Skip to main content
Top
Published in: Medical Oncology 11/2019

01-11-2019 | Ovarian Cancer | Original Paper

Ubiquitin-specific peptidase 39 regulates the process of proliferation and migration of human ovarian cancer via p53/p21 pathway and EMT

Authors: Congcong Yan, Jiahui Yuan, Jiajia Xu, Gongye Zhang, Xiaomei Li, Bing Zhang, Tianhui Hu, Xiaohua Huang, Yubin Mao, Gang Song

Published in: Medical Oncology | Issue 11/2019

Login to get access

Abstract

Ovarian cancer is one of the most lethal gynecological cancers; owning to its late detection and chemoresistance, understanding the pathogenesis of this malignant tumor is much critical. Previous studies have reported that ubiquitin-specific peptidase 39 (USP39) is generally overexpressed in a variety of cancers, including hepatocellular carcinoma, gastric cancer and so forth. Furthermore, USP39 is proved to be associated with the proliferation of malignant tumors. However, the function and mechanism of USP39 in ovarian cancer have not been elucidated. In the present study, we observed that USP39 was frequently overexpressed in human ovarian cancer and was highly correlated with TNM stage. Suppression of USP39 markedly inhibited the growth and migration of ovarian cancer cell lines HO-8910 and SKOV3 and induced cell cycle G2/M arrest. Moreover, knockdown of USP39 inhibited ovarian tumor growth in a xenograft model. In addition, our findings indicated that cell cycle arrest induced by USP39 knockdown might be involved in p53/p21 signaling pathway. Furthermore, we found that the depletion of USP39 inhibited the migration of ovarian cancer cells via blocking epithelial–mesenchymal transition. Taken together, these results suggest that USP39 may play vital roles in the genesis and progression and may serve as a potential biomarker for diagnosis and therapeutic target of ovarian cancer.
Literature
1.
go back to reference Torre LA, Trabert B, DeSantis CE, et al. Ovarian cancer statistics, 2018. CA Cancer J Clin. 2018;68:284–96.CrossRef Torre LA, Trabert B, DeSantis CE, et al. Ovarian cancer statistics, 2018. CA Cancer J Clin. 2018;68:284–96.CrossRef
2.
go back to reference Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30.CrossRef Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30.CrossRef
3.
go back to reference Kuan AS, Teng CJ, Wu HH, et al. Risk of ischemic stroke in patients with ovarian cancer: a nationwide population-based study. BMC Med. 2014;12:53.CrossRef Kuan AS, Teng CJ, Wu HH, et al. Risk of ischemic stroke in patients with ovarian cancer: a nationwide population-based study. BMC Med. 2014;12:53.CrossRef
4.
go back to reference Fraile JM, Quesada V, Rodriguez D, Freije JM, Lopez-Otin C. Deubiquitinases in cancer: new functions and therapeutic options. Oncogene. 2012;31:2373–88.CrossRef Fraile JM, Quesada V, Rodriguez D, Freije JM, Lopez-Otin C. Deubiquitinases in cancer: new functions and therapeutic options. Oncogene. 2012;31:2373–88.CrossRef
5.
go back to reference van Leuken RJ, Luna-Vargas MP, Sixma TK, Wolthuis RM, Medema RH. Usp39 is essential for mitotic spindle checkpoint integrity and controls mRNA-levels of aurora B. Cell Cycle. 2008;7:2710–9.CrossRef van Leuken RJ, Luna-Vargas MP, Sixma TK, Wolthuis RM, Medema RH. Usp39 is essential for mitotic spindle checkpoint integrity and controls mRNA-levels of aurora B. Cell Cycle. 2008;7:2710–9.CrossRef
6.
go back to reference Hadjivassiliou H, Rosenberg OS, Guthrie C. The crystal structure of S. cerevisiae Sad1, a catalytically inactive deubiquitinase that is broadly required for pre-mRNA splicing. RNA. 2014;20:656–69.CrossRef Hadjivassiliou H, Rosenberg OS, Guthrie C. The crystal structure of S. cerevisiae Sad1, a catalytically inactive deubiquitinase that is broadly required for pre-mRNA splicing. RNA. 2014;20:656–69.CrossRef
