Abstract
Cuproptosis is characterized by the aggregation of lipoylated enzymes of the tricarboxylic acid cycle and subsequent loss of iron-sulfur cluster proteins as a unique copper-dependent form of regulated cell death. As dysregulation of copper homeostasis can induce cuproptosis, there is emerging interest in exploiting cuproptosis for cancer therapy. However, the molecular drivers of cancer cell evasion of cuproptosis were previously undefined. Here, we found that cuproptosis activates the Wnt/β-catenin pathway. Mechanistically, copper binds PDK1 and promotes its interaction with AKT, resulting in activation of the Wnt/β-catenin pathway and cancer stem cell (CSC) properties. Notably, aberrant activation of Wnt/β-catenin signaling conferred resistance of CSCs to cuproptosis. Further studies showed the β-catenin/TCF4 transcriptional complex directly binds the ATP7B promoter, inducing its expression. ATP7B effluxes copper ions, reducing intracellular copper and inhibiting cuproptosis. Knockdown of TCF4 or pharmacological Wnt/β-catenin blockade increased the sensitivity of CSCs to elesclomol-Cu-induced cuproptosis. These findings reveal a link between copper homeostasis regulated by the Wnt/β-catenin pathway and cuproptosis sensitivity, and suggest a precision medicine strategy for cancer treatment through selective cuproptosis induction.
This is a preview of subscription content, access via your institution
Access options
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
Data availability
RNA-seq and CUT&Tag data have been deposited in GEO under the following accession numbers: GSE248083, GSE248084, and GSE248246.
References
Ge E, Bush A, Casini A, Cobine P, Cross J, DeNicola G, et al. Connecting copper and cancer: from transition metal signalling to metalloplasia. Nat Rev Cancer. 2022;22:102–13.
Brady DC, Crowe MS, Turski ML, Hobbs GA, Yao X, Chaikuad A, et al. Copper is required for oncogenic BRAF signalling and tumorigenesis. Nature. 2014;509:492–96.
Das A, Ash D, Fouda A, Sudhahar V, Kim Y, Hou Y, et al. Cysteine oxidation of copper transporter CTR1 drives VEGFR2 signalling and angiogenesis. Nat Cell Biol. 2022;24:35–50.
Tsang T, Posimo J, Gudiel A, Cicchini M, Feldser D, Brady D. Copper is an essential regulator of the autophagic kinases ULK1/2 to drive lung adenocarcinoma. Nat Cell Biol. 2020;22:412–24.
Ramchandani D, Berisa M, Tavarez D, Li Z, Miele M, Bai Y, et al. Copper depletion modulates mitochondrial oxidative phosphorylation to impair triple negative breast cancer metastasis. Nat Commun. 2021;12:7311.
Chen L, Min J, Wang F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct Target Ther. 2022;7:378.
Wang W, Mo W, Hang Z, Huang Y, Yi H, Sun Z, et al. Cuproptosis: harnessing transition metal for cancer therapy. ACS Nano. 2023;17:19581–99.
Xie J, Yang Y, Gao Y, He J. Cuproptosis: mechanisms and links with cancers. Mol Cancer. 2023;22:46.
Ohgami R, Campagna D, McDonald A, Fleming M. The Steap proteins are metalloreductases. Blood. 2006;108:1388–94.
Su Y, Zhang X, Li S, Xie W, Guo J. Emerging roles of the copper-CTR1 axis in tumorigenesis. Mol Cancer Res. 2022;20:1339–53.
La Fontaine S, Mercer J. Trafficking of the copper-ATPases, ATP7A and ATP7B: role in copper homeostasis. Arch Biochem Biophys. 2007;463:149–67.
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;375:1254–61.
Kahlson M, Dixon S. Copper-induced cell death. Science. 2022;375:1231–32.
Xiong C, Ling H, Hao Q, Zhou X. Cuproptosis: p53-regulated metabolic cell death? Cell Death Differ. 2023;30:876–84.
Schulz V, Basu S, Freibert S-A, Webert H, Boss L, Mühlenhoff U, et al. Functional spectrum and specificity of mitochondrial ferredoxins FDX1 and FDX2. Nat Chem Biol. 2023;19:206–17.
Sun L, Zhang Y, Yang B, Sun S, Zhang P, Luo Z, et al. Lactylation of METTL16 promotes cuproptosis via mA-modification on FDX1 mRNA in gastric cancer. Nat Commun. 2023;14:6523.
Cobine P, Brady D. Cuproptosis: cellular and molecular mechanisms underlying copper-induced cell death. Mol Cell. 2022;82:1786–87.
Jiang X, Ke J, Jia L, An X, Ma H, Li Z, et al. A novel cuproptosis-related gene signature of prognosis and immune microenvironment in head and neck squamous cell carcinoma cancer. J Cancer Res Clin Oncol. 2023;149:203–18.
