Skip to main content
Log in

DLAT as a Cuproptosis Promoter and a Molecular Target of Elesclomol in Hepatocellular Carcinoma

  • Published:
Current Medical Science Aims and scope Submit manuscript

Abstract

Objective

Cuproptosis is a novel cell death pathway that was newly discovered in early 2022. However, cuproptosis is still in its infancy in many respects and warrants further research in hepatocellular carcinoma (HCC). This study aimed to analyze the mechanism of cuprptosis in HCC.

Methods

Herein, the tumor microenvironment infiltration landscape of molecular subtypes was illustrated using GSVA, ssGSEA, TIMER, CIBERSORT, and ESTIMATE algorithms based on the expression profile of cuproptosis-related genes (CRGs) from TCGA and GEO databases. Then, the least absolute shrinkage and selection operator regression method was applied to construct a cuproptosis signature to quantify the cuproptosis profile of HCC. Further, we explored the expression of three hub CRGs in cell lines and clinical patient tissues of HCC by Western blotting, qRT-PCR and immunohistochemistry. Finally, we examined the function of dihydrolipoamide S-acetyltransferase (DLAT) in cuproptosis in HCC by loss-of-function strategy, Western blotting and CCK8 assay.

Results

Three distinct molecular subtypes were identified. Cluster 2 had the greatest infiltration of immune cells with best prognosis. The cuproptosis signature was indicative of tumor subtype, immunity, and prognosis for HCC, and specifically, a low cuproptosis score foreshadowed good prognosis. DLAT was highly expressed in liver cancer cell lines and HCC tissues and positively correlated with clinical stage and grade. We also found that potent copper ionophore elesclomol could induce cuproptosis in a copper-dependent manner. Selective Cu++ chelator ammonium tetrathiomolybdate and downregulating DLAT expression by siRNA could effectively inhibit cuproptosis.

Conclusion

Cuproptosis and DLAT as a promising biomarker could help to determine the prognosis of HCC and may offer novel insights for effective treatment.

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.

Similar content being viewed by others

References

  1. Cao M, Luo X, Wu K, et al. Targeting lysosomes in human disease: from basic research to clinical applications. Signal Transduct Target Ther, 2021,6(1):379

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Tsvetkov P, Coy S, Petrova B, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science, 2022,375(6586):1254–1261

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lin S, Yang H. Ovarian cancer risk according to circulating zinc and copper concentrations: A metaanalysis and Mendelian randomization study. Clin Nutr, 2021,40(4):2464–2468

    Article  CAS  PubMed  Google Scholar 

  4. Xiao Y, Chen DI, Zhang X, et al. Molecular study on copper-mediated tumor proteasome inhibition and cell death. Int J Oncol, 2010,37(1):81–87

    CAS  PubMed  Google Scholar 

  5. Blockhuys S, Celauro E, Hildesjö C, et al. Defining the human copper proteome and analysis of its expression variation in cancers. Metallomics, 2017,9(2):112–123

    Article  CAS  PubMed  Google Scholar 

  6. Ishida S, Andreux P, Poitry-Yamate C, et al. Bioavailable copper modulates oxidative phosphorylation and growth of tumors. Proc Natl Acad Sci USA, 2013,110(48): 19507–19512

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ge EJ, Bush AI, Casini A, et al. Connecting copper and cancer: from transition metal signalling to metalloplasia. Nat Rev Cancer, 2022,22(2):102–113

    Article  CAS  PubMed  Google Scholar 

  8. Zuo XL, Chen JM, Zhou X, et al. Levels of selenium, zinc, copper, and antioxidant enzyme activity in patients with leukemia. Biol Trace Elem Res, 2006,114(1–3):41–53

    Article  CAS  PubMed  Google Scholar 

  9. Lutsenko S. Human copper homeostasis: a network of interconnected pathways. Curr Opin Chem Biol, 2010,14(2):211–217

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Jiang Y, Huo Z, Qi X, et al. Copper-induced tumor cell death mechanisms and antitumor theragnostic applications of copper complexes. Nanomedicine (Lond), 2022,17(5):303–324

    Article  CAS  PubMed  Google Scholar 

  11. Voli F, Valli E, Lerra L, et al. Intratumoral Copper Modulates PD-L1 Expression and Influences Tumor Immune Evasion. Cancer Res, 2020,80(19):4129–4144

    Article  CAS  PubMed  Google Scholar 

  12. Liao Y, Zhao J, Bulek K, et al. Inflammation mobilizes copper metabolism to promote colon tumorigenesis via an IL-17-STEAP4-XIAP axis. Nat Commun, 2020,11(1):900

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Chan N, Willis A, Kornhauser N, et al. Influencing the Tumor Microenvironment: A Phase II Study of Copper Depletion Using Tetrathiomolybdate in Patients with Breast Cancer at High Risk for Recurrence and in Preclinical Models of Lung Metastases. Clin Cancer Res, 2017,23(3):666–676

    Article  CAS  PubMed  Google Scholar 

  14. Gupte A, Mumper RJ. Elevated copper and oxidative stress in cancer cells as a target for cancer treatment. Cancer Treat Rev, 2009,35(1):32–46

    Article  CAS  PubMed  Google Scholar 

  15. Yoshii J, Yoshiji H, Kuriyama S, et al. The copper-chelating agent, trientine, suppresses tumor development and angiogenesis in the murine hepatocellular carcinoma cells. Int J Cancer, 2001,94(6):768–773