7.
go back to reference Makarova OV, Makarov EM, Luhrmann R. The 65 and 110 kDa SR-related proteins of the U4/U6.U5 tri-snRNP are essential for the assembly of mature spliceosomes. EMBO J. 2001;20:2553–63.CrossRef Makarova OV, Makarov EM, Luhrmann R. The 65 and 110 kDa SR-related proteins of the U4/U6.U5 tri-snRNP are essential for the assembly of mature spliceosomes. EMBO J. 2001;20:2553–63.CrossRef
8.
go back to reference He X, Zhang P. Serine/arginine-rich splicing factor 3 (SRSF3) regulates homologous recombination-mediated DNA repair. Mol Cancer. 2015;14:158.CrossRef He X, Zhang P. Serine/arginine-rich splicing factor 3 (SRSF3) regulates homologous recombination-mediated DNA repair. Mol Cancer. 2015;14:158.CrossRef
9.
go back to reference Patwardhan GA, Hosain SB, Liu DX, et al. Ceramide modulates pre-mRNA splicing to restore the expression of wild-type tumor suppressor p53 in deletion-mutant cancer cells. Biochem Biophys Acta. 1841;1571–1580:2014. Patwardhan GA, Hosain SB, Liu DX, et al. Ceramide modulates pre-mRNA splicing to restore the expression of wild-type tumor suppressor p53 in deletion-mutant cancer cells. Biochem Biophys Acta. 1841;1571–1580:2014.
10.
go back to reference Gautrey HL, Tyson-Capper AJ. Regulation of Mcl-1 by SRSF1 and SRSF5 in cancer cells. PLoS ONE. 2012;7:e51497.CrossRef Gautrey HL, Tyson-Capper AJ. Regulation of Mcl-1 by SRSF1 and SRSF5 in cancer cells. PLoS ONE. 2012;7:e51497.CrossRef
11.
go back to reference Wang H, Ji X, Liu X, et al. Lentivirus-mediated inhibition of USP39 suppresses the growth of breast cancer cells in vitro. Oncol Rep. 2013;30:2871–7.CrossRef Wang H, Ji X, Liu X, et al. Lentivirus-mediated inhibition of USP39 suppresses the growth of breast cancer cells in vitro. Oncol Rep. 2013;30:2871–7.CrossRef
12.
go back to reference Pan Z, Pan H, Zhang J, et al. Lentivirus mediated silencing of ubiquitin specific peptidase 39 inhibits cell proliferation of human hepatocellular carcinoma cells in vitro. Biol Res. 2015;48:18.CrossRef Pan Z, Pan H, Zhang J, et al. Lentivirus mediated silencing of ubiquitin specific peptidase 39 inhibits cell proliferation of human hepatocellular carcinoma cells in vitro. Biol Res. 2015;48:18.CrossRef
13.
go back to reference An Y, Yang S, Guo K, Ma B, Wang Y. Reduced USP39 expression inhibits malignant proliferation of medullary thyroid carcinoma in vitro. World J Surg Oncol. 2015;13:255.CrossRef An Y, Yang S, Guo K, Ma B, Wang Y. Reduced USP39 expression inhibits malignant proliferation of medullary thyroid carcinoma in vitro. World J Surg Oncol. 2015;13:255.CrossRef
14.
go back to reference Wen D, Xu Z, Xia L, et al. Important role of SUMOylation of Spliceosome factors in prostate cancer cells. J Proteome Res. 2014;13:3571–82.CrossRef Wen D, Xu Z, Xia L, et al. Important role of SUMOylation of Spliceosome factors in prostate cancer cells. J Proteome Res. 2014;13:3571–82.CrossRef
15.
go back to reference Tong Y, Zhang G, Li Y, et al. Corilagin inhibits breast cancer growth via reactive oxygen species-dependent apoptosis and autophagy. J Cell Mol Med. 2018;22:3795–807.CrossRef Tong Y, Zhang G, Li Y, et al. Corilagin inhibits breast cancer growth via reactive oxygen species-dependent apoptosis and autophagy. J Cell Mol Med. 2018;22:3795–807.CrossRef
16.
go back to reference Stegmeier F, Rape M, Draviam VM, et al. Anaphase initiation is regulated by antagonistic ubiquitination and deubiquitination activities. Nature. 2007;446:876–81.CrossRef Stegmeier F, Rape M, Draviam VM, et al. Anaphase initiation is regulated by antagonistic ubiquitination and deubiquitination activities. Nature. 2007;446:876–81.CrossRef
17.
