SENP1 promotes hypoxia-induced cancer stemness by HIF-1α deSUMOylation and SENP1/HIF-1α positive feedback loop

Chun-Ping Cui, Carmen Chak-Lui Wong, Alan Ka-Lun Kai, Daniel Wai-Hung Ho, Eunice Yuen-Ting Lau, Yu-Man Tsui, Lo-Kong Chan, Tan-To Cheung, Kenneth Siu-Ho Chok, Albert C Y Chan, Regina Cheuk-Lam Lo, Joyce Man-Fong Lee, Terence Kin-Wah Lee, Irene Oi Lin Ng, Chun-Ping Cui, Carmen Chak-Lui Wong, Alan Ka-Lun Kai, Daniel Wai-Hung Ho, Eunice Yuen-Ting Lau, Yu-Man Tsui, Lo-Kong Chan, Tan-To Cheung, Kenneth Siu-Ho Chok, Albert C Y Chan, Regina Cheuk-Lam Lo, Joyce Man-Fong Lee, Terence Kin-Wah Lee, Irene Oi Lin Ng

Abstract

Objective: We investigated the effect and mechanism of hypoxic microenvironment and hypoxia-inducible factors (HIFs) on hepatocellular carcinoma (HCC) cancer stemness.

Design: HCC cancer stemness was analysed by self-renewal ability, chemoresistance, expression of stemness-related genes and cancer stem cell (CSC) marker-positive cell population. Specific small ubiquitin-like modifier (SUMO) proteases 1 (SENP1) mRNA level was examined with quantitative PCR in human paired HCCs. Immunoprecipitation was used to examine the binding of proteins and chromatin immunoprecipitation assay to detect the binding of HIFs with hypoxia response element sequence. In vivo characterisation was performed in immunocompromised mice and stem cell frequency was analysed.

Results: We showed that hypoxia enhanced the stemness of HCC cells and hepatocarcinogenesis through enhancing HIF-1α deSUMOylation by SENP1 and increasing stabilisation and transcriptional activity of HIF-1α. Furthermore, we demonstrated that SENP1 is a direct target of HIF-1/2α and a previously unrecognised positive feedback loop exists between SENP1 and HIF-1α.

Conclusions: Taken together, our findings suggest the significance of this positive feedback loop between HIF-1α and SENP1 in contributing to the increased cancer stemness in HCC and hepatocarcinogenesis under hypoxia. Drugs that specifically target SENP1 may offer a potential novel therapeutic approach for HCC.

Keywords: HEPATOCELLULAR CARCINOMA; MOLECULAR BIOLOGY; MOLECULAR PATHOLOGY; SIGNALING.

Conflict of interest statement

Competing interests: None declared.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.

