DOI QR코드

DOI QR Code

Apigenin causes necroptosis by inducing ROS accumulation, mitochondrial dysfunction, and ATP depletion in malignant mesothelioma cells

  • Lee, Yoon-Jin (Department of Biochemistry, Soonchunhyang University College of Medicine) ;
  • Park, Kwan-Sik (Department of Biochemistry, Soonchunhyang University College of Medicine) ;
  • Nam, Hae-Seon (Division of Molecular Cancer Research, Soonchunhyang Medical Research Institute, Soonchunhyang University) ;
  • Cho, Moon-Kyun (Division of Molecular Cancer Research, Soonchunhyang Medical Research Institute, Soonchunhyang University) ;
  • Lee, Sang-Han (Department of Biochemistry, Soonchunhyang University College of Medicine)
  • 투고 : 2020.06.17
  • 심사 : 2020.09.26
  • 발행 : 2020.11.01

초록

Apigenin, a naturally occurring flavonoid, is known to exhibit significant anticancer activity. This study was designed to determine the effects of apigenin on two malignant mesothelioma cell lines, MSTO-211H and H2452, and to explore the underlying mechanism(s). Apigenin significantly inhibited cell viability with a concomitant increase in intracellular reactive oxygen species (ROS) and caused the loss of mitochondrial membrane potential (ΔΨm), and ATP depletion, resulting in apoptosis and necroptosis in monolayer cell culture. Apigenin upregulated DNA damage response proteins, including the DNA double strand break marker phospho (p)-histone H2A.X. and caused a transition delay at the G2/M phase of cell cycle. Western blot analysis showed that apigenin treatment upregulated protein levels of cleaved caspase-3, cleaved PARP, p-MLKL, and p-RIP3 along with an increased Bax/Bcl-2 ratio. ATP supplementation restored cell viability and levels of DNA damage-, apoptosisand necroptosis-related proteins that apigenin caused. In addition, N-acetylcysteine reduced ROS production and improved ΔΨm loss and cell death that were caused by apigenin. In a 3D spheroid culture model, ROS-dependent necroptosis was found to be a mechanism involved in the anti-cancer activity of apigenin against malignant mesothelioma cells. Taken together, our findings suggest that apigenin can induce ROS-dependent necroptotic cell death due to ATP depletion through mitochondrial dysfunction. This study provides us a possible mechanism underlying why apigenin could be used as a therapeutic candidate for treating malignant mesothelioma.

