DOI QR코드

DOI QR Code

Combination treatment with 2-methoxyestradiol overcomes bortezomib resistance of multiple myeloma cells

  • Song, In-Sung (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Jeong, Yu Jeong (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Jeong, Seung Hun (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Heo, Hye Jin (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Kim, Hyoung Kyu (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Lee, Sung Ryul (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Ko, Tae Hee (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Youm, Jae Boum (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Kim, Nari (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Ko, Kyung Soo (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Rhee, Byoung Doo (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University) ;
  • Han, Jin (National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University)
  • Published : 2013.10.31

Abstract

Bortezomib is a proteasome inhibitor used for the treatment of relapsed/refractory multiple myeloma (MM). However, intrinsic and acquired resistance to bortezomib has already been observed in MM patients. In a previous report, we demonstrated that changes in the expression of mitochondrial genes lead to changes in mitochondrial activity and bortezomib susceptibility or resistance, and their combined effects contribute to the differential sensitivity or resistance of MM cells to bortezomib. Here we report that the combination treatment of bortezomib and 2-methoxyestradiol (2ME), a natural estrogen metabolite, induces mitochondria-mediated apoptotic cell death of bortezomib-resistant MM KMS20 cells via mitochondrial reactive oxygen species (ROS) overproduction. Bortezomib plus 2ME treatment induces a higher level of cell death compared with treatment with bortezomib alone and increases mitochondrial ROS and $Ca^{2+}$ levels in KMS20 cells. Pretreatment with the antioxidant N-acetyl-L-cysteine scavenges mitochondrial ROS and decreases cell death after treatment with bortezomib plus 2ME in KMS20 cells. Moreover, we observed that treatment with bortezomib plus 2ME maintains the activation of c-Jun N-terminal kinase (JNK) and mitogen-activated protein kinase kinase kinase 4/7 (MKK4/7). Collectively, combination treatment with bortezomib and 2ME induces cell death via JNK-MKK4/7 activation by overproduction of mitochondrial ROS. Therefore, combination therapy with specific mitochondrial-targeting drugs may prove useful to the development of novel strategies for the treatment of bortezomib-resistant MM patients.

