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:

UFBP1, a key component in ufmylation, enhances drug sensitivity by promoting proteasomal degradation of oxidative stress-response transcription factor Nrf2

Abstract

The key component in the UFM1 conjugation system, UFM1-binding and PCI domain-containing protein 1 (UFBP1), regulates many biological processes. Recently it has been shown that low UFBP1 protein level is associated with the worse outcome of gastric cancer patients. However, how it responses to the sensitivity of gastric cancer to chemotherapy drugs and the underlying molecular mechanism remain elusive. Here, we discovered that high UFBP1 expression increases the progression-free survival of advanced gastric cancer patients treated with platinum-based chemotherapy. Cell-line based studies unveiled that UFBP1 expression enhances while UFBP1 knockdown attenuates the sensitivity of gastric cancer cells to cisplatin. High-throughput SILAC-based quantitative proteomic analysis revealed that the protein level of aldo-keto reductase 1Cs (AKR1Cs) is significantly downregulated by UFBP1. Flow cytometry analysis showed that UFBP1 expression increases while UFBP1 knockdown reduces reactive oxygen species upon cisplatin treatment. We further disclosed that UFBP1 attenuates the gene expression of AKR1Cs and the transcription activity of the master oxidative stress-response transcription factor Nrf2 (nuclear factor erythroid-2-related factor 2). Detailed mechanistic studies manifested that UFBP1 promotes the formation of K48-linked polyubiquitin chains on Nrf2 and thus augments its proteasome-mediated degradation. Experiments using genetic depletion and pharmacological activation in vitro and in vivo demonstrated that UFBP1 enhances the sensitivity of gastric cancer cells to cisplatin through the Nrf2/AKR1C axis. Overall, this work discovered a novel prognostic biomarker for gastric cancer patients treated with platinum-based chemotherapy and elucidated the underlying molecular mechanism, which may benefit to future personalized chemotherapy.

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

Access options

Buy this article

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

Fig. 1: UFBP1 enhances the sensitivity of gastric cancer to cisplatin (CDDP).
Fig. 2: UFBP1 downregulates AKR1C1/2/3 and enhances oxidative reduction process in the gastric cancer cells.
Fig. 3: UFBP1 downregulates AKR1C protein and mRNA levels and Nrf2 protein level.
Fig. 4: UFBP1 promotes Nrf2 degradation through the ubiquitin-proteasome system.
Fig. 5: UFBP1 enhances the sensitivity of gastric cancer cells to cisplatin through Nrf2/AKR1C axis.
Fig. 6: Nrf2/AKR1C axis mediates the UFBP1-enhanced sensitivity of gastric cancer cells to cisplatin in xenograft mouse model.

Similar content being viewed by others

Data availability

Additional Materials and methods were provided in the Supplementary Materials. The proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE [43] partner repository with the dataset identifier PXD013590.

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.

    Google Scholar 

  2. Van Cutsem E, Sagaert X, Topal B, Haustermans K, Prenen H. Gastric cancer. Lancet 2016;388:2654–64.

    Article  Google Scholar 

  3. Ivanova T, Zouridis H, Wu Y, Cheng LL, Tan IB, Gopalakrishnan V, et al. Integrated epigenomics identifies BMP4 as a modulator of cisplatin sensitivity in gastric cancer. Gut 2013;62:22–33.

    Article  CAS  Google Scholar 

  4. Xi P, Ding D, Zhou J, Wang M, Cong YS. DDRGK1 regulates NF-κ activity by modulating IκBα stability. PLoS One. 2013;8:e64231.

  5. Lin JX, Xie XS, Weng XF, Zheng CH, Xie JW, Wang JB, et al. Low expression of CDK5RAP3 and DDRGK1 indicates a poor prognosis in patients with gastric cancer. World J Gastroenterol. 2018;24:3898–907.

    Article  CAS  Google Scholar 

  6. Cai Y, Zhu G, Liu S, Pan Z, Quintero M, Poole CJ, et al. Indispensable role of the ubiquitin-fold modifier 1-specific E3 ligase in maintaining intestinal homeostasis and controlling gut inflammation. Cell Discov. 2019;5:7.

