Zerumbone mediates apoptosis and induces secretion of proinflammatory cytokines in breast carcinoma cell culture

Document Type : Original Article

Authors

1 Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur 50603, Malaysia

2 Department of Pharmacology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur 50603, Malaysia

3 Center of Toxicology and Health Risk Studies (CORE), Faculty of Health Sciences, National University of Malaysia, Jalan Raja Muda Aziz, Kuala Lumpur 50300, Malaysia

4 Department of Surgery, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur 50603, Malaysia

Abstract

Objective(s): To investigate the potential anti-breast cancer activity of zerumbone in regulating apoptotic mediators and cytokines in comparison with paclitaxel (positive control). 
Materials and Methods: In this study, assays such as viability, apoptosis, reactive oxygen species, cell cycle, DNA fragmentation, and cytokines were carried out on MCF-7 cells after treatment with zerumbone and paclitaxel.
Results: The results showed that zerumbone demonstrated a higher (18-fold) IC50 value (126.7 µg/ml) than paclitaxel (7.29 µg/ml) in order to suppress proliferation and induce cell death of MCF-7. The cell cycle arrest at the G0/G1 phase and excessive intracellular ROS production during the in vitro zerumbone treatment indicated occurrence of apoptotic cell death although nuclear DNA fragmentation was not observed. The flow cytometer analysis of treated cells revealed secretion of proinflammatory cytokines suggesting the potential immunomodulatory activity of zerumbone. 
Conclusion: Although, zerumbone exhibited a higher IC50 value compared with paclitaxel yet its anticancer activity against MCF-7 cells is still parallel to paclitaxel hence zerumbone has the potential to be an antineoplastic agent in the treatment of breast cancer especially the luminal type A.