Figures

Figure 1
Figure 1
The effect of hypoxia and hypoxia-inducible factor (HIF)-1/2α on the stemness of MHCC-97L cells. (A–C) The in vitro cell abilities for self-renewal (A), migration (B) and chemoresistance (C) to doxorubicin and sorafenib were enhanced under hypoxic condition (1% O2) in MHCC-97L cells and the knockdown of HIF-1α or HIF-2α suppressed these hypoxia-induced effects. (D) Hypoxia-induced increase of mRNA expression of stemness-related genes (Oct3/4, Nanog, BMI-1 and Notch1) was inhibited in HIF-1α or HIF-2α knockdown MHCC-97L cells. (E) CD24+ and CD133+ cell populations were increased in hypoxia-treated MHCC-97L cells, but the knockdown of HIF-1α or HIF-2α blunted the effects. (F) CD24+ population was analysed by FACS in sorted CD24+ (left) and CD24− (right) cells from MHCC-97L cells after cultured for indicated time under hypoxia or normoxia. (*p<0.05, **p<0.01, ***p<0.001, compared with the negative control in normoxia (20% O2); #p<0.05, ##p<0.01, ###p<0.001, compared with the negative control in hypoxia (1% O2)).
Figure 2
Figure 2
Upregulation of specific SUMO proteases 1 (SENP1) in hepatocellular carcinoma (HCC) tissues and the correlation between SENP1 and hypoxia-inducible factor (HIF)-α target genes. (A and B) SENP1 was consistently and significantly upregulated among the six SENP family members examined in our RNA-sequencing data on 16 pairs of human HCCs and their corresponding non-tumourous livers (NT-Ls) and data from The Cancer Genome Atlas (TCGA) of National Cancer Institute, USA (202 HCC tumours of all aetiologies including ‘unspecified’ aetiologies). (C) Using quantitative PCR on a larger cohort of 107 pairs of HCC tumour and corresponding NT-Ls, the similar result of SENP1 upregulation in HCC tumours was observed. (D) Correlation analysis of relative mRNA levels of SENP1 and VEGFa, VEGFb, LOX, LOXL2, PLOD2 or CD24 using our RNA-sequencing data on 16 pairs of human HCCs and their corresponding NT-Ls.
Figure 3
Figure 3
Effect of specific SUMO proteases 1 (SENP1) overexpression on the stemness of hepatocellular carcinoma (HCC) cells. (A–E) The effects of overexpression of SENP1, SENP1mut and non-target control (NTC) on the stemness of HCC cells shown by in vitro abilities of self-renewal (A), migration (B), CD24+ cell population (C) and mRNA expression of stemness-related genes (D) and chemoresistance (E), in hypoxic condition. (F) Limiting dilution xenograft formation of Huh-7 cells with NTC or SENP1 overexpression. (*p<0.05, **p<0.01, compared with the negative control in normoxia (20% O2), #p<0.05; ##p<0.01, compared with the negative control in hypoxia (1% O2)).
Figure 4
Figure 4
Effect of specific SUMO proteases 1 (SENP1) knockdown on the stemness of hepatocellular carcinoma (HCC) cells. (A–E) The effects of knockdown of SENP1 (shSENP1-#1 and shSENP1-#4) and non-target control (NTC) on the stemness of HCC cells shown by in vitro abilities of self-renewal (A), migration (B) and chemoresistance (C), CD24+ cell population (D) and mRNA expression of stemness-related genes (E) in hypoxic condition. (F) Limiting dilution xenograft formation of MHCC-97L cells with NTC or SENP1 knockdown. (*p<0.05, **p<0.01, ***p<0.001, as compared with the negative control in normoxia (20% O2); #p<0.05, ##p<0.01, ###p<0.001, compared with the negative control in hypoxia (1% O2)).
Figure 5
Figure 5
Specific SUMO proteases 1 (SENP1) play a role as the direct target gene of hypoxia-inducible factor (HIF)-1/2α in hepatocellular carcinoma (HCC) cells. (A and B) Protein (A) and mRNA (B) levels of SENP1 were increased under hypoxia in HIF-1α-dependent and HIF-2α-dependent manner in MHCC-97L cells. (C) Chromatin immunoprecipitation assay was used to determine the bind of HIF-1α, HIF-2α and HIF-1β to the hypoxia response element (HRE) sequence in SENP1 promoter. (*p2); #p<0.05, ##p<0.01, ###p<0.001, as compared with the negative control in hypoxia (1% O2)).