  7. Lee JY, Moon S, Kim YK, Lee SH, Lee BS, Park MY, et al. Genome-based exome sequencing analysis identifies GYG1, DIS3L and DDRGK1 are associated with myocardial infarction in Koreans. J Genet. 2017;96:1041–6.

    Article  CAS  Google Scholar 

  8. Egunsola AT, Bae Y, Jiang MM, Liu DS, Chen-Evenson Y, Bertin T, et al. Loss of DDRGK1 modulates SOX9 ubiquitination in spondyloepimetaphyseal dysplasia. J Clin Invest. 2017;127:1475–84.

    Article  Google Scholar 

  9. Cai Y, Pi W, Sivaprakasam S, Zhu X, Zhang M, Chen J, et al. UFBP1, a key component of the Ufm1 conjugation system, is essential for ufmylation-mediated regulation of erythroid development. PLoS Genet. 2015;11:e1005643.

    Article  Google Scholar 

  10. Rukova B, Staneva R, Hadjidekova S, Stamenov G, Milanova V, Toncheva D. Whole genome methylation analyses of schizophrenia patients before and after treatment. Biotechnol Biotechnol Equip. 2014;28:518–24.

    Article  CAS  Google Scholar 

  11. Tanaka Y, Kurosaki M, Nishida N, Sugiyama M, Matsuura K, Sakamoto N, et al. Genome-wide association study identified ITPA/DDRGK1 variants reflecting thrombocytopenia in pegylated interferon and ribavirin therapy for chronic hepatitis C. Hum Mol Genet. 2011;20:3507–16.

    Article  CAS  Google Scholar 

  12. Neziri D, Pajenda S, Amuge R, Ilhan A, Wewalka M, Hormann G, et al. DDRGK1 in urine indicative of tubular cell injury in intensive care patients with serious infections. J Nephropathol. 2016;5:65–71.

    Article  Google Scholar 

  13. Yoo HM, Kang SH, Kim JY, Lee JE, Seong MW, Lee SW, et al. Modification of ASC1 by UFM1 is crucial for ERα transactivation and breast cancer development. Mol Cell. 2014;56:261–74.

    Article  CAS  Google Scholar 

  14. Yoo HM, Park JH, Jeon YJ, Chung CH. Ubiquitin-fold modifier 1 acts as a positive regulator of breast cancer. Front Endocrinol (Lausanne). 2015;6:36.

    Article  Google Scholar 

  15. Walczak CP, Leto DE, Zhang L, Riepe C, Muller RY, DaRosa PA, et al. Ribosomal protein RPL26 is the principal target of UFMylation. Proc Natl Acad Sci USA. 2019;116:1299–308.

    Article  CAS  Google Scholar 

  16. Wang L, Xu Y, Rogers H, Saidi L, Noguchi CT, Li H, et al. UFMylation of RPL26 links translocation-associated quality control to endoplasmic reticulum protein homeostasis. Cell Res. 2020;30:5–20.

    Article  CAS  Google Scholar 

  17. Liang JR, Lingeman E, Luong T, Ahmed S, Muhar M, Nguyen T, et al. A genome-wide ER-phagy screen highlights key roles of mitochondrial metabolism and ER-resident UFMylation. Cell 2020;180:1160–77.

  18. Qin B, Yu J, Nowsheen S, Wang M, Tu X, Liu T, et al. UFL1 promotes histone H4 ufmylation and ATM activation. Nat Commun. 2019;10:1242.

    Article  Google Scholar 

  19. Wang Z, Gong Y, Peng B, Shi R, Fan D, Zhao H, et al. MRE11 UFMylation promotes ATM activation. Nucleic Acids Res. 2019;47:4124–35.

    Article  CAS  Google Scholar 

  20. Liu J, Guan D, Dong M, Yang J, Wei H, Liang Q, et al. UFMylation maintains tumour suppressor p53 stability by antagonizing its ubiquitination. Nat Cell Biol. 2020;22:1056–63.

