DOI QR코드

DOI QR Code

USP15 inhibits multiple myeloma cell apoptosis through activating a feedback loop with the transcription factor NF-κBp65

  • Zhou, Lili (Shanghai Jiahui International Hospital Cancer Center) ;
  • Jiang, Hua (Department of Hematology, Shanghai Changzheng Hospital, Second Military Medical University) ;
  • Du, Juan (Department of Hematology, Shanghai Changzheng Hospital, Second Military Medical University) ;
  • Li, Lu (Department of Hematology, Shanghai Changzheng Hospital, Second Military Medical University) ;
  • Li, Rong (Department of Hematology, Shanghai Changzheng Hospital, Second Military Medical University) ;
  • Lu, Jing (Department of Hematology, Shanghai Changzheng Hospital, Second Military Medical University) ;
  • Fu, Weijun (Department of Hematology, Shanghai Changzheng Hospital, Second Military Medical University) ;
  • Hou, Jian (Department of Hematology, Renji Hospital affiliated to Shanghai Jiaotong University School of Medicine)
  • Received : 2018.05.16
  • Accepted : 2018.09.04
  • Published : 2018.11.30

Abstract

USP15 has been shown to stabilize transcription factors, to be amplified in many cancers and to mediate cancer cell survival. However, the underlying mechanism by which USP15 regulates multiple myeloma (MM) cell proliferation and apoptosis has not been established. Here, our results showed that USP15 mRNA expression was upregulated in MM patients. USP15 silencing induced MM cell proliferation inhibition, apoptosis, and the expression of nuclear and cytoplasmic NF-${\kappa}Bp65$, while USP15 overexpression exhibited an inverse effect. Moreover, in vivo experiments indicated that USP15 silencing inhibited MM tumor growth and NF-${\kappa}Bp65$ expression. PDTC treatment significantly inhibited USP15 overexpression-induced cell proliferation, apoptosis inhibition, and NF-${\kappa}Bp65$ expression. USP15 overexpression promoted NF-${\kappa}Bp65$ expression through inhibition of its ubiquitination, whereas NF-${\kappa}Bp65$ promoted USP15 expression as a positive regulator. Taken together, the USP15-NF-${\kappa}Bp65$ loop is involved in MM tumorigenesis and may be a potential therapeutic target for MM.