Keywords


1. Dolatkhah R, Somi MH, Jafarabadi MA, Hosseinalifam M, Sepahi S, Belalzadeh M, et al. Breast cancer survival and incidence: 10 Years Cancer Registry Data in the Northwest, Iran. Int J Breast Cancer 2020; 2020:1–6. 
2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 0:caac.21660. 
3. Feng Y, Spezia M, Huang S, Yuan C, Zeng Z, Zhang L, et al. Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis 2018; 5:77–106. 
4. Moo TA, Sanford R, Dang C, and Morrow M. Overview of Breast Cancer Therapy. Vol. 13, PET Clinics. 2018; 339–354. 
5. Berardi R, Morgese F, Rinaldi S, Torniai M, Mentrasti G, Scortichini L, et al. Benefits and limitations of a multidisciplinary approach in cancer patient management. Cancer Manag Res 2020; 12:9363–9374. 
6. Tong CWS, Wu M, Cho WCS, To KKW. Recent advances in the treatment of breast cancer. Front Oncol 2018; 8. 
7. Nounou MI, ElAmrawy F, Ahmed N, Abdelraouf K, Goda S, and Syed-Sha-Qhattal H. Breast cancer: Conventional diagnosis and treatment modalities and recent patents and technologies. Breast Cancer (Auckl) 2015; 9:17–34. 
8. Vuong TV. Natural products and their derivatives with antibacterial, anti-oxidant and anticancer activities. Antibiotics 2021; 10:70-72. 
9. Gallego-Jara J, Lozano-Terol G, Sola-Martínez RA, Cánovas-Díaz M, and de Diego Puente T. A Compressive Review about Taxol®: History and Future Challenges. Molecules 2020; 25:5986. 
10. Weaver BA. How Taxol/paclitaxel kills cancer cells. Bement W, editor. Mol Biol Cell 2014; 25:2677–2681. 
11. Carvalho C, Santos R, Cardoso S, Correia S, Oliveira P, Santos M, et al. Doxorubicin: The good, the bad and the ugly effect. Curr Med Chem 2009; 16:3267–3285. 
12. Thorn CF, Oshiro C, Marsh S, Hernandez-Boussard T, McLeod H, Klein TE, et al. Doxorubicin pathways. Pharmacogenet Genomics 2011; 21:440–446. 
13. Qin J, Wang W, and Zhang R. Novel natural product therapeutics targeting both inflammation and cancer. Chin J Nat Med 2017; 15:401–416. 
14. Ashley RE and Osheroff N. Natural products as topoisomerase II poisons: Effects of thymoquinone on DNA cleavage mediated by human topoisomerase IIα. Chem Res Toxicol 2014; 27:787–793. 
15. Zhao Y, Butler EB, and Tan M. Targeting cellular metabolism to improve cancer therapeutics. Cell Death Dis 2013; 4:e532. 
16. Dalla Via L, García-Argáez AN, Martínez-Vázquez M, Grancara S, Martinis P, and Toninello A. Mitochondrial permeability transition as target of anticancer drugs. Curr Pharm Des 2014; 20:223–244. 
17. Endo S, Hoshi M, Matsunaga T, Inoue T, Ichihara K, and Ikari A. Autophagy inhibition enhances anticancer efficacy of artepillin C, a cinnamic acid  derivative in Brazilian green propolis. Biochem Biophys Res Commun 2018; 497:437–443. 
18. Falzone L, Salomone S, and Libra M. Evolution of cancer pharmacological treatments at the turn of the third millennium. Front Pharmacol 2018; 9:1-26. 
19. E. M. Eid E, S. Alanazi A, Koosha S, A. Alrasheedy A, Azam F, M. Taban I, et al. Zerumbone induces apoptosis in breast cancer cells by targeting αvβ3 integrin upon co-administration with TP5-iRGD peptide. Molecules 2019; 24:2554-2568. 
20. Girisa S, Shabnam B, Monisha J, Fan L, Halim C, Arfuso F, et al. Potential of zerumbone as an anti-cancer agent. Molecules 2019; 24:734-753. 
21. Sellés Vidal L, Kelly CL, Mordaka PM, and Heap JT. Review of NAD(P)H-dependent oxidoreductases: Properties, engineering and application. Biochim Biophys Acta - Proteins Proteomics 2018; 1866:327–347. 
22. Aung TN, Qu Z, Kortschak RD, and Adelson DL. Understanding the effectiveness of natural compound mixtures in cancer through their molecular mode of action. Int J Mol Sci 2017; 18. 
23. Jamalzadeh L, Ghafoori H, Aghamaali M, and Sariri R. Induction of apoptosis in human breast cancer MCF-7 cells by a semisynthetic derivative of artemisinin: A caspase-related mechanism. Iran J Biotechnol 2017; 15:157–165. 
24. Daddiouaissa D, Amid A, Kabbashi NA, Fuad FAA, Elnour AM, and Epandy MAKMS. Antiproliferative activity of ionic liquid-graviola fruit extract against human breast cancer (MCF-7) cell lines using flow cytometry techniques. J Ethnopharmacol 2019; 236:466–473. 
25. Nagata S, Suzuki J, Segawa K, and Fujii T. Exposure of phosphatidylserine on the cell surface. Cell Death Differ 2016; 23:952–961. 
26. Ucker DS and Levine JS. Exploitation of Apoptotic Regulation in Cancer. Front Immunol 2018; 9:241. 
27. Siddiqui WA, Ahad A, and Ahsan H. The mystery of BCL2 family: Bcl-2 proteins and apoptosis: an update. Arch Toxicol 2015; 89:289–317. 
28. Sjostrom J. How apoptosis is regulated, and what goes wrong in cancer. BMJ 2001; 322:1538–1539. 
29. Park M-T and Lee S-J. Cell cycle and cancer. J Biochem Mol Biol 2003; 36:60–65. 
30. Willis N and Rhind N. Regulation of DNA replication by the S-phase DNA damage checkpoint. Cell Div 2009; 4:13. 
31. Liu J-D, Wang Y-J, Chen C-H, Yu C-F, Chen L-C, Lin J-K, et al. Molecular mechanisms of G0/G1 cell-cycle arrest and apoptosis induced by terfenadine in human cancer cells. Mol Carcinog 2003; 37:39–50. 
32. Milkovic L, Cipak Gasparovic A, Cindric M, Mouthuy P-A, and Zarkovic N. Short Overview of ROS as Cell Function Regulators and Their Implications in Therapy Concepts. Cells 2019; 8:793. 
33. Schieber M and Chandel NS. ROS Function in Redox Signaling and Oxidative Stress. Curr Biol 2014; 24:R453–R462. 
34. Elmore S. Apoptosis: A Review of Programmed Cell Death. Toxicol Pathol 2007; 35:495–516. 
35. Naik E and Dixit VM. Mitochondrial reactive oxygen species drive proinflammatory cytokine production. J Exp Med 2011; 208:417–420. 
36. Iglesias-Guimarais V, Gil-Guiñon E, Sánchez-Osuna M, Casanelles E, García-Belinchón M, Comella JX, et al. Chromatin collapse during caspase-dependent apoptotic cell death requires DNA  fragmentation factor, 40-kDa subunit-/caspase-activated deoxyribonuclease-mediated 3’-OH single-strand DNA breaks. J Biol Chem 2013; 288:9200–9215. 
37. Semenov D V, Aronov PA, Kuligina E V, Potapenko MO,  Richter VA. Oligonucleosomal DNA fragmentation in MCF-7 cells undergoing palmitate-induced apoptosis. Biochem 2003; 68:1335–1341. 
38. Porter AG and Jänicke RU. Emerging roles of caspase-3 in apoptosis. Cell Death Differ 1999; 6:99–104. 
39. Wajant H, Pfizenmaier K, and Scheurich P. Tumor necrosis factor signaling. Cell Death Differ 2003; 10:45–65. 
40. Ma Y, Ren Y, Dai Z-J, Wu C-J, Ji Y-H, and Xu J. IL-6, IL-8 and TNF-α levels correlate with disease stage in breast cancer patients. Adv Clin Exp Med 2017; 26:421–426. 
41. Dennis KL, Blatner NR, Gounari F, and Khazaie K. Current status of interleukin-10 and regulatory T-cells in cancer. Curr Opin Oncol 2013; 25:637–645. 
42. Barathan M, Mariappan V, Shankar EM, Abdullah BJ, Goh KL, and Vadivelu J. Hypericin-photodynamic therapy leads to interleukin-6 secretion by HepG2 cells and their apoptosis via recruitment of BH3 interacting-domain death agonist and caspases. Cell Death Dis 2013; 4:1–10. 
43. Zhou J, Wang G, Chen Y, Wang H, Hua Y, and Cai Z. Immunogenic cell death in cancer therapy: Present and emerging inducers. J Cell Mol Med 2019; 23:4854–4865. 
44. Rapoport B and Anderson R. Realizing the clinical potential of immunogenic cell death in cancer chemotherapy and radiotherapy. Int J Mol Sci 2019; 20:959. 
45. Bose S, Panda AK, Mukherjee S, and Sa G. Curcumin and tumor immune-editing: resurrecting the immune system. Cell Div 2015; 10:6-19. 
46. Huang F-Y, Lei J, Sun Y, Yan F, Chen B, Zhang L, et al. Induction of enhanced immunogenic cell death through ultrasound-controlled release of doxorubicin by liposome-microbubble complexes. Oncoimmunology 2018; 7:e1446720.