키워드

참고문헌

  1. Hartwell LH, Weinert TA. Checkpoints: controls that ensure the order of cell cycle events. Science. 1989;246:629-634. https://doi.org/10.1126/science.2683079
  2. Zannini L, Delia D, Buscemi G. CHK2 kinase in the DNA damage response and beyond. J Mol Cell Biol. 2014;6:442-457. https://doi.org/10.1093/jmcb/mju045
  3. Murphy MP. How mitochondria produce reactive oxygen species. Biochem J. 2009;417:1-13. https://doi.org/10.1042/BJ20081386
  4. Higuchi Y. Glutathione depletion-induced chromosomal DNA fragmentation associated with apoptosis and necrosis. J Cell Mol Med. 2004;8:455-464. https://doi.org/10.1111/j.1582-4934.2004.tb00470.x
  5. Liu J, Wang Z. Increased oxidative stress as a selective anticancer therapy. Oxid Med Cell Longev. 2015;2015:294303. https://doi.org/10.1155/2015/294303
  6. Nagata Y, Sawada R, Takashima A, Shoji H, Honma Y, Iwasa S, Amano K, Kato K, Hamaguchi T, Shimada Y, Saruta M, Boku N. Efficacy and safety of pemetrexed plus cisplatin as first-line chemotherapy in advanced malignant peritoneal mesothelioma. Jpn J Clin Oncol. 2019;49:1004-1008. https://doi.org/10.1093/jjco/hyz104
  7. Fennell DA, Rudd RM. Defective core-apoptosis signalling in diffuse malignant pleural mesothelioma: opportunities for effective drug development. Lancet Oncol. 2004;5:354-362. https://doi.org/10.1016/S1470-2045(04)01492-5
  8. Galani V, Varouktsi A, Papadatos SS, Mitselou A, Sainis I, Constantopoulos S, Dalavanga Y. The role of apoptosis defects in malignant mesothelioma pathogenesis with an impact on prognosis and treatment. Cancer Chemother Pharmacol. 2019;84:241-253. https://doi.org/10.1007/s00280-019-03878-3
  9. Grootjans S, Vanden Berghe T, Vandenabeele P. Initiation and execution mechanisms of necroptosis: an overview. Cell Death Differ. 2017;24:1184-1195. https://doi.org/10.1038/cdd.2017.65
  10. Chen D, Yu J, Zhang L. Necroptosis: an alternative cell death program defending against cancer. Biochim Biophys Acta. 2016;1865:228-236.
  11. Diederich M, Cerella C. Non-canonical programmed cell death mechanisms triggered by natural compounds. Semin Cancer Biol. 2016;40-41:4-34. https://doi.org/10.1016/j.semcancer.2016.06.001
  12. Salehi B, Venditti A, Sharifi-Rad M, Kregiel D, Sharifi-Rad J, Durazzo A, Lucarini M, Santini A, Souto EB, Novellino E, Antolak H, Azzini E, Setzer WN, Martins N. The therapeutic potential of apigenin. Int J Mol Sci. 2019;20:1305. https://doi.org/10.3390/ijms20061305
  13. Yan X, Qi M, Li P, Zhan Y, Shao H. Apigenin in cancer therapy: anti-cancer effects and mechanisms of action. Cell Biosci. 2017;7:50. https://doi.org/10.1186/s13578-017-0179-x
  14. Shukla S, Gupta S. Apigenin: a promising molecule for cancer prevention. Pharm Res. 2010;27:962-978. https://doi.org/10.1007/s11095-010-0089-7
  15. Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol. 2014;12:207-218. https://doi.org/10.1089/adt.2014.573
  16. Nunes AS, Barros AS, Costa EC, Moreira AF, Correia IJ. 3D tumor spheroids as in vitro models to mimic in vivo human solid tumors resistance to therapeutic drugs. Biotechnol Bioeng. 2019;116:206-226. https://doi.org/10.1002/bit.26845
  17. Chambers KF, Mosaad EM, Russell PJ, Clements JA, Doran MR. 3D cultures of prostate cancer cells cultured in a novel high-throughput culture platform are more resistant to chemotherapeutics compared to cells cultured in monolayer. PLoS One. 2014;9:e111029. https://doi.org/10.1371/journal.pone.0111029
  18. Federici C, Petrucci F, Caimi S, Cesolini A, Logozzi M, Borghi M, D'Ilio S, Lugini L, Violante N, Azzarito T, Majorani C, Brambilla D, Fais S. Exosome release and low pH belong to a framework of resistance of human melanoma cells to cisplatin. PLoS One. 2014;9:e88193. https://doi.org/10.1371/journal.pone.0088193
  19. Giglia-Mari G, Zotter A, Vermeulen W. DNA damage response. Cold Spring Harb Perspect Biol. 2011;3:a000745. https://doi.org/10.1101/cshperspect.a000745
  20. Masuelli L, Benvenuto M, Mattera R, Di Stefano E, Zago E, Taffera G, Tresoldi I, Giganti MG, Frajese GV, Berardi G, Modesti A, Bei R. In vitro and in vivo anti-tumoral effects of the flavonoid apigenin in malignant mesothelioma. Front Pharmacol. 2017;8:373. https://doi.org/10.3389/fphar.2017.00373
  21. Tait SW, Ichim G, Green DR. Die another way--non-apoptotic mechanisms of cell death. J Cell Sci. 2014;127(Pt 10):2135-2144. https://doi.org/10.1242/jcs.093575
  22. Liu L, Fan J, Ai G, Liu J, Luo N, Li C, Cheng Z. Berberine in combination with cisplatin induces necroptosis and apoptosis in ovarian cancer cells. Biol Res. 2019;52:37. https://doi.org/10.1186/s40659-019-0243-6
  23. Tummers B, Green DR. Caspase-8: regulating life and death. Immunol Rev. 2017;277:76-89. https://doi.org/10.1111/imr.12541
  24. Safa AR. Roles of c-FLIP in apoptosis, necroptosis, and autophagy. J Carcinog Mutagen. 2013;Suppl 6:003.
  25. Carlisi D, Buttitta G, Di Fiore R, Scerri C, Drago-Ferrante R, Vento R, Tesoriere G. Parthenolide and DMAPT exert cytotoxic effects on breast cancer stem-like cells by inducing oxidative stress, mitochondrial dysfunction and necrosis. Cell Death Dis. 2016;7:e2194. https://doi.org/10.1038/cddis.2016.94
  26. Chen S, Lv X, Hu B, Zhao L, Li S, Li Z, Qing X, Liu H, Xu J, Shao Z. Critical contribution of RIPK1 mediated mitochondrial dysfunction and oxidative stress to compression-induced rat nucleus pulposus cells necroptosis and apoptosis. Apoptosis. 2018;23:299-313. https://doi.org/10.1007/s10495-018-1455-x
  27. Eguchi Y, Shimizu S, Tsujimoto Y. Intracellular ATP levels determine cell death fate by apoptosis or necrosis. Cancer Res. 1997;57:1835-1840.

피인용 문헌

  1. Differences of Key Proteins between Apoptosis and Necroptosis vol.2021, 2021, https://doi.org/10.1155/2021/3420168
  2. Codonopsis pilosula Polysaccharides Alleviate Aβ1-40-Induced PC12 Cells Energy Dysmetabolism via CD38/NAD+ Signaling Pathway vol.18, pp.3, 2021, https://doi.org/10.2174/1567205018666210608103831
  3. Optimization of Pre-Inoculum, Fermentation Process Parameters and Precursor Supplementation Conditions to Enhance Apigenin Production by a Recombinant Streptomyces albus Strain vol.7, pp.3, 2020, https://doi.org/10.3390/fermentation7030161