Skip to main content

Advertisement

Log in

Identification and development of a novel risk model based on cuproptosis-associated RNA methylation regulators for predicting prognosis and characterizing immune status in hepatocellular carcinoma

  • Original Article
  • Published:
Hepatology International Aims and scope Submit manuscript

Abstract

Background

Cuproptosis, a novel cell death caused by excess copper, is quite obscure in hepatocellular carcinoma (HCC) and needs more investigation.

Methods

RNA-seq and clinical data of HCC patients TCGA database were analyzed to establish a predictive model through LASSO Cox regression analysis. External dataset ICGC was used for the verification. GSEA and CIBERSORT were applied to investigate the molecular mechanisms and immune microenvironment of HCC. Cuproptosis induced by elesclomol was confirmed via various in vitro experiments. The expression of prognostic genes was verified in HCC tissues using qRT-PCR analysis.

Results

Initially, 18 cuproptosis-associated RNA methylation regulators (CARMRs) were selected for prognostic analysis. A nine-gene signature was created by applying the LASSO Cox regression method. Survival and ROC assays were carried out to validate the model using TCGA and ICGC database. Moreover, there exhibited obvious differences in drug sensitivity in terms of common drugs. A higher tumor mutation burden was shown in the high-risk group. Additionally, significant discrepancies were found between the two groups in metabolic pathways and RNA processing via GSEA analysis. Meanwhile, CIBERSORT analysis indicated different infiltrating levels of various immune cells between the two groups. Elesclomol treatment caused a unique form of programmed cell death accompanied by loss of lipoylated mitochondrial proteins and Fe–S cluster protein. The results of qRT-PCR indicated that most prognostic genes were differentially expressed in the HCC tissues.

Conclusion

Overall, our predictive signature displayed potential value in the prediction of overall survival of HCC patients and might provide valuable clues for personalized therapies.

Graphical abstract

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Availability of data and materials

Most of the datasets used and analyzed in the study are publicly available data from TCGA and ICGC database. Further inquiries can be directed to the corresponding authors.

References

  1. Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA, et al. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology. 2020;159:335-349.e315

    Article  Google Scholar 

  2. Franses JW, Zhu AX. Neoadjuvant approaches in hepatocellular carcinoma: there’s no time like the present. Clin Cancer Res. 2022;28:2738–2743

    Article  CAS  Google Scholar 

  3. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Prim. 2021;7:6

    Article  Google Scholar 

  4. Schulze K, Imbeaud S, Letouzé E, Alexandrov LB, Calderaro J, Rebouissou S, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet. 2015;47:505–511

    Article  CAS  Google Scholar 

  5. Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 2012;44:694–698

    Article  CAS  Google Scholar 

  6. Strasser A, Vaux DL. Cell death in the origin and treatment of cancer. Mol Cell. 2020;78:1045–1054

    Article  CAS  Google Scholar 

  7. Qi X, Li Q, Che X, Wang Q, Wu G. Application of regulatory cell death in cancer: based on targeted therapy and immunotherapy. Front Immunol. 2022;13: 837293

    Article  CAS  Google Scholar 

  8. Koren E, Fuchs Y. Modes of regulated cell death in cancer. Cancer Discov. 2021;11:245–265

    Article  CAS  Google Scholar 

  9. Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022;375:1254–1261

    Article  CAS  Google Scholar 

  10. Li Z, Zhang H, Wang X, Wang Q, Xue J, Shi Y, et al. Identification of cuproptosis-related subtypes, characterization of tumor microenvironment infiltration, and development of a prognosis model in breast cancer. Front Immunol. 2022;13: 996836

    Article  CAS  Google Scholar 

  11. Zhang G, Chen X, Fang J, Tai P, Chen A, Cao K. Cuproptosis status affects treatment options about immunotherapy and targeted therapy for patients with kidney renal clear cell carcinoma. Front Immunol. 2022;13: 954440

    Article  CAS  Google Scholar 

  12. Yang M, Zheng H, Xu K, Yuan Q, Aihaiti Y, Cai Y, et al. A novel signature to guide osteosarcoma prognosis and immune microenvironment: cuproptosis-related lncRNA. Front Immunol. 2022;13: 919231

