Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

UHRF1 promotes renal cell carcinoma progression through epigenetic regulation of TXNIP

Subjects

Abstract

UHRF1 is an important epigenetic regulator that belongs to the UHRF family. Overexpression of UHRF1 has been found in many kinds of tumors and its overexpression is associated with poor prognosis and short survival in certain cancer types. However, its function in renal cell carcinoma (RCC) is not clear. Here we report that RCC tumor tissues had obviously higher UHRF1 expression than normal renal tissues. Downregulation of UHRF1 by siRNA or shRNA in RCC cell lines resulted in decreased cell viability, inhibited cell migration and invasion, and increased apoptosis. UHRF1 knockdown RCC xenografts also resulted in obviously inhibited tumor growth in vivo. After downregulation of UHRF1 in RCC cells, the expression of TXNIP was upregulated. In addition, after UHRF1 and TXNIP were simultaneously downregulated, cell viability and cell invasion increased, whereas cell apoptosis decreased compared with UHRF1 single downregulated cells. We also showed that UHRF1 could recruit HDAC1 to the TXNIP promoter and mediate the deacetylation of histone H3K9, resulting in the inhibition of TXNIP expression. Our results confirm that UHRF1 has oncogenic function in RCC and UHRF1 may promote tumor progression through epigenetic regulation of TXNIP. UHRF1 might be used as a therapeutic target for RCC treatment.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Lopez-Beltran A, Carrasco JC, Cheng L, Scarpelli M, Kirkali Z, Montironi R. 2009 update on the classification of renal epithelial tumors in adults. Int J Urol. 2009;16:432–43.

    Article  Google Scholar 

  2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66:7–30.

    Article  Google Scholar 

  3. Bhatt JR, Finelli A. Landmarks in the diagnosis and treatment of renal cell carcinoma. Nat Rev Urol. 2014;11:517–25.

    Article  Google Scholar 

  4. PA C. Optimizing patient adherence to targeted therapies in renal cell carcinoma. Clin J Oncol Nurs. 2014;18:694–700.

    Article  Google Scholar 

  5. Rini BI, Campbell SC, Escudier B. Renal cell carcinoma. Lancet. 2009;373:1119–32.

    Article  CAS  Google Scholar 

  6. Tauber M, Fischle W. Conserved linker regions and their regulation determine multiple chromatin-binding modes of UHRF1. Nucleus. 2015;6:123–32.

    Article  Google Scholar 

  7. Arita KAM, Tochio H, Nakamura Y, Shirakawa M. Recognition of hemi-methylated DNA by the SRA protein UHRF1 by a base-flipping mechanism. Nature. 2008;455:818–21.

    Article  CAS  Google Scholar 

  8. Hashimoto H, Horton JR, Zhang X, Bostick M, Jacobsen SE, Cheng X. The SRA domain of UHRF1 flips 5-methylcytosine out of the DNA helix. Nature. 2008;455:826–9.

    Article  CAS  Google Scholar 

  9. Bostick M, Kim JK, Esteve PO, Clark A, Pradhan S, Jacobsen SE. UHRF1 plays a role in maintaining DNA methylation in mammalian cells. Science. 2007;317:1760–4.

    Article  CAS  Google Scholar 

  10. Sharif J, Muto M, Takebayashi S, Suetake I, Iwamatsu A, Endo TA, et al. The SRA protein Np95 mediates epigenetic inheritance by recruiting Dnmt1 to methylated DNA. Nature. 2007;450:908–12.

    Article  CAS  Google Scholar 

  11. Hervouet E, Vallette FM, Cartron PF. Dnmt1/Transcription factor interactions: an alternative mechanism of DNA methylation inheritance. Genes Cancer. 2010;1:434–43.

    Article  CAS  Google Scholar 

  12. Geng Y, Gao YF, Ju HX, Yan F. Diagnostic and prognostic value of plasma and tissue ubiquitin-like, containing PHD and RING finger domains 1 in breast cancer patients. Cancer Sci. 2013;104:194–9.

    Article  CAS  Google Scholar 

  13. Ge TT, Yang M, Chen Z, Lou G, Gu T. UHRF1 gene silencing inhibits cell proliferation and promotes cell apoptosis in human cervical squamous cell carcinoma CaSki cells. J Ovarian Res. 2016;9:42.

    Article  Google Scholar 

  14. Nakamura KBY, Kosumi K, Harada K, Shigaki H, Miyake K, Kiyozumi Y, et al. UHRF1 regulates global DNA hypomethylation and is associated with poor prognosis in esophageal squamous cell carcinoma. Oncotarget. 2016;7:57821–31.