Keywords

Acknowledgement

Supported by : National Natural Science Foundation of China

References

  1. Viziteu, E. et al. RECQ1 helicase is involved in replication stress survival and drug resistance in multiple myeloma. Leukemia 31, 2104-2113 (2017). https://doi.org/10.1038/leu.2017.54
  2. Fakhri, B., Fiala, M. A., Tuchman, S. A. & Wildes, T. M. Undertreatment of older patients with newly diagnosed multiple myeloma in the era of novel therapies. Clin. Lymphoma Myeloma Leuk. 18, 219-224 (2018). https://doi.org/10.1016/j.clml.2018.01.005
  3. Kazandjian, D. Multiple myeloma epidemiology and survival: a unique malignancy. Semin. Oncol. 43, 676-681 (2016). https://doi.org/10.1053/j.seminoncol.2016.11.004
  4. Fulciniti, M. et al. Functional role and therapeutic targeting of p21-activated kinase 4 in multiple myeloma. Blood 129, 2233-2245 (2017). https://doi.org/10.1182/blood-2016-06-724831
  5. Neemat, K., Rania, K., Tarek, M. & Hamdy, A. A. Effect of 13q deletion on IL-6 production in patients with multiple myeloma: a hypothesis may hold true. Clin. Lab. 60, 1393-1399 (2014).
  6. Iida, S. et al. Bortezomib plus dexamethasone versus thalidomide plus dexamethasone for relapsed or refractory multiple myeloma. Cancer Sci. 109, 1552-1561 (2018). https://doi.org/10.1111/cas.13550
  7. Pulte, D., Gondos, A. & Brenner, H. Improvement in survival of older adults with multiple myeloma: results of an updated period analysis of SEER data. Oncologist 16, 1600-1603 (2011). https://doi.org/10.1634/theoncologist.2011-0229
  8. Chen, Z. et al. MicroRNA-300 regulates the ubiquitination of PTEN through the CRL4B(DCAF13) E3 ligase in osteosarcoma cells. Mol. Ther. Nucleic Acids 10, 254-268 (2018). https://doi.org/10.1016/j.omtn.2017.12.010
  9. Wang, M. et al. SYNJ2BP promotes the degradation of PTEN through the lysosome-pathway and enhances breast tumor metastasis via PI3K/AKT/ SNAI1 signaling. Oncotarget 8, 89692-89706 (2017).
  10. Torre, S. & Polyak, M. J. USP15 regulates type I interferon response and is required for pathogenesis of neuroinflammation. Nat. Immunol. 18, 54-63 (2017). https://doi.org/10.1038/ni.3581
  11. Padmanabhan, A., Candelaria, N. & Wong, K. K. USP15-dependent lysosomal pathway controls p53-R175H turnover in ovarian cancer cells. Nat. Commun. 9, 1270 (2018). https://doi.org/10.1038/s41467-018-03599-w
  12. Liu, W. T. et al. TGF-beta upregulates the translation of USP15 via the PI3K/AKT pathway to promote p53 stability. Oncogene 36, 2715-2723 (2017). https://doi.org/10.1038/onc.2016.424
  13. Schweitzer, K., Bozko, P. M., Dubiel, W. & Naumann, M. CSN controls NFkappaB by deubiquitinylation of IkappaBalpha. EMBO J. 26, 1532-1541 (2007). https://doi.org/10.1038/sj.emboj.7601600
  14. Borner, C. & Kraus, J. Inhibition of NF-kappaB by opioids in T cells. J. Immunol. 191, 4640-4647 (2013). https://doi.org/10.4049/jimmunol.1300320
  15. Kim, J. et al. Regulation of STAT3 and NF-kappaB activations by S-nitrosylation in multiple myeloma. Free Radic. Biol. Med. 106, 245-253 (2017). https://doi.org/10.1016/j.freeradbiomed.2017.02.039
  16. Milano, A., Perri, F. & Caponigro, F. The ubiquitin-proteasome system as a molecular target in solid tumors: an update on bortezomib. Onco. Targets Ther. 2, 171-178 (2009).
  17. Meshram, S. N. et al. FBXO32 activates NF-kappaB through IkappaBalpha degradation in inflammatory and genotoxic stress. Int. J. Biochem. Cell Biol. 92, 134-140 (2017). https://doi.org/10.1016/j.biocel.2017.09.021
  18. Hosono, M. et al. Interaction of Ca(2+)-dependent activator protein for secretion 1 (CAPS1) with septin family proteins in mouse brain. Neurosci. Lett. 617, 232-235 (2016). https://doi.org/10.1016/j.neulet.2016.02.035
  19. Bar-Natan, M. et al. Bone marrow stroma protects myeloma cells from cytotoxic damage via induction of the oncoprotein MUC1. Br. J. Haematol. 176, 929-938 (2017). https://doi.org/10.1111/bjh.14493