Figure 6
Figure 6
Increased stability and transcriptional activity of hypoxia-inducible factor (HIF)-1α through deSUMOylation by specific SUMO proteases 1 (SENP1) in hepatocellular carcinoma (HCC) cells under hypoxic condition. (A) Western blot analyses were used to examine the protein level of HIF-1α and HIF-2α in SENP1-knockdown and non-target control (NTC) HCC cells under hypoxia or normoxia. (B) immunoprecipitation (IP) assay was used to detect the SUMOylation of HIF-1/2α by SUMO-1 or SUMO-2/3 in HCC cells. IP was conducted to obtain the complex protein with anti-HIF-1α or anti-HIF-2α antibody in SENP1-knockdown or vehicle-infected SMMC-7721 cells, which were incubated under normoxia or hypoxia for 24 hours. Western blot analyses were then carried out on the whole cell lysates and the IP complex with anti-SUMO-1 or SUMO-2/3 antibody. Inputs are presented in the right-most panel. (C) Fold change of the relative luciferase activity was examined by luciferase-reporter assay in SENP1-knockdown and NTC SMMC-7721 cells which were incubated under normoxia or hypoxia for 24 hours. (D) Fold change of the relative luciferase activity was examined in NTC, SENP1 overexpression (SENP1 OE) and SENP1mut overexpression (SENP1mut OE) Huh-7 cells which were incubated under normoxia or hypoxia for 36 hours. (E) IP assay was used to detect the binding of exogenous HIF-1α or the mutants with SUMO-1 in SMMC-7721 cells in normoxia. First, IP was conducted to obtain the complex protein with anti-HIF-1α antibody in SENP1-knockdown or vehicle-infected SMMC-7721 cells, in which HA-SUMO-1 was cotransfected with RGS-HIF-1α wild type (WT), RGS-HIF-1α K391R, RGS-HIF-1α K477R, or RGS-HIF-1α SM. Western blot analyses for indicated proteins in the whole cell lysates and the IP complex with anti-HA antibody. (F) Fold of the relative luciferase activity was examined in SENP1-knockdown and NTC SMMC-7721 cells (in normoxia), in which HA-SUMO-1 was cotransfected with RGS-HIF-1α WT, RGS-HIF-1α K391R, RGS-HIF-1α K477R or RGS-HIF-1α SM. (*p2); #p<0.05, ##p<0.01, ###p<0.001, compared with the negative control in hypoxia (1% O2)).
Figure 7
Figure 7
Mutation of SUMO sites rescues the loss of hypoxia-induced stemness in specific SUMO proteases 1 (SENP1)-knockdown hepatocellular carcinoma (HCC) cells. The effects of SENP1 knockdown on the stemness of HCC cells with overexpressing hypoxia-inducible factor (HIF)-1α wild type (WT), HIF-1α K391R, HIF-1α K477R or HIF-1α SM. In vitro abilities of self-renewal (A), migration (B) and chemoresistance (C), CD24+ cell population (D) and mRNA expression of stemness-related genes (E) were determined in hypoxic condition. SENP1 knockdown suppressed the effects of HIF-1α WT, while the three HIF-1α mutants partially to almost completely rescued HIF-1α-induced enhancement of HCC cell stemness in shSENP1 HCC cells. (F) A cartoon summarising our findings. HIF-1/2α induced by the hypoxic microenvironment increases the transcription of SENP1 in HCC cells. SENP1 represses the SUMOylation of HIF-1α at K391 and K477 by its SUMO protease activity, thus enhancing the stability and transcriptional activity of HIF-1α. These in turn upregulate the expression of HIF target genes, including SENP1 and stemness-related genes, and contribute to the increased stemness properties. (*p<0.05, **p<0.01, ***p<0.001, as compared with the negative control).