  21. Wang XJ, Hayes JD, Wolf CR. Generation of a stable antioxidant response element-driven reporter gene cell line and its use to show redox-dependent activation of Nrf2 by cancer chemotherapeutic agents. Cancer Res. 2006;66:10983–94.

    Article  CAS  Google Scholar 

  22. Rojo de la Vega M, Chapman E, Zhang DD. NRF2 and the hallmarks of cancer. Cancer Cell. 2018;34:21–43.

    Article  CAS  Google Scholar 

  23. Bortolozzi R, Bresolin S, Rampazzo E, Paganin M, Maule F, Mariotto E, et al. AKR1C enzymes sustain therapy resistance in paediatric T-ALL. Br J Cancer. 2018;118:985–94.

    Article  CAS  Google Scholar 

  24. Chowdhry S, Zhang Y, McMahon M, Sutherland C, Cuadrado A, Hayes JD. Nrf2 is controlled by two distinct β-TrCP recognition motifs in its Neh6 domain, one of which can be modulated by GSK-3 activity. Oncogene 2013;32:3765–81.

    Article  CAS  Google Scholar 

  25. Wu T, Zhao F, Gao B, Tan C, Yagishita N, Nakajima T, et al. Hrd1 suppresses Nrf2-mediated cellular protection during liver cirrhosis. Genes Dev. 2014;28:708–22.

    Article  CAS  Google Scholar 

  26. Chen CC, Chu CB, Liu KJ, Huang CY, Chang JY, Pan WY, et al. Gene expression profiling for analysis acquired oxaliplatin resistant factors in human gastric carcinoma TSGH-S3 cells: the role of IL-6 signaling and Nrf2/AKR1C axis identification. Biochem Pharm. 2013;86:872–87.

    Article  CAS  Google Scholar 

  27. Jung KA, Kwak MK. Enhanced 4-hydroxynonenal resistance in KEAP1 silenced human colon cancer cells. Oxid Med Cell Longev. 2013;2013:423965.

    Article  Google Scholar 

  28. Zhu Y, Lei Q, Li D, Zhang Y, Jiang X, Hu Z, et al. Proteomic and biochemical analyses reveal a novel mechanism for promoting protein ubiquitination and degradation by UFBP1, a key component of ufmylation. J Proteome Res. 2018;17:1509–20.

    Article  CAS  Google Scholar 

  29. Gambardella V, Gimeno-Valiente F, Tarazona N, Ciarpaglini CM, Roda D, Fleitas T, et al. NRF2 through RPS6 activation is related to anti-HER2 drug resistance in HER2-amplified gastric cancer. Clin Cancer Res. 2019;25:1639–49.

    Article  CAS  Google Scholar 

  30. Homma S, Ishii Y, Morishima Y, Yamadori T, Matsuno Y, Haraguchi N, et al. Nrf2 enhances cell proliferation and resistance to anticancer drugs in human lung cancer. Clin Cancer Res. 2009;15:3423–32.

    Article  CAS  Google Scholar 

  31. Roh JL, Kim EH, Jang H, Shin D. Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis. Redox Biol. 2017;11:254–62.

    Article  CAS  Google Scholar 

  32. Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Tissue-based map of the human proteome. Science. 2015;347:1260419.

    Article  Google Scholar 

  33. Lin JX, Xie XS, Weng XF, Qiu SL, Yoon C, Lian NZ, et al. UFM1 suppresses invasive activities of gastric cancer cells by attenuating the expression of PDK1 through PI3K/AKT signaling. J Exp Clin Cancer Res. 2019;38:410.

    Article  Google Scholar 

  34. Wang S, Juan J, Zhang Z, Du Y, Xu Y, Tong J, et al. Inhibition of the deubiquitinase USP5 leads to c-Maf protein degradation and myeloma cell apoptosis. Cell Death Dis. 2017;8:e3058.

    Article  CAS  Google Scholar 

  35. Zhao Y, Liu D, Proksch P, Yu S, Lin W. Isocoumarin derivatives from the sponge-associated fungus Peyronellaea glomerata with antioxidant activities. Chem Biodivers. 2016;13:1186–93.