Keywords

References

  1. Kyle RA, Rajkumar SV. Multiple myeloma. N Engl J Med 2004; 351: 1860-1873. https://doi.org/10.1056/NEJMra041875
  2. Rajkumar SV, Richardson PG, Hideshima T, Anderson KC. Proteasome inhibition as a novel therapeutic target in human cancer. J Clin Oncol 2005; 23: 630-639. https://doi.org/10.1200/JCO.2005.11.030
  3. Chauhan D, Li G, Podar K, Hideshima T, Mitsiades C, Schlossman R et al. Targeting mitochondria to overcome conventional and bortezomib/proteasome inhibitor PS-341 resistance in multiple myeloma (MM) cells. Blood 2004; 104: 2458-2466. https://doi.org/10.1182/blood-2004-02-0547
  4. Caldera H, Giralt S. Stem cell transplantation for multiple myeloma: current status and future directions. Curr Hematol Rep 2004; 3: 249-256.
  5. Richardson PG, Barlogie B, Berenson J, Singhal S, Jagannath S, Irwin D et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med 2003; 348: 2609-2617. https://doi.org/10.1056/NEJMoa030288
  6. Ocio EM, Mateos MV, Maiso P, Pandiella A, San-Miguel JF. New drugs in multiple myeloma: mechanisms of action and phase I/II clinical findings. Lancet Oncol 2008; 9: 1157-1165. https://doi.org/10.1016/S1470-2045(08)70304-8
  7. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000; 100: 57-70. https://doi.org/10.1016/S0092-8674(00)81683-9
  8. Song IS, Kim HK, Lee SR, Jeong SH, Kim N, Ko KS et al. Mitochondrial modulation decreases the bortezomib-resistance in multiple myeloma cells. Int J Cancer 2013; 133: 1357-1367. https://doi.org/10.1002/ijc.28149
  9. Seegers JC, Aveling ML, Van Aswegen CH, Cross M, Koch F, Joubert WS. The cytotoxic effects of estradiol-17 beta, catecholestradiols and methoxyestradiols on dividing MCF-7 and HeLa cells. J Steroid Biochem 1989; 32: 797-809. https://doi.org/10.1016/0022-4731(89)90455-X
  10. Sutherland TE, Anderson RL, Hughes RA, Altmann E, Schuliga M, Ziogas J et al. 2-Methoxyestradiol-a unique blend of activities generating a new class of anti-tumour/anti-inflammatory agents. Drug Discov Today 2007; 12: 577-584. https://doi.org/10.1016/j.drudis.2007.05.005
  11. Attalla H, Westberg JA, Andersson LC, Adlercreutz H, Makela TP. 2-Methoxyestradiol-induced phosphorylation of Bcl-2: uncoupling from JNK/SAPK activation. Biochem Biophys Res Commun 1998; 247: 616-619. https://doi.org/10.1006/bbrc.1998.8870
  12. Hamel E, Lin CM, Flynn E, D'Amato RJ. Interactions of 2-methoxyestradiol, an endogenous mammalian metabolite, with unpolymerized tubulin and with tubulin polymers. Biochemistry 1996; 35: 1304-1310. https://doi.org/10.1021/bi951559s
  13. Huang P, Feng L, Oldham EA, Keating MJ, Plunkett W. Superoxide dismutase as a target for the selective killing of cancer cells. Nature 2000; 407: 390-395. https://doi.org/10.1038/35030140
  14. Kumar AP, Garcia GE, Slaga TJ. 2-methoxyestradiol blocks cell-cycle progression at G(2)/M phase and inhibits growth of human prostate cancer cells. Mol Carcinog 2001; 31: 111-124. https://doi.org/10.1002/mc.1046
  15. Lin HL, Liu TY, Chau GY, Lui WY, Chi CW. Comparison of 2-methoxyestradiol-induced, docetaxel- induced, and paclitaxel-induced apoptosis in hepatoma cells and its correlation with reactive oxygen species. Cancer 2000; 89: 983-994. https://doi.org/10.1002/1097-0142(20000901)89:5<983::AID-CNCR7>3.0.CO;2-G
  16. Song IS, Kim SU, Oh NS, Kim J, Yu DY, Huang SM et al. Peroxiredoxin I contributes to TRAIL resistance through suppression of redox-sensitive caspase activation in human hepatoma cells. Carcinogenesis 2009; 30: 1106-1114. https://doi.org/10.1093/carcin/bgp104
  17. Mukhopadhyay T, Roth JA. Induction of apoptosis in human lung cancer cells after wild-type p53 activation by methoxyestradiol. Oncogene 1997; 14: 379-384. https://doi.org/10.1038/sj.onc.1200835
  18. Qadan LR, Perez-Stable CM, Anderson C, D'Ippolito G, Herron A, Howard GA et al. 2-Methoxyestradiol induces G2/M arrest and apoptosis in prostate cancer. Biochem Biophys Res Commun 2001; 285: 1259-1266. https://doi.org/10.1006/bbrc.2001.5320
  19. Qanungo S, Basu A, Das M, Haldar S. 2-Methoxyestradiol induces mitochondria dependent apoptotic signaling in pancreatic cancer cells. Oncogene 2002; 21: 4149-4157. https://doi.org/10.1038/sj.onc.1205508
  20. Song IS, Jun SY, Na HJ, Kim HT, Jung SY, Ha GH et al. Inhibition of MKK7-JNK by the TOR signaling pathway regulator-like protein contributes to resistance of HCC cells to TRAIL-induced apoptosis. Gastroenterology 2012; 143: 1341-1351. https://doi.org/10.1053/j.gastro.2012.07.103
  21. Ventura JJ, Cogswell P, Flavell RA, Baldwin AS Jr., Davis RJ. JNK potentiates TNF-stimulated necrosis by increasing the production of cytotoxic reactive oxygen species. Genes Dev 2004; 18: 2905-2915. https://doi.org/10.1101/gad.1223004
  22. Derijard B, Raingeaud J, Barrett T, Wu IH, Han J, Ulevitch RJ et al. Independent human MAP-kinase signal transduction pathways defined by MEK and MKK isoforms. Science 1995; 267: 682-685. https://doi.org/10.1126/science.7839144