    Article  CAS  Google Scholar 

  13. Zhang C, Zeng Y, Guo X, Shen H, Zhang J, Wang K, et al. Pan-cancer analyses confirmed the cuproptosis-related gene FDX1 as an immunotherapy predictor and prognostic biomarker. Front Genet. 2022;13: 923737

    Article  CAS  Google Scholar 

  14. Yang B, Wang JQ, Tan Y, Yuan R, Chen ZS, Zou C. RNA methylation and cancer treatment. Pharmacol Res. 2021;174: 105937

    Article  CAS  Google Scholar 

  15. Lan Q, Liu PY, Haase J, Bell JL, Hüttelmaier S, Liu T. The critical role of RNA m(6)A methylation in cancer. Cancer Res. 2019;79:1285–1292

    Article  CAS  Google Scholar 

  16. He R, Man C, Huang J, He L, Wang X, Lang Y, et al. Identification of RNA methylation-related lncRNAs signature for predicting hot and cold tumors and prognosis in colon cancer. Front Genet. 2022;13: 870945

    Article  CAS  Google Scholar 

  17. Lan Q, Liu PY, Bell JL, Wang JY, Hüttelmaier S, Zhang XD, et al. The emerging roles of RNA m(6)A methylation and demethylation as critical regulators of tumorigenesis, drug sensitivity, and resistance. Cancer Res. 2021;81:3431–3440

    Article  CAS  Google Scholar 

  18. Gu Y, Wu X, Zhang J, Fang Y, Pan Y, Shu Y, et al. The evolving landscape of N(6)-methyladenosine modification in the tumor microenvironment. Mol Ther. 2021;29:1703–1715

    Article  CAS  Google Scholar 

  19. Huang H, Weng H, Chen J. The biogenesis and precise control of RNA m(6)A methylation. Trends Genet. 2020;36:44–52

    Article  CAS  Google Scholar 

  20. Einstein JM, Perelis M, Chaim IA, Meena JK, Nussbacher JK, Tankka AT, et al. Inhibition of YTHDF2 triggers proteotoxic cell death in MYC-driven breast cancer. Mol Cell. 2021;81:3048-3064.e3049

    Article  CAS  Google Scholar 

  21. Woo HH, Chambers SK. Human ALKBH3-induced m(1)A demethylation increases the CSF-1 mRNA stability in breast and ovarian cancer cells. Biochim Biophys Acta Gene Regul Mech. 2019;1862:35–46

    Article  CAS  Google Scholar 

  22. Chen X, Li A, Sun BF, Yang Y, Han YN, Yuan X, et al. 5-Methylcytosine promotes pathogenesis of bladder cancer through stabilizing mRNAs. Nat Cell Biol. 2019;21:978–990

    Article  CAS  Google Scholar 

  23. Han H, Zheng S, Lin S. N(7)-methylguanosine (m(7)G) tRNA modification: a novel autophagy modulator in cancer. Autophagy. 2022:1–3.

  24. Tibshirani R. The lasso method for variable selection in the Cox model. Stat Med. 1997;16:385–395

    Article  CAS  Google Scholar 

  25. Blanche P, Dartigues JF, Jacqmin-Gadda H. Estimating and comparing time-dependent areas under receiver operating characteristic curves for censored event times with competing risks. Stat Med. 2013;32:5381–5397

    Article  Google Scholar 

  26. Eaton A, Therneau T, Le-Rademacher J. Designing clinical trials with (restricted) mean survival time endpoint: practical considerations. Clin Trials. 2020;17:285–294

    Article  Google Scholar 

  27. Mi JX, Zhang YN, Lai Z, Li W, Zhou L, Zhong F: Principal component analysis based on nuclear norm minimization. Neural Netw 2019;118:1–16.