    PubMed  PubMed Central  Google Scholar 

  15. Liu X, Ou H, Xiang L, Li X, Huang Y, Yang D. Elevated UHRF1 expression contributes to poor prognosis by promoting cell proliferation and metastasis in hepatocellular carcinoma. Oncotarget. 2017;8:10510–22.

    PubMed  PubMed Central  Google Scholar 

  16. Wan X, Yang S, Huang W, Wu D, Chen H, Wu M, et al. UHRF1 overexpression is involved in cell proliferation and biochemical recurrence in prostate cancer after radical prostatectomy. J Exp Clin Cancer Res. 2016;35:34.

    Article  Google Scholar 

  17. Jenkins Y, Markovtsov V, Lang W, Sharma P, Pearsall D, Warner J, et al. Critical role of the ubiquitin ligase activity of UHRF1, a nuclear RING finger protein, in tumor cell growth. Mol Biol Cell. 2005;16:5621–9.

    Article  CAS  Google Scholar 

  18. Yan F, Shao L-J, Hu X-Y. Knockdown of UHRF1 by lentivirus-mediated shRNA inhibits ovarian cancer cell growth. Asian Pac J Cancer Prev. 2015;16:1343–8.

    Article  Google Scholar 

  19. Qin Y, Wang J, Gong W, Zhang M, Tang Z, Zhang J, et al. UHRF1 depletion suppresses growth of gallbladder cancer cells through induction of apoptosis and cell cycle arrest. Oncol Rep. 2014;31:2635–43.

    Article  CAS  Google Scholar 

  20. Zhang ZY, Cai JJ, Hong J, Li KK, Ping Z, Wang Y, et al. Clinicopathological analysis of UHRF1 expression in medulloblastoma tissues and its regulation on tumor cell proliferation. Med Oncol. 2016;33:99.

    Article  Google Scholar 

  21. Sheng Y, Wang H, Liu D, Zhang C, Deng Y, Yang F, et al. Methylation of tumor suppressor gene CDH13 and SHP1 promoters and their epigenetic regulation by the UHRF1/PRMT5 complex in endometrial carcinoma. Gynecol Oncol. 2016;140:145–51.

    Article  CAS  Google Scholar 

  22. Daskalos A, Oleksiewicz U, Filia A, Nikolaidis G, Xinarianos G, Gosney JR, et al. UHRF1-mediated tumor suppressor gene inactivation in nonsmall cell lung cancer. Cancer. 2011;117:1027–37.

    Article  CAS  Google Scholar 

  23. Mudbhary R, Hoshida Y, Chernyavskaya Y, Jacob V, Villanueva A, Fiel MI, et al. UHRF1 overexpression drives DNA hypomethylation and hepatocellular carcinoma. Cancer Cell. 2014;25:196–209.

    Article  CAS  Google Scholar 

  24. Zhuo H, Tang J, Lin Z, Jiang R, Zhang X, Ji J, et al. The aberrant expression of MEG3 regulated by UHRF1 predicts the prognosis of hepatocellular carcinoma. Mol Carcinog. 2016;55:209–19.

    Article  CAS  Google Scholar 

  25. Hervouet E, Lalier L, Debien E, Cheray M, Geairon A, Rogniaux H, et al. Disruption of Dnmt1/PCNA/UHRF1 interactions promotes tumorigenesis from human and mice glial cells. PLoS ONE. 2010;5:e11333.

    Article  Google Scholar 

  26. Babbio F, Pistore C, Curti L, Castiglioni I, Kunderfranco P, Brino L, et al. The SRA protein UHRF1 promotes epigenetic crosstalks and is involved in prostate cancer progression. Oncogene. 2012;31:4878–87.

    Article  CAS  Google Scholar 

  27. Sabatino L, Fucci A, Pancione M, Carafa V, Nebbioso A, Pistore C, et al. UHRF1 coordinates peroxisome proliferator activated receptor gamma (PPARG) epigenetic silencing and mediates colorectal cancer progression. Oncogene. 2012;31:5061–72.

    Article  CAS  Google Scholar 

  28. Unoki M, Nishidate T, Nakamura Y. ICBP90, an E2F-1 target, recruits HDAC1 and binds to methyl-CpG through its SRA domain. Oncogene. 2004;23:7601–10.

    Article  CAS  Google Scholar 

  29. Cartron PF, Blanquart C, Hervouet E, Gregoire M, Vallette FM. HDAC1-mSin3a-NCOR1, Dnmt3b-HDAC1-Egr1 and Dnmt1-PCNA-UHRF1-G9a regulate the NY-ESO1 gene expression. Mol Oncol. 2013;7:452–63.

    Article  CAS  Google Scholar 

  30. Zhou J, Yu Q, Chng WJ. TXNIP (VDUP-1, TBP-2): a major redox regulator commonly suppressed in cancer by epigenetic mechanisms. Int J Biochem Cell Biol. 2011;43:1668–73.