  23. Richardson PG, Hideshima T, Mitsiades C, Anderson KC. The emerging role of novel therapies for the treatment of relapsed myeloma. J Natl Compr Canc Netw 2007; 5: 149-162. https://doi.org/10.6004/jnccn.2007.0015
  24. Lonial S, Waller EK, Richardson PG, Jagannath S, Orlowski RZ, Giver CR et al. Risk factors and kinetics of thrombocytopenia associated with bortezomib for relapsed, refractory multiple myeloma. Blood 2005; 106: 3777-3784. https://doi.org/10.1182/blood-2005-03-1173
  25. Djavaheri-Mergny M, Wietzerbin J, Besancon F. 2-Methoxyestradiol induces apoptosis in Ewing sarcoma cells through mitochondrial hydrogen peroxide production. Oncogene 2003; 22: 2558-2567. https://doi.org/10.1038/sj.onc.1206356
  26. Benhar M, Engelberg D, ROS Levitzki A. Stress-activated kinases and stress signaling in cancer. EMBO Rep 2002; 3: 420-425. https://doi.org/10.1093/embo-reports/kvf094
  27. Aoki H, Kang PM, Hampe J, Yoshimura K, Noma T, Matsuzaki M et al. Direct activation of mitochondrial apoptosis machinery by c-Jun N-terminal kinase in adult cardiac myocytes. J Biol Chem 2002; 277: 10244-10250. https://doi.org/10.1074/jbc.M112355200
  28. Hashimoto M, Hsu LJ, Rockenstein E, Takenouchi T, Mallory M, Masliah E. alpha-Synuclein protects against oxidative stress via inactivation of the c-Jun N-terminal kinase stress-signaling pathway in neuronal cells. J Biol Chem 2002; 277: 11465-11472. https://doi.org/10.1074/jbc.M111428200
  29. Chen YR, Wang X, Templeton D, Davis RJ, Tan TH. The role of c-Jun N-terminal kinase (JNK) in apoptosis induced by ultraviolet C and gamma radiation. Duration of JNK activation may determine cell death and proliferation. J Biol Chem 1996; 271: 31929-31936. https://doi.org/10.1074/jbc.271.50.31929
  30. Guo YL, Baysal K, Kang B, Yang LJ, Williamson JR. Correlation between sustained c-Jun N-terminal protein kinase activation and apoptosis induced by tumor necrosis factor-alpha in rat mesangial cells. J Biol Chem 1998; 273: 4027-4034. https://doi.org/10.1074/jbc.273.7.4027
  31. Roulston A, Reinhard C, Amiri P, Williams LT. Early activation of c-Jun N-terminal kinase and p38 kinase regulate cell survival in response to tumor necrosis factor alpha. J Biol Chem 1998; 273: 10232-10239. https://doi.org/10.1074/jbc.273.17.10232
  32. Martindale JL, Holbrook NJ. Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol 2002; 192: 1-15. https://doi.org/10.1002/jcp.10119
  33. Sano M, Fukuda K, Sato T, Kawaguchi H, Suematsu M, Matsuda S et al. ERK and p38 MAPK, but not NF-kappaB, are critically involved in reactive oxygen species-mediated induction of IL-6 by angiotensin II in cardiac fibroblasts. Circ Res 2001; 89: 661-669. https://doi.org/10.1161/hh2001.098873
  34. Song IS, Kim HK, Jeong SH, Lee SR, Kim N, Rhee BD et al. Mitochondrial peroxiredoxin III is a potential target for cancer therapy. Int J Mol Sci 2011; 12: 7163-7185. https://doi.org/10.3390/ijms12107163
  35. Guyton KZ, Liu Y, Gorospe M, Xu Q, Holbrook NJ. Activation of mitogenactivated protein kinase by $H_2O_2$. Role in cell survival following oxidant injury. J Biol Chem 1996; 271: 4138-4142. https://doi.org/10.1074/jbc.271.8.4138
  36. Chen K, Vita JA, Berk BC, Keaney JF Jr. c-Jun N-terminal kinase activation by hydrogen peroxide in endothelial cells involves SRC-dependent epidermal growth factor receptor transactivation. J Biol Chem 2001; 276: 16045-16050. https://doi.org/10.1074/jbc.M011766200
  37. Davis RJ. Signal transduction by the JNK group of MAP kinases. Cell 2000; 103: 239-252. https://doi.org/10.1016/S0092-8674(00)00116-1
  38. Ventura JJ, Hubner A, Zhang C, Flavell RA, Shokat KM, Davis RJ. Chemical genetic analysis of the time course of signal transduction by JNK. Mol Cell 2006; 21: 701-710. https://doi.org/10.1016/j.molcel.2006.01.018
  39. Papa S, Zazzeroni F, Bubici C, Jayawardena S, Alvarez K, Matsuda S et al. Gadd45 beta mediates the NF-kappa B suppression of JNK signalling by targeting MKK7/JNKK2. Nat Cell Biol 2004; 6: 146-153. https://doi.org/10.1038/ncb1093
  40. Sakon S, Xue X, Takekawa M, Sasazuki T, Okazaki T, Kojima Y et al. NF-kappaB inhibits TNF-induced accumulation of ROS that mediate prolonged MAPK activation and necrotic cell death. EMBO J 2003; 22: 3898-3909. https://doi.org/10.1093/emboj/cdg379
  41. Kang SW, Chang TS, Lee TH, Kim ES, Yu DY, Rhee SG. Cytosolic peroxiredoxin attenuates the activation of Jnk and p38 but potentiates that of Erk in Hela cells stimulated with tumor necrosis factor-alpha. J Biol Chem 2004; 279: 2535-2543. https://doi.org/10.1074/jbc.M307698200
  42. Gotoh Y, Cooper JA. Reactive oxygen species- and dimerizationinduced activation of apoptosis signal-regulating kinase 1 in tumor necrosis factor-alpha signal transduction. J Biol Chem 1998; 273: 17477-17482. https://doi.org/10.1074/jbc.273.28.17477
  43. Saitoh M, Nishitoh H, Fujii M, Takeda K, Tobiume K, Sawada Y et al. Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. EMBO J 1998; 17: 2596-2606. https://doi.org/10.1093/emboj/17.9.2596