References

    1. Lee TK, Cheung VC, Ng IO. Liver tumor-initiating cells as a therapeutic target for hepatocellular carcinoma. Cancer Lett 2013;338:101–9. 10.1016/j.canlet.2012.05.001
    1. Ma S, Chan KW, Hu L, et al. . Identification and characterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology 2007;132:2542–56. 10.1053/j.gastro.2007.04.025
    1. Yang ZF, Ho DW, Ng MN, et al. . Significance of CD90+ cancer stem cells in human liver cancer. Cancer Cell 2008;13:153–66. 10.1016/j.ccr.2008.01.013
    1. Yamashita T, Ji J, Budhu A, et al. . EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology 2009;136:1012–24. 10.1053/j.gastro.2008.12.004
    1. Lee TK, Castilho A, Cheung VC, et al. . CD24(+) liver tumor-initiating cells drive self-renewal and tumor initiation through STAT3-mediated NANOG regulation. Cell stem cell 2011;9:50–63. 10.1016/j.stem.2011.06.005
    1. Lee TK, Cheung VC, Lu P, et al. . Blockade of CD47-mediated cathepsin S/protease-activated receptor 2 signaling provides a therapeutic target for hepatocellular carcinoma. Hepatology 2014;60:179–91. 10.1002/hep.27070
    1. Keith B, Johnson RS, Simon MC. HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression. Nat Rev Cancer 2011;12:9–22. 10.1038/nrc3183
    1. Zheng SS, Chen XH, Yin X, et al. . Prognostic significance of HIF-1α expression in hepatocellular carcinoma: a meta-analysis. PLoS ONE 2013;8:e65753 10.1371/journal.pone.0065753
    1. Liang Y, Zheng T, Song R, et al. . Hypoxia-mediated sorafenib resistance can be overcome by EF24 through Von Hippel-Lindau tumor suppressor-dependent HIF-1α inhibition in hepatocellular carcinoma. Hepatology 2013;57:1847–57. 10.1002/hep.26224
    1. Zhao D, Zhai B, He C, et al. . Upregulation of HIF-2α induced by sorafenib contributes to the resistance by activating the TGF-α/EGFR pathway in hepatocellular carcinoma cells. Cell Signal 2014;26:1030–9. 10.1016/j.cellsig.2014.01.026
    1. Lin Q, Yun Z. Impact of the hypoxic tumor microenvironment on the regulation of cancer stem cell characteristics. Cancer Biol Ther 2010;9:949–56. 10.4161/cbt.9.12.12347
    1. Pietras A, Gisselsson D, Ora I, et al. . High levels of HIF-2alpha highlight an immature neural crest-like neuroblastoma cell cohort located in a perivascular niche. J Pathol 2008;214:482–8. 10.1002/path.2304
    1. Pietras A, Hansford LM, Johnsson AS, et al. . HIF-2alpha maintains an undifferentiated state in neural crest-like human neuroblastoma tumor-initiating cells. Proc Natl Acad Sci USA 2009;106:16805–10. 10.1073/pnas.0904606106
    1. Li Z, Bao S, Wu Q, et al. . Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell 2009;15:501–13. 10.1016/j.ccr.2009.03.018
    1. Muramatsu S, Tanaka S, Mogushi K, et al. . Visualization of stem cell features in human hepatocellular carcinoma reveals in vivo significance of tumor-host interaction and clinical course. Hepatology 2013;58:218–28. 10.1002/hep.26345
    1. Flotho A, Melchior F. Sumoylation: a regulatory protein modification in health and disease. Annu Rev Biochem 2013;82:357–85. 10.1146/annurev-biochem-061909-093311
    1. Johnson ES. Protein modification by SUMO. Annu Rev Biochem 2004;73:355–82. 10.1146/annurev.biochem.73.011303.074118
    1. Hay RT. SUMO: a history of modification. Mol Cell 2005;18:1–12. 10.1016/j.molcel.2005.03.012
    1. Kim JH, Baek SH. Emerging roles of desumoylating enzymes. Biochim Biophys Acta 2009;1792:155–62. 10.1016/j.bbadis.2008.12.008
    1. Deng R, Zhao X, Qu Y, et al. . Shp2 SUMOylation promotes ERK activation and hepatocellular carcinoma development. Oncotarget 2015;6:9355–69. 10.18632/oncotarget.3323
    1. Jiang QF, Tian YW, Shen Q, et al. . SENP2 regulated the stability of β-catenin through WWOX in hepatocellular carcinoma cell. Tumour Biol 2014;35:9677–82. 10.1007/s13277-014-2239-8
    1. Liu J, Sha M, Wang Q, et al. . Small ubiquitin-related modifier 2/3 interacts with p65 and stabilizes it in the cytoplasm in HBV-associated hepatocellular carcinoma. BMC cancer 2015;15:675 10.1186/s12885-015-1665-3