    Article  CAS  Google Scholar 

  31. Sheth SS, Bodnar JS, Ghazalpour A, Thipphavong CK, Tsutsumi S, Tward AD, et al. Hepatocellular carcinoma in Txnip-deficient mice. Oncogene. 2006;25:3528–36.

    Article  CAS  Google Scholar 

  32. Han SH, Jeon JH, Ju HR, Jung U, Kim KY, Yoo HS, et al. VDUP1 upregulated by TGF-beta1 and 1,25-dihydorxyvitamin D3 inhibits tumor cell growth by blocking cell-cycle progression. Oncogene. 2003;22:4035–46.

    Article  CAS  Google Scholar 

  33. Ikarashi M, Takahashi Y, Ishii Y, Nagata T, Asai S, Ishikawa K. Vitamin D3 up-regulated protein 1 (VDUP1) expression in gastrointestinal cancer and its relation to stage of disease. Anticancer Res. 2002;22:4045–8.

    CAS  PubMed  Google Scholar 

  34. Shen L, O’Shea JM, Kaadige MR, Cunha S, Wilde BR, Cohen AL, et al. Metabolic reprogramming in triple-negative breast cancer through Myc suppression of TXNIP. Proc Natl Acad Sci USA. 2015;112:5425–30.

    Article  CAS  Google Scholar 

  35. Li J, Yue Z, Xiong W, Sun P, You K, Wang J. TXNIP overexpression suppresses proliferation and induces apoptosis in SMMC7221 cells through ROS generation and MAPK pathway activation. Oncol Rep. 2017;37:3369–76.

    Article  CAS  Google Scholar 

  36. Morrison JA, Pike LA, Sams SB, Sharma V, Zhou Q, Severson JJ, et al. Thioredoxin interacting protein (TXNIP) is a novel tumor suppressor in thyroid cancer. Mol Cancer. 2014;13:62.

    Article  Google Scholar 

  37. Dutta KK, Nishinaka Y, Masutani H, Akatsuka S, Aung TT, Shirase T, et al. Two distinct mechanisms for loss of thioredoxin-binding protein-2 in oxidative stress-induced renal carcinogenesis. Lab Investig. 2005;85:798–807.

    Article  CAS  Google Scholar 

  38. Butler LM, Zhou X, Xu WS, Scher HI, Rifkind RA, Marks PA, et al. The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc Natl Acad Sci USA. 2002;99:11700–5.

    Article  CAS  Google Scholar 

  39. Zhou J, Bi C, Cheong LL, Mahara S, Liu SC, Tay KG, et al. The histone methyltransferase inhibitor, DZNep, up-regulates TXNIP, increases ROS production, and targets leukemia cells in AML. Blood. 2011;118:2830–9.

    Article  Google Scholar 

  40. Kim JK, Esteve PO, Jacobsen SE, Pradhan S. UHRF1 binds G9a and participates in p21 transcriptional regulation in mammalian cells. Nucleic Acids Res. 2009;37:493–505.

    Article  CAS  Google Scholar 

  41. Qin J, Wu SP, Creighton CJ, Dai F, Xie X, Cheng CM, et al. COUP-TFII inhibits TGF-beta-induced growth barrier to promote prostate tumorigenesis. Nature. 2013;493:236–40.

    Article  CAS  Google Scholar 

  42. Wei M, Jiao D, Han D, Wu J, Wei F, Zheng G, et al. Knockdown of RNF2 induces cell cycle arrest and apoptosis in prostate cancer cells through the upregulation of TXNIP. Oncotarget. 2017;8:5323–38.

    PubMed  Google Scholar 

  43. Shi T, Dimitrov I, Zhang Y, Tax FE, Yi J, Gou X, et al. Accelerated rates of protein evolution in barley grain and pistil biased genes might be legacy of domestication. Plant Mol Biol. 2015;89:253–61.

    Article  CAS  Google Scholar 

  44. Shang Y, Hu X, DiRenzo J, Lazar MA, Brown M. Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription. Cell. 2000;103:843–52.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Natural Science Foundation of China (No. 81772734, 81220108011, 81372225, and 81372771) and the open project program of the State Key Laboratory of Cancer Biology (Fourth Military Medical University) (CBSKL201705).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Weijun Qin, He Wang or Weihong Wen.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jiao, D., Huan, Y., Zheng, J. et al. UHRF1 promotes renal cell carcinoma progression through epigenetic regulation of TXNIP. Oncogene 38, 5686–5699 (2019). https://doi.org/10.1038/s41388-019-0822-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-019-0822-6

This article is cited by

Search

Quick links