go back to reference Chen FZ, Zhao XK. Ubiquitin-proteasome pathway and prostate cancer. Onkologie. 2013;36:592–6.CrossRef Chen FZ, Zhao XK. Ubiquitin-proteasome pathway and prostate cancer. Onkologie. 2013;36:592–6.CrossRef
18.
go back to reference Yuan X, Sun X, Shi X, et al. USP39 promotes colorectal cancer growth and metastasis through the Wnt/beta-catenin pathway. Oncol Rep. 2017;37:2398–404.CrossRef Yuan X, Sun X, Shi X, et al. USP39 promotes colorectal cancer growth and metastasis through the Wnt/beta-catenin pathway. Oncol Rep. 2017;37:2398–404.CrossRef
19.
go back to reference Stevenson LF, Sparks A, Allende-Vega N, Xirodimas DP, Lane DP, Saville MK. The deubiquitinating enzyme USP2a regulates the p53 pathway by targeting Mdm2. EMBO J. 2007;26:976–86.CrossRef Stevenson LF, Sparks A, Allende-Vega N, Xirodimas DP, Lane DP, Saville MK. The deubiquitinating enzyme USP2a regulates the p53 pathway by targeting Mdm2. EMBO J. 2007;26:976–86.CrossRef
20.
go back to reference Zhang L, Zhou FF, Drabsch Y, et al. USP4 is regulated by AKT phosphorylation and directly deubiquitylates TGF-beta type I receptor. Nat Cell Biol. 2012;14:717–26.CrossRef Zhang L, Zhou FF, Drabsch Y, et al. USP4 is regulated by AKT phosphorylation and directly deubiquitylates TGF-beta type I receptor. Nat Cell Biol. 2012;14:717–26.CrossRef
21.
go back to reference Dayal S, Sparks A, Jacob J, Allende-Vega N, Lane DP, Saville MK. Suppression of the deubiquitinating enzyme USP5 causes the accumulation of unanchored polyubiquitin and the activation of p53. J Biol Chem. 2009;284:5030–41.CrossRef Dayal S, Sparks A, Jacob J, Allende-Vega N, Lane DP, Saville MK. Suppression of the deubiquitinating enzyme USP5 causes the accumulation of unanchored polyubiquitin and the activation of p53. J Biol Chem. 2009;284:5030–41.CrossRef
22.
go back to reference Qian J, Pentz K, Zhu Q, et al. USP7 modulates UV-induced PCNA monoubiquitination by regulating DNA polymerase eta stability. Oncogene. 2015;34:4791–6.CrossRef Qian J, Pentz K, Zhu Q, et al. USP7 modulates UV-induced PCNA monoubiquitination by regulating DNA polymerase eta stability. Oncogene. 2015;34:4791–6.CrossRef
23.
go back to reference Liu J, Xia H, Kim M, et al. Beclin1 controls the levels of p53 by regulating the deubiquitination activity of USP10 and USP13. Cell. 2011;147:223–34.CrossRef Liu J, Xia H, Kim M, et al. Beclin1 controls the levels of p53 by regulating the deubiquitination activity of USP10 and USP13. Cell. 2011;147:223–34.CrossRef
24.
go back to reference Wang X, Yu Q, Huang L, Yu P. Lentivirus-mediated inhibition of USP39 suppresses the growth of gastric cancer cells via PARP activation. Mol Med Rep. 2016;14:301–6.CrossRef Wang X, Yu Q, Huang L, Yu P. Lentivirus-mediated inhibition of USP39 suppresses the growth of gastric cancer cells via PARP activation. Mol Med Rep. 2016;14:301–6.CrossRef
25.
go back to reference Naidu KA, Fang Q, Naidu KA, Cheng JQ, Nicosia SV, Coppola D. P53 enhances ascorbyl stearate-induced G2/M arrest of human ovarian cancer cells. Anticancer Res. 2007;27:3927–34.PubMed Naidu KA, Fang Q, Naidu KA, Cheng JQ, Nicosia SV, Coppola D. P53 enhances ascorbyl stearate-induced G2/M arrest of human ovarian cancer cells. Anticancer Res. 2007;27:3927–34.PubMed
26.
go back to reference Hnit SS, Xie C, Yao M, et al. p27(Kip1) signaling: transcriptional and post-translational regulation. Int J Biochem Cell Biol. 2015;68:9–14.CrossRef Hnit SS, Xie C, Yao M, et al. p27(Kip1) signaling: transcriptional and post-translational regulation. Int J Biochem Cell Biol. 2015;68:9–14.CrossRef
27.
go back to reference Horvath V, Soucek K, Svihalkova-Sindlerova L, et al. Different cell cycle modulation following treatment of human ovarian carcinoma cells with a new platinum(IV) complex vs cisplatin. Investig New Drugs. 2007;25:435–43.CrossRef Horvath V, Soucek K, Svihalkova-Sindlerova L, et al. Different cell cycle modulation following treatment of human ovarian carcinoma cells with a new platinum(IV) complex vs cisplatin. Investig New Drugs. 2007;25:435–43.CrossRef