Cited by

  1. Inhibition of PGC-1α after chemotherapy-mediated insult confines multiple myeloma cell survival by affecting ROS accumulation vol.33, pp.2, 2013, https://doi.org/10.3892/or.2014.3635
  2. Dissolution Chemistry and Biocompatibility of Silicon- and Germanium-Based Semiconductors for Transient Electronics vol.7, pp.17, 2013, https://doi.org/10.1021/acsami.5b02526
  3. DNA strand breaks induced by nuclear hijacking of neuronal NOS as an anti-cancer effect of 2-methoxyestradiol vol.6, pp.17, 2013, https://doi.org/10.18632/oncotarget.3913
  4. KSP inhibitor SB743921 induces death of multiple myeloma cells via inhibition of the NF-κB signaling pathway vol.48, pp.10, 2015, https://doi.org/10.5483/bmbrep.2015.48.10.015
  5. Soluble and Cell–Cell-Mediated Drivers of Proteasome Inhibitor Resistance in Multiple Myeloma vol.9, pp.None, 2013, https://doi.org/10.3389/fendo.2018.00218
  6. The Influence of Metabolism on Drug Response in Cancer vol.8, pp.None, 2013, https://doi.org/10.3389/fonc.2018.00500
  7. Promising Anti-Mitochondrial Agents for Overcoming Acquired Drug Resistance in Multiple Myeloma vol.10, pp.2, 2013, https://doi.org/10.3390/cells10020439