    Article  CAS  Google Scholar 

  36. Xu G, Jiang X, Jaffrey SR. A mental retardation-linked nonsense mutation in cereblon is rescued by proteasome inhibition. J Biol Chem. 2013;288:29573–85.

    Article  CAS  Google Scholar 

  37. Hou X, Si J, Ren H, Chen D, Wang H, Ying Z, et al. Parkin represses 6-hydroxydopamine-induced apoptosis via stabilizing scaffold protein p62 in PC12 cells. Acta Pharm Sin. 2015;36:1300–7.

    Article  CAS  Google Scholar 

  38. Duan Q, Li D, Xiong L, Chang Z, Xu G. SILAC quantitative proteomics and biochemical analyses reveal a novel molecular mechanism by which ADAM12S promotes the proliferation, migration, and invasion of small cell lung cancer cells through upregulating hexokinase 1. J Proteome Res. 2019;18:2903–14.

    Article  CAS  Google Scholar 

  39. Hu SQ, Wang R, Cui W, Mak SH, Li G, Hu YJ, et al. Dimeric bis (heptyl)-cognitin blocks Alzheimer’s β-amyloid neurotoxicity via the inhibition of Aβ fibrils formation and disaggregation of preformed fibrils. CNS Neurosci Ther. 2015;21:953–61.

    Article  CAS  Google Scholar 

  40. Guo DK, Zhu Y, Sun HY, Xu XY, Zhang S, Hao ZB, et al. Pharmacological activation of REV-ERBα represses LPS-induced microglial activation through the NF-κB pathway. Acta Pharm Sin. 2019;40:26–34.

    Article  CAS  Google Scholar 

  41. Washington K. 7th edition of the AJCC cancer staging manual: stomach. Ann Surg Oncol. 2010;17:3077–9.

    Article  Google Scholar 

  42. Yakirevich E, Resnick MB, Mangray S, Wheeler M, Jackson CL, Lombardo KA, et al. Oncogenic ALK fusion in rare and aggressive subtype of colorectal adenocarcinoma as a potential therapeutic target. Clin Cancer Res. 2016;22:3831–40.

    Article  CAS  Google Scholar 

  43. Perez-Riverol Y, Csordas A, Bai J, Bernal-Llinares M, Hewapathirana S, Kundu DJ, et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 2019;47:D442–50.

Download references

Acknowledgements

We are grateful to Drs. Xinliang Mao (Soochow University) and Siwang Yu (Peking University) for kindly providing plasmids and to Dr. Guanghui Wang (Soochow University) for fruitful discussion. MS analyses were performed at the Mass Spectrometry core facility of the Medical School of Soochow University. This work was supported by the National Key R&D Program of China (2019YFA0802400), the National Natural Science Foundation of China (31700722 & 31971353), Suzhou Bureau of Science and Technology (Basic Research in Medical and Health Sciences, SYS201718), Talent Program in Six Major Disciplines in Jiangsu Province (SWYY-080), Open Project of Jiangsu Key Laboratory of Neuropsychiatric Diseases (KIS1722), Open Project Program of the State Key Laboratory of Proteomics (SKLP-O201905), Postgraduate Research & Practice Innovation Program of Jiangsu Province (KYCX17_2040), Natural Science Foundation of Jiangsu Higher Education Institutes of China (17KJA180010), National Center for International Research (2017B01012), a project funded by the Priority Academic Program Development (PAPD) of Jiangsu Higher Education Institutions.

Author information

Authors and Affiliations

Authors

Contributions

ZH and GX conceived and designed the experiments; ZH, XW, DL, LC, and HC performed the experiments and analyzed the data; ZH and GX wrote the paper; all authors revised the paper.

Corresponding author

Correspondence to Guoqiang Xu.

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

Hu, Z., Wang, X., Li, D. et al. UFBP1, a key component in ufmylation, enhances drug sensitivity by promoting proteasomal degradation of oxidative stress-response transcription factor Nrf2. Oncogene 40, 647–662 (2021). https://doi.org/10.1038/s41388-020-01551-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-020-01551-1

This article is cited by

Search

Quick links