    Article  Google Scholar 

  28. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–15550

    Article  CAS  Google Scholar 

  29. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12:453–457

    Article  CAS  Google Scholar 

  30. Geeleher P, Cox N, Huang RS. pRRophetic: an R package for prediction of clinical chemotherapeutic response from tumor gene expression levels. PLoS ONE. 2014;9: e107468

    Article  Google Scholar 

  31. Hsu CY, Liu PH, Hsia CY, Lee YH, Al Juboori A, Lee RC, et al. Nomogram of the Barcelona clinic liver cancer system for individual prognostic prediction in hepatocellular carcinoma. Liver Int. 2016;36:1498–1506

    Article  Google Scholar 

  32. Sarkar FH, Li Y. Using chemopreventive agents to enhance the efficacy of cancer therapy. Cancer Res. 2006;66:3347–3350

    Article  CAS  Google Scholar 

  33. Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 2008;27:5904–5912

    Article  CAS  Google Scholar 

  34. Xi Q, Zhang J, Yang G, Zhang L, Chen Y, Wang C, et al. Restoration of miR-340 controls pancreatic cancer cell CD47 expression to promote macrophage phagocytosis and enhance antitumor immunity. J Immunother Cancer. 2020,8.

  35. Ercal N, Gurer-Orhan H, Aykin-Burns N. Toxic metals and oxidative stress part I: mechanisms involved in metal-induced oxidative damage. Curr Top Med Chem. 2001;1:529–539

    Article  CAS  Google Scholar 

  36. Fan X, Li Y, Yi X, Chen G, Jin S, Dai Y, et al. Epigenome-wide DNA methylation profiling of portal vein tumor thrombosis (PVTT) tissues in hepatocellular carcinoma patients. Neoplasia. 2020;22:630–643

    Article  CAS  Google Scholar 

  37. Wong MC, Jiang JY, Goggins WB, Liang M, Fang Y, Fung FD, et al. International incidence and mortality trends of liver cancer: a global profile. Sci Rep. 2017;7:45846

    Article  CAS  Google Scholar 

  38. Constantinidou A, Alifieris C, Trafalis DT. Targeting programmed cell death-1 (PD-1) and ligand (PD-L1): a new era in cancer active immunotherapy. Pharmacol Ther. 2019;194:84–106

    Article  CAS  Google Scholar 

  39. Song W, Shen L, Wang Y, Liu Q, Goodwin TJ, Li J, et al. Synergistic and low adverse effect cancer immunotherapy by immunogenic chemotherapy and locally expressed PD-L1 trap. Nat Commun. 2018;9:2237

    Article  Google Scholar 

  40. Holzer K, Ori A, Cooke A, Dauch D, Drucker E, Riemenschneider P, et al. Nucleoporin Nup155 is part of the p53 network in liver cancer. Nat Commun. 2019;10:2147

    Article  Google Scholar 

  41. Gray LR, Tompkins SC, Taylor EB. Regulation of pyruvate metabolism and human disease. Cell Mol Life Sci. 2014;71:2577–2604

    Article  CAS  Google Scholar 

  42. Li SR, Bu LL, Cai L. Cuproptosis: lipoylated TCA cycle proteins-mediated novel cell death pathway. Signal Transduct Target Ther. 2022;7:158

    Article  CAS  Google Scholar 

  43. Cronan JE. Progress in the enzymology of the mitochondrial diseases of lipoic acid requiring enzymes. Front Genet. 2020;11:510

    Article  CAS  Google Scholar 

  44. Hanna A, Shevde LA. Hedgehog signaling: modulation of cancer properies and tumor mircroenvironment. Mol Cancer. 2016;15:24

    Article  Google Scholar 

  45. Wei Y, Lao XM, Xiao X, Wang XY, Wu ZJ, Zeng QH, et al. Plasma cell polarization to the immunoglobulin G phenotype in hepatocellular carcinomas involves epigenetic alterations and promotes hepatoma progression in mice. Gastroenterology. 2019;156(1890–1904): e1816

    Google Scholar 

  46. Liu X, Wu S, Yang Y, Zhao M, Zhu G, Hou Z. The prognostic landscape of tumor-infiltrating immune cell and immunomodulators in lung cancer. Biomed Pharmacother. 2017;95:55–61