28.
go back to reference Nurse P. Universal control mechanism regulating onset of M-phase. Nature. 1990;344:503–8.CrossRef Nurse P. Universal control mechanism regulating onset of M-phase. Nature. 1990;344:503–8.CrossRef
29.
go back to reference Choi EJ, Kim GH. Apigenin causes G(2)/M arrest associated with the modulation of p21(Cip1) and Cdc2 and activates p53-dependent apoptosis pathway in human breast cancer SK-BR-3 cells. J Nutr Biochem. 2009;20:285–90.CrossRef Choi EJ, Kim GH. Apigenin causes G(2)/M arrest associated with the modulation of p21(Cip1) and Cdc2 and activates p53-dependent apoptosis pathway in human breast cancer SK-BR-3 cells. J Nutr Biochem. 2009;20:285–90.CrossRef
30.
go back to reference Takai M, Terai Y, Kawaguchi H, et al. The EMT (epithelial-mesenchymal-transition)-related protein expression indicates the metastatic status and prognosis in patients with ovarian cancer. J Ovarian Res. 2014;7:76.CrossRef Takai M, Terai Y, Kawaguchi H, et al. The EMT (epithelial-mesenchymal-transition)-related protein expression indicates the metastatic status and prognosis in patients with ovarian cancer. J Ovarian Res. 2014;7:76.CrossRef
31.
go back to reference Mao Y, Xu J, Li Z, Zhang N, Yin H, Liu Z. The role of nuclear beta-catenin accumulation in the Twist2-induced ovarian cancer EMT. PLoS ONE. 2013;8:e78200.CrossRef Mao Y, Xu J, Li Z, Zhang N, Yin H, Liu Z. The role of nuclear beta-catenin accumulation in the Twist2-induced ovarian cancer EMT. PLoS ONE. 2013;8:e78200.CrossRef
32.
go back to reference Schmalhofer O, Brabletz S, Brabletz T. E-cadherin, beta-catenin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev. 2009;28:151–66.CrossRef Schmalhofer O, Brabletz S, Brabletz T. E-cadherin, beta-catenin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev. 2009;28:151–66.CrossRef
33.
go back to reference Beiter K, Hiendlmeyer E, Brabletz T, et al. beta-Catenin regulates the expression of tenascin-C in human colorectal tumors. Oncogene. 2005;24:8200–4.CrossRef Beiter K, Hiendlmeyer E, Brabletz T, et al. beta-Catenin regulates the expression of tenascin-C in human colorectal tumors. Oncogene. 2005;24:8200–4.CrossRef
34.
go back to reference MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell. 2009;17:9–26.CrossRef MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell. 2009;17:9–26.CrossRef
35.
go back to reference Gordon MD, Nusse R. Wnt signaling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem. 2006;281:22429–33.CrossRef Gordon MD, Nusse R. Wnt signaling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem. 2006;281:22429–33.CrossRef
36.
go back to reference Orsulic S, Huber O, Aberle H, Arnold S, Kemler R. E-cadherin binding prevents beta-catenin nuclear localization and beta-catenin/LEF-1-mediated transactivation. J Cell Sci. 1999;112(Pt 8):1237–45.PubMed Orsulic S, Huber O, Aberle H, Arnold S, Kemler R. E-cadherin binding prevents beta-catenin nuclear localization and beta-catenin/LEF-1-mediated transactivation. J Cell Sci. 1999;112(Pt 8):1237–45.PubMed
Metadata
Title
Ubiquitin-specific peptidase 39 regulates the process of proliferation and migration of human ovarian cancer via p53/p21 pathway and EMT
Authors
Congcong Yan
Jiahui Yuan
Jiajia Xu
Gongye Zhang
Xiaomei Li
Bing Zhang
Tianhui Hu
Xiaohua Huang
Yubin Mao
Gang Song
Publication date
01-11-2019
Publisher
Springer US
Published in
Medical Oncology / Issue 11/2019
Print ISSN: 1357-0560
Electronic ISSN: 1559-131X
DOI
https://doi.org/10.1007/s12032-019-1308-7

Other articles of this Issue 11/2019

Medical Oncology 11/2019 Go to the issue