Nusse R, Clevers H. Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 2017;169:985–99.
Bugter J, Fenderico N, Maurice M. Mutations and mechanisms of WNT pathway tumour suppressors in cancer. Nat Rev Cancer. 2021;21:5–21.
Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther. 2020;5:8.
Wang Y, Zheng L, Shang W, Yang Z, Li T, Liu F, et al. Wnt/beta-catenin signaling confers ferroptosis resistance by targeting GPX4 in gastric cancer. Cell Death Differ. 2022;29:2190–202.
Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12:31–46.
Zou Y, Zheng S, Xie X, Ye F, Hu X, Tian Z, et al. N6-methyladenosine regulated FGFR4 attenuates ferroptotic cell death in recalcitrant HER2-positive breast cancer. Nat Commun. 2022;13:2672.
Huster D, Purnat TD, Burkhead JL, Ralle M, Fiehn O, Stuckert F, et al. High copper selectively alters lipid metabolism and cell cycle machinery in the mouse model of Wilson disease. J Biol Chem. 2007;282:8343–55.
Voli F, Valli E, Lerra L, Kimpton K, Saletta F, Giorgi F, et al. Intratumoral copper Modulates PD-L1 expression and influences tumor immune evasion. Cancer Res. 2020;80:4129–44.
Guo J, Cheng J, Zheng N, Zhang X, Dai X, Zhang L, et al. Copper promotes tumorigenesis by activating the PDK1-AKT oncogenic pathway in a copper transporter 1 dependent manner. Adv Sci. 2021;8:e2004303.
Lu J, Liu X, Li X, Li H, Shi L, Xia X, et al. Copper regulates the host innate immune response against bacterial infection via activation of ALPK1 kinase. Proc Natl Acad Sci USA. 2024;121:e2311630121.
Zulkifli M, Spelbring AN, Zhang Y, Soma S, Chen S, Li L, et al. FDX1-dependent and independent mechanisms of elesclomol-mediated intracellular copper delivery. Proc Natl Acad Sci USA. 2023;120:e2216722120.
Xiao Z, Brose J, Schimo S, Ackland SM, La Fontaine S, Wedd AG. Unification of the copper(I) binding affinities of the metallo-chaperones Atx1, Atox1, and related proteins: detection probes and affinity standards. J Biol Chem. 2011;286:11047–55.
Chen L, Li N, Zhang M, Sun M, Bian J, Yang B, et al. APEX2-based proximity labeling of Atox1 identifies CRIP2 as a nuclear copper-binding protein that regulates autophagy activation. Angew Chem Int Ed. 2021;60:25346–55.
Wang Z, Wu VH, Allevato MM, Gilardi M, He Y, Luis Callejas-Valera J, et al. Syngeneic animal models of tobacco-associated oral cancer reveal the activity of in situ anti-CTLA-4. Nat Commun. 2019;10:5546.
Chen D, Wang C. Targeting cancer stem cells in squamous cell carcinoma. Precis Clin Med. 2019;2:152–65.
Chen Y, Zhao H, Liang W, Jiang E, Zhou X, Shao Z, et al. Autophagy regulates the cancer stem cell phenotype of head and neck squamous cell carcinoma through the noncanonical FOXO3/SOX2 axis. Oncogene. 2022;41:634–46.
Sun S, Liu S, Duan SZ, Zhang L, Zhou H, Hu Y, et al. Targeting the c-Met/FZD8 signaling axis eliminates patient-derived cancer stem-like cells in head and neck squamous carcinomas. Cancer Res. 2014;74:7546–59.
Fang L, Zhu Q, Neuenschwander M, Specker E, Wulf-Goldenberg A, Weis W, et al. A small-molecule antagonist of the β-catenin/TCF4 interaction blocks the self-renewal of cancer stem cells and suppresses tumorigenesis. Cancer Res. 2016;76:891–901.
Guo B, Yang F, Zhang L, Zhao Q, Wang W, Yin L, et al. Cuproptosis induced by ROS responsive nanoparticles with elesclomol and copper combined with αPD-L1 for enhanced cancer immunotherapy. Adv Mater. 2023;35:e2212267.
Banci L, Bertini I, Ciofi-Baffoni S, Kozyreva T, Zovo K, Palumaa P. Affinity gradients drive copper to cellular destinations. Nature. 2010;465:645–48.
Li L, Jensen R. Understanding and overcoming immunosuppression shaped by cancer stem cells. Cancer Res. 2023;83:2096–104.
Castellano M, Pollock P, Walters M, Sparrow L, Down L, Gabrielli B, et al. CDKN2A/p16 is inactivated in most melanoma cell lines. Cancer Res. 1997;57:4868–75.