    Article  CAS  Google Scholar 

  47. Shirota H, Klinman DM, Ito SE, Ito H, Kubo M, Ishioka C. IL4 from T follicular helper cells downregulates antitumor immunity. Cancer Immunol Res. 2017;5:61–71

    Article  CAS  Google Scholar 

  48. Tu JF, Ding YH, Ying XH, Wu FZ, Zhou XM, Zhang DK, et al. Regulatory T cells, especially ICOS(+) FOXP3(+) regulatory T cells, are increased in the hepatocellular carcinoma microenvironment and predict reduced survival. Sci Rep. 2016;6:35056

    Article  CAS  Google Scholar 

  49. Cho Y, Miyamoto M, Kato K, Fukunaga A, Shichinohe T, Kawarada Y, et al. CD4+ and CD8+ T cells cooperate to improve prognosis of patients with esophageal squamous cell carcinoma. Cancer Res. 2003;63:1555–1559

    CAS  Google Scholar 

  50. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–949

    Article  CAS  Google Scholar 

  51. Kubo H, Mensurado S, Goncalves-Sousa N, Serre K, Silva-Santos B. Primary tumors limit metastasis formation through induction of IL15-mediated cross-talk between patrolling monocytes and NK cells. Cancer Immunol Res. 2017;5:812–820

    Article  CAS  Google Scholar 

  52. Wang H, Wu J, Ling R, Li F, Yang Q, He J, et al. Fibroblast-derived LPP as a biomarker for treatment response and therapeutic target in gastric cancer. Mol Ther Oncolytics. 2022;24:547–560

    Article  CAS  Google Scholar 

  53. Komi DEA, Redegeld FA. Role of mast cells in shaping the tumor microenvironment. Clin Rev Allergy Immunol. 2020;58:313–325

    Article  CAS  Google Scholar 

  54. Chen Y, Li C, Xie H, Fan Y, Yang Z, Ma J, et al. Infiltrating mast cells promote renal cell carcinoma angiogenesis by modulating PI3K→AKT→GSK3beta→AM signaling. Oncogene. 2017;36:2879–2888

    Article  CAS  Google Scholar 

  55. Xiong D, Wang Y, You M. Tumor intrinsic immunity related proteins may be novel tumor suppressors in some types of cancer. Sci Rep. 2019;9:10918

    Article  Google Scholar 

  56. Xu F, Jin T, Zhu Y, Dai C. Immune checkpoint therapy in liver cancer. J Exp Clin Cancer Res. 2018;37:110

    Article  Google Scholar 

Download references

Funding

This work was supported by National Key Research and Development Project (2017YFC0110802), Zhejiang province Key Research and Development Project (2020C01059), National Natural Science Foundation of China (81872297, and 81874059), Zhejiang Engineering Research Center of Cognitive Healthcare (2017E10011), National Natural Science Foundation of China (82102105), Natural Science Foundation of Zhejiang Province (LQ22H160017) and China Postdoctoral Science Foundation (2021M702825).

Author information

Authors and Affiliations

Authors

Contributions

HL, JY, and XXF conceived and designed the analysis. SXJ, PC, YYZ, and GQC collected the data. DGL, ZQS, and XLL performed analysis. DGL, ZQS, and XLL wrote the paper. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Xiaoxiao Fan, Jing Yang or Hui Lin.

Ethics declarations

Conflict of interest

Duguang Li, Zhaoqi Shi, Xiaolong Liu, Shengxi Jin, Peng Chen, Yiyin Zhang, Guoqiao Chen, Xiaoxiao Fan, Jing Yang and Hui Lin have no relevant financial or non-financial interests to disclose.

Ethics approval and consent to participate

The study was conducted in accordance with the Declaration of Helsinki, and was approved by the Institutional Review Board of SRRSH (ethical code: 20210729-282) and written informed consent was obtained from all patients.

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

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, D., Shi, Z., Liu, X. et al. Identification and development of a novel risk model based on cuproptosis-associated RNA methylation regulators for predicting prognosis and characterizing immune status in hepatocellular carcinoma. Hepatol Int 17, 112–130 (2023). https://doi.org/10.1007/s12072-022-10460-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12072-022-10460-2

Keywords

Navigation