  20. Zheng, Z. et al. Inhibition of thioredoxin activates mitophagy and overcomes adaptive bortezomib resistance in multiple myeloma. J. Hematol. Oncol. 11, 29 (2018). https://doi.org/10.1186/s13045-018-0575-7
  21. Guo, W. et al. Up-regulated deubiquitinase USP4 plays an oncogenic role in melanoma. J. Cell Mol. Med. 22, 2944-2954 (2018). https://doi.org/10.1111/jcmm.13603
  22. Peng, L., Hu, Y., Chen, D., Jiao, S. & Sun, S. Ubiquitin specific peptidase 21 regulates interleukin-8 expression, stem-cell like property of human renal cell carcinoma. Oncotarget 7, 42007-42016 (2016).
  23. Chen, X. et al. Ubiquitin-specific protease 14 regulates cell proliferation and apoptosis in oral squamous cell carcinoma. Int. J. Biochem. Cell Biol. 79, 350-359 (2016). https://doi.org/10.1016/j.biocel.2016.08.038
  24. Zou, Q. et al. USP15 stabilizes MDM2 to mediate cancer-cell survival and inhibit antitumor T cell responses. Nat. Immunol. 15, 562-570 (2014).
  25. Eichhorn, P. J. et al. USP15 stabilizes TGF-beta receptor I and promotes oncogenesis through the activation of TGF-beta signaling in glioblastoma. Nat. Med. 18, 429-435 (2012). https://doi.org/10.1038/nm.2619
  26. Mialki, R. K., Zhao, J., Wei, J., Mallampalli, D. F. & Zhao, Y. Overexpression of USP14 protease reduces I-kappaB protein levels and increases cytokine release in lung epithelial cells. J. Biol. Chem. 288, 15437-15441 (2013). https://doi.org/10.1074/jbc.C112.446682
  27. Xiao, N. et al. Ubiquitin-specific protease 4 (USP4) targets TRAF2 and TRAF6 for deubiquitination and inhibits TNFalpha-induced cancer cell migration. Biochem. J. 441, 979-986 (2012). https://doi.org/10.1042/BJ20111358
  28. Liu, S., Zheng, L. L., Zhu, Y. M. & Shen, H. J. Knockdown of REGgamma inhibits the proliferation and migration and promotes the apoptosis of multiple myeloma cells by downregulating NF-kappaB signal pathway. Hematology 23, 277-283 (2018). https://doi.org/10.1080/10245332.2017.1385194
  29. Sun, C. et al. Resveratrol downregulates the constitutional activation of nuclear factor-kappaB in multiple myeloma cells, leading to suppression of proliferation and invasion, arrest of cell cycle, and induction of apoptosis. Cancer Genet. Cytogenet. 165, 9-19 (2006). https://doi.org/10.1016/j.cancergencyto.2005.06.016
  30. Chen, G. et al. An anti-leishmanial thiadiazine agent induces multiple myeloma cell apoptosis by suppressing the nuclear factor kappaB signalling pathway. Br. J. Cancer 110, 63-70 (2014). https://doi.org/10.1038/bjc.2013.711
  31. Takeda, T. et al. Mangiferin enhances the sensitivity of human multiple myeloma cells to anticancer drugs through suppression of the nuclear factor kappaB pathway. Int. J. Oncol. 48, 2704-2712 (2016). https://doi.org/10.3892/ijo.2016.3470

Cited by

  1. DUBbing Down Translation: The Functional Interaction of Deubiquitinases with the Translational Machinery vol.18, pp.9, 2018, https://doi.org/10.1158/1535-7163.mct-19-0307
  2. De-regulated STAT5A/miR-202-5p/USP15/Caspase-6 regulatory axis suppresses CML cell apoptosis and contributes to Imatinib resistance vol.39, pp.1, 2018, https://doi.org/10.1186/s13046-019-1502-7
  3. In vitro and in vivo efficacy of the novel oral proteasome inhibitor NNU546 in multiple myeloma vol.12, pp.22, 2020, https://doi.org/10.18632/aging.104023
  4. Current Methods of Post-Translational Modification Analysis and Their Applications in Blood Cancers vol.13, pp.8, 2018, https://doi.org/10.3390/cancers13081930
  5. Deubiquitinases: Modulators of Different Types of Regulated Cell Death vol.22, pp.9, 2018, https://doi.org/10.3390/ijms22094352
  6. The Multifaceted Roles of USP15 in Signal Transduction vol.22, pp.9, 2018, https://doi.org/10.3390/ijms22094728
  7. Deubiquitinases in hematological malignancies vol.9, pp.1, 2021, https://doi.org/10.1186/s40364-021-00320-w