    1. Mukhopadhyay D, Dasso M. Modification in reverse: the SUMO proteases. Trends Biochem Sci 2007;32:286–95. 10.1016/j.tibs.2007.05.002
    1. Yang ST, Yen CJ, Lai CH, et al. . SUMOylated CPAP is required for IKK-mediated NF-κB activation and enhances HBx-induced NF-κB signaling in HCC. J Hepatol 2013;58:1157–64. 10.1016/j.jhep.2013.01.025
    1. Tomasi ML, Tomasi I, Ramani K, et al. . S-adenosyl methionine regulates ubiquitin-conjugating enzyme 9 protein expression and sumoylation in murine liver and human cancers. Hepatology 2012;56:982–93. 10.1002/hep.25701
    1. Cheng J, Kang X, Zhang S, et al. . SUMO-specific protease 1 is essential for stabilization of HIF1alpha during hypoxia. Cell 2007;131:584–95. 10.1016/j.cell.2007.08.045
    1. Berta MA, Mazure N, Hattab M, et al. . SUMOylation of hypoxia-inducible factor-1α reduces its transcriptional activity. Biochem Biophys Res Commun 2007;360:646–52. 10.1016/j.bbrc.2007.06.103
    1. Carbia-Nagashima A, Gerez J, Perez-Castro C, et al. . RSUME, a small RWD-containing protein, enhances SUMO conjugation and stabilizes HIF-1α during hypoxia. Cell 2007;131:309–23. 10.1016/j.cell.2007.07.044
    1. Parhira S, Zhu GY, Jiang RW, et al. . 2′-Epi-uscharin from the latex of Calotropis gigantea with HIF-1 inhibitory activity. Sci Rep 2014;4:4748 10.1038/srep04748
    1. Wong CC, Tse AP, Huang YP, et al. . Lysyl oxidase-like 2 is critical to tumor microenvironment and metastatic niche formation in hepatocellular carcinoma. Hepatology 2014;60:1645–58. 10.1002/hep.27320
    1. Ma S, Lee TK, Zheng BJ, et al. . CD133+ HCC cancer stem cells confer chemoresistance by preferential expression of the Akt/PKB survival pathway. Oncogene 2008;27:1749–58. 10.1038/sj.onc.1210811
    1. Li C, Heidt DG, Dalerba P, et al. . Identification of pancreatic cancer stem cells. Cancer Res 2007;67:1030–7. 10.1158/0008-5472.CAN-06-2030
    1. Ke J, Wu X, Wu X, et al. . A subpopulation of CD24+ cells in colon cancer cell lines possess stem cell characteristics. Neoplasma 2012;59:282–8. 10.4149/neo_2012_036
    1. Vermeulen L, Todaro M, de Sousa Mello F, et al. . Single-cell cloning of colon cancer stem cells reveals a multi-lineage differentiation capacity. Proc Natl Acad Sci USA 2008;105:13427–32. 10.1073/pnas.0805706105
    1. Ishizawa K, Rasheed ZA, Karisch R, et al. . Tumor-initiating cells are rare in many human tumors. Cell Stem Cell 2010;7:279–82. 10.1016/j.stem.2010.08.009
    1. Thomas S, Harding MA, Smith SC, et al. . CD24 is an effector of HIF-1-driven primary tumor growth and metastasis. Cancer Res 2012;72:5600–12. 10.1158/0008-5472.CAN-11-3666
    1. Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods 2009;347:70–8. 10.1016/j.jim.2009.06.008
    1. Xu Y, Zuo Y, Zhang H, et al. . Induction of SENP1 in endothelial cells contributes to hypoxia-driven VEGF expression and angiogenesis. J Biol Chem 2010;285:36682–8. 10.1074/jbc.M110.164236
    1. Du L, Li YJ, Fakih M, et al. . Role of SUMO activating enzyme in cancer stem cell maintenance and self-renewal. Nat Commun 2016;7:12326 10.1038/ncomms12326
    1. Antico Arciuch VG, Tedesco L, Fuertes M, et al. . Role of RSUME in inflammation and cancer. FEBS Lett 2015;589:3330–5. 10.1016/j.febslet.2015.07.048
    1. Li J, Xu Y, Long XD, et al. . Cbx4 governs HIF-1α to potentiate angiogenesis of hepatocellular carcinoma by its SUMO E3 ligase activity. Cancer Cell 2014;25:118–31. 10.1016/j.ccr.2013.12.008
    1. Mei Z, Jiao H, Wang W, et al. . Polycomb chromobox 4 enhances migration and pulmonary metastasis of hepatocellular carcinoma cell line MHCC97L. Sci China Life Sci 2014;57:610–17. 10.1007/s11427-014-4663-9
    1. van Hagen M, Overmeer RM, Abolvardi SS, et al. . RNF4 and VHL regulate the proteasomal degradation of SUMO-conjugated Hypoxia-Inducible Factor-2α. Nucleic Acids Res 2010;38:1922–31. 10.1093/nar/gkp1157

Source: PubMed

3
Abonneren