Dok R, Kalev P, Van Limbergen E, Asbagh L, Vázquez I, Hauben E, et al. p16INK4a impairs homologous recombination-mediated DNA repair in human papillomavirus-positive head and neck tumors. Cancer Res. 2014;74:1739–51.
Spranger S, Bao R, Gajewski T. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231–5.
Liu Y, Sun Z. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics. 2021;11:5365–86.
Takeuchi Y, Tanegashima T, Sato E, Irie T, Sai A, Itahashi K, et al. Highly immunogenic cancer cells require activation of the WNT pathway for immunological escape. Sci Immunol. 2021;6:eabc6424.
Solier S, Müller S, Cañeque T, Versini A, Mansart A, Sindikubwabo F, et al. A druggable copper-signalling pathway that drives inflammation. Nature. 2023;617:386–94.
Pezacki AT, Matier CD, Gu X, Kummelstedt E, Bond SE, Torrente L, et al. Oxidation state-specific fluorescent copper sensors reveal oncogene-driven redox changes that regulate labile copper(II) pools. Proc Natl Acad Sci USA. 2022;119:e2202736119.
Tang D, Kroemer G, Kang R. Targeting cuproplasia and cuproptosis in cancer. Nat Rev Clin Oncol. 2024;21:370–88.
Nagai M, Vo NH, Shin Ogawa L, Chimmanamada D, Inoue T, Chu J, et al. The oncology drug elesclomol selectively transports copper to the mitochondria to induce oxidative stress in cancer cells. Free Radic Biol Med. 2012;52:2142–50.
Liu Y, Li S, Wang S, Yang Q, Wu Z, Zhang M, et al. LIMP-2 enhances cancer stem-like cell properties by promoting autophagy-induced GSK3β degradation in head and neck squamous cell carcinoma. Int J Oral Sci. 2023;15:24.
Chen L, Yang Q, Li Y, Yang L, Liu J, Li H, et al. Targeting CMTM6 suppresses stem cell-like properties and enhances antitumor immunity in head and neck squamous cell carcinoma. Cancer Immunol Res. 2020;8:179–91.
Arnesano F, Natile G. Interference between copper transport systems and platinum drugs. Semin Cancer Biol. 2021;76:173–88.
Członkowska A, Litwin T, Dusek P, Ferenci P, Lutsenko S, Medici V, et al. Wilson disease. Nat Rev Dis Primers. 2018;4:21.
Wang S, Wu Z, Zhu S, Wan S, Zhang M, Zhang B, et al. CTLA-4 blockade induces tumor pyroptosis via CD8 T cells in head and neck squamous cell carcinoma. Mol Ther. 2023;31:2154–68.
Kaya-Okur H, Wu S, Codomo C, Pledger E, Bryson T, Henikoff J, et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat Commun. 2019;10:1930.
Barbie D, Tamayo P, Boehm J, Kim S, Moody S, Dunn I, et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature. 2009;462:108–12.
Funding
This work was financially supported by National Natural Science Foundation of China 82273202, 82072996, 82002893, 82103670, National Key Research and Development Program 2022YFC2504200, Interdisciplinary innovative foundation of Wuhan University XNJC202303, and the Fundamental Research Funds for the Central Universities (2042022dx0003).
Author information
Authors and Affiliations
Contributions
Y.T. Liu: Data curation, conceptualization, methodology, formal analysis, investigation, writing-original draft. L. Chen: Data curation, methodology, formal analysis, writing-original draft. S.J. Li: Data curation, methodology. W.Y. Wang: Investigation, methodology. Y.Y. Wang: Data curation. Q.C. Yang: Data curation. A. Song: Methodology. M.J. Zhang: Formal analysis. W.T. Mo: Formal analysis. H. Li: Supervision. C.Y. Hu: Writing-review and editing, funding acquisition. Z.J. Sun: Conceptualization, supervision, funding acquisition, writing-original draft, project administration, resources, writing-review and editing.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethical approval and consent to participate
Approval for human tissue samples utilization was granted by the Medical Ethics Committee of the School and Hospital of Stomatology, Wuhan University (2016LUNSHENZI62), following the guidelines of Declaration of Helsinki. All patients provided written informed consent. All animal experiments were carried out with the approval of the center for Animal Experiment of Wuhan University (WP20230467) and performed according to institutional guidelines.
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.
About this article
Cite this article
Liu, YT., Chen, L., Li, SJ. et al. Dysregulated Wnt/β-catenin signaling confers resistance to cuproptosis in cancer cells. Cell Death Differ (2024). https://doi.org/10.1038/s41418-024-01341-2
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41418-024-01341-2