    Article  CAS  PubMed  Google Scholar 

  16. Chen D, Cui QC, Yang H, et al. Disulfiram, a clinically used anti-alcoholism drug and copper-binding agent, induces apoptotic cell death in breast cancer cultures and xenografts via inhibition of the proteasome activity. Cancer Res, 2006,66(21):10425–10433

    Article  CAS  PubMed  Google Scholar 

  17. Karginova O, Weekley CM, Raoul A, et al. Inhibition of Copper Transport Induces Apoptosis in Triple-Negative Breast Cancer Cells and Suppresses Tumor Angiogenesis. Mol Cancer Ther, 2019,18(5):873–885

    Article  CAS  PubMed  Google Scholar 

  18. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin, 2021,71(3):209–249

    Article  PubMed  Google Scholar 

  19. Luo XY, Wu KM, He XX. Advances in drug development for hepatocellular carcinoma: clinical trials and potential therapeutic targets. J Exp Clin Cancer Res, 2021,40(1):172

    Article  PubMed  PubMed Central  Google Scholar 

  20. Lv H, Liu X, Zeng X, et al. Comprehensive Analysis of Cuproptosis-Related Genes in Immune Infiltration and Prognosis in Melanoma. Front Pharmacol, 2022,13:930041

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chen Q, Wang Y, Yang L, et al. PM2.5 promotes NSCLC carcinogenesis through translationally and transcriptionally activating DLAT-mediated glycolysis reprograming. J Exp Clin Cancer Res, 2022,41(1):229

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Goh WQ, Ow GS, Kuznetsov VA, et al. DLAT subunit of the pyruvate dehydrogenase complex is upregulated in gastric cancer-implications in cancer therapy. Am J Transl Res, 2015,7(6):1140–1151

    PubMed  PubMed Central  Google Scholar 

  23. Stone S, Jiang P, Dayananth P, et al. Complex structure and regulation of the P16 (MTS1) locus. Cancer Res, 1995,55(14):2988–2994

    CAS  PubMed  Google Scholar 

  24. Song W, Ren J, Xiang R, et al. Identification of pyroptosis-related subtypes, the development of a prognosis model, and characterization of tumor microenvironment infiltration in colorectal cancer. Oncoimmunology, 2021,10(1):1987636

    Article  PubMed  PubMed Central  Google Scholar 

  25. Kim Y, Kang JW, Kang J, et al. Novel deep learning-based survival prediction for oral cancer by analyzing tumor-infiltrating lymphocyte profiles through CIBER-SORT. Oncoimmunology, 2021,10(1):1904573

    Article  PubMed  PubMed Central  Google Scholar 

  26. Wang T, Dai L, Shen S, et al. Comprehensive Molecular Analyses of a Macrophage-Related Gene Signature With Regard to Prognosis, Immune Features, and Biomarkers for Immunotherapy in Hepatocellular Carcinoma Based on WGCNA and the LASSO Algorithm. Front Immunol, 2022,13:843408

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Tang Z, Kang B, Li C, et al. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res, 2019,47(W1):W556–W560

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Li T, Fan J, Wang B, et al. TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. Cancer Res, 2017,77(21):e108–e110

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Li SR, Bu LL, Cai L. Cuproptosis: lipoylated TCA cycle proteins-mediated novel cell death pathway. Signal Transduct Target Ther, 2022,7(1):158

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Buccarelli M, D’Alessandris Q, Matarrese P, et al. Elesclomol-induced increase of mitochondrial reactive oxygen species impairs glioblastoma stem-like cell survival and tumor growth. J Exp Clin Cancer Res, 2021,40(1):228

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wangpaichitr M, Wu C, You M, et al. N′,N′-Dimethyl-N′,N′-bis(phenylcarbonothioyl) Propanedihydrazide (Elesclomol) Selectively Kills Cisplatin Resistant Lung Cancer Cells through Reactive Oxygen Species (ROS). Cancers (Basel), 2009,1(1):23–38

    Article  CAS  PubMed  Google Scholar 

  32. Gao W, Huang Z, Duan J, et al. Elesclomol induces copper-dependent ferroptosis in colorectal cancer cells via degradation of ATP7A. Mol Oncol, 2021,15(12): 3527–3544

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. O’Day S, Eggermont A, Chiarion-Sileni V, et al. Final results of phase III SYMMETRY study: randomized, double-blind trial of elesclomol plus paclitaxel versus paclitaxel alone as treatment for chemotherapy-naive patients with advanced melanoma. J Clin Oncol, 2013,31(9):1211–1218

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Ying Chang or Xing-xing He.

Additional information

Conflict of Interest Statement

The authors have no conflict of interest.

This research was financially supported by grants from the National Natural Science Foundation of China (No. 82073095, No. 82172938 and No. 81670554) and Science and Technology Innovation Cultivation Fund of Zhongnan Hospital of Wuhan University (No. CXPY2020042).

Supplementary data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gao, F., Yuan, Y., Ding, Y. et al. DLAT as a Cuproptosis Promoter and a Molecular Target of Elesclomol in Hepatocellular Carcinoma. CURR MED SCI 43, 526–538 (2023). https://doi.org/10.1007/s11596-023-2755-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11596-023-2755-0

Key words

Navigation