-64. https://doi.org/10.1016/j.freeradbiomed.2017.01.004
  42. Lee YJ, Yoon JJ, Lee SM, Kim JS, Kang DG, Lee HS. Inhibitory effect of Zanthoxylum schinifolium on vascular smooth muscle proliferation. Immunopharmacol Immunotoxicol 2012;34:354-61. https://doi.org/10.3109/08923973.2011.608070