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:

Palmitoylation of GPX4 via the targetable ZDHHC8 determines ferroptosis sensitivity and antitumor immunity

Abstract

Ferroptosis is closely linked with various pathophysiological processes, including aging, neurodegeneration, ischemia-reperfusion injury, viral infection and, notably, cancer progression; however, its post-translational regulatory mechanisms remain incompletely understood. Here we revealed a crucial role of S-palmitoylation in regulating ferroptosis through glutathione peroxidase 4 (GPX4), a pivotal enzyme that mitigates lipid peroxidation. We identified that zinc finger DHHC-domain containing protein 8 (zDHHC8), an S-acyltransferase that is highly expressed in multiple tumors, palmitoylates GPX4 at Cys75. Through small-molecule drug screening, we identified PF-670462, a zDHHC8-specific inhibitor that promotes the degradation of zDHHC8, consequently attenuating GPX4 palmitoylation and enhancing ferroptosis sensitivity. PF-670462 inhibition of zDHHC8 facilitates the CD8+ cytotoxic T cell-induced ferroptosis of tumor cells, thereby improving the efficacy of cancer immunotherapy in a B16-F10 xenograft model. Our findings reveal the prominent role of the zDHHC8–GPX4 axis in regulating ferroptosis and highlight the potential application of zDHHC8 inhibitors in anticancer therapy.

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: Small-molecule screening revealed that palmitoylation inhibitor 2-BP can increase the ferroptosis sensitivity of tumor cells.
Fig. 2: Palmitoylation of GPX4 is essential for ferroptosis resistance.
Fig. 3: zDHHC8 mediates GPX4 palmitoylation.
Fig. 4: ZDHHC8 deficiency increases the sensitivity of tumor cells to ferroptosis.
Fig. 5: zDHHC8-mediated GPX4 palmitoylation is negatively correlated with immune cell infiltration.
Fig. 6: PF-670462 increases ferroptosis sensitivity of tumor cells by targeting zDHHC8.
Fig. 7: PF-670462 inhibits immune escape of tumor through ferroptosis in vivo.

Similar content being viewed by others

Data availability

All data supporting the findings of this study are included in the paper and its supplementary files. The crystal structure of GPX4 used in this study is available from the Protein Data Bank (6hn3). The predicted structure of zDHHC8 was obtained from the AlphaFold Protein Structure Database (AF-Q9ULC8-F1). The predicted interaction between GPX4 and zDHHC8 was performed via HADDOCK2.4 (v.2.5; https://wenmr.science.uu.nl/haddock2.4/). The crystal structure of the GPX4 and zDHHC8 complex was extracted by PyMoL software v.2.5.2 (San Carlos). The human cancer data were derived from the TCGA Research Network (http://cancergenome.nih.gov/). Normal sample data were derived from the GTEx database (https://gtexportal.org/home/). Data of the immune infiltration analysis in SKCM, COAD and READ were obtained from TCGA Research Network and analyzed via the Tumor Immune Estimation Resource (https://cistrome.shinyapps.io/timer/). The whole-genome sequencing data of ZDHHC8-knockout HT-1080 cells generated in this study have been deposited in the Sequence Read Archive database under BioProject accession no. PRJNA1210772. Source data are provided with this paper.

Code availability

No unique code was developed for this study.

References

  1. Morad, G., Helmink, B. A., Sharma, P. & Wargo, J. A. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell 184, 5309–5337 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Topalian, S. L. et al. Neoadjuvant immune checkpoint blockade: a window of opportunity to advance cancer immunotherapy. Cancer Cell 41, 1551–1566 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Dagher, O. K. & Posey, A. D. Jr. Forks in the road for CAR T and CAR NK cell cancer therapies. Nat. Immunol. 24, 1994–2007 (2023).

    Article  CAS  PubMed  Google Scholar 

  4. Tay, C., Tanaka, A. & Sakaguchi, S. Tumor-infiltrating regulatory T cells as targets of cancer immunotherapy. Cancer Cell 41, 450–465 (2023).

    Article  CAS  PubMed  Google Scholar 

  5. Albelda, S. M. CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat. Rev. Clin. Oncol. 21, 47–66 (2024).

    Article  PubMed  Google Scholar 

  6. Singh, N. & Maus, M. V. Synthetic manipulation of the cancer-immunity cycle: CAR-T cell therapy. Immunity 56, 2296–2310 (2023).

    Article  CAS  PubMed  Google Scholar 

  7. Nong, C., Guan, P., Li, L., Zhang, H. & Hu, H. Tumor immunotherapy: mechanisms and clinical applications. Med. Comm. Oncol. https://doi.org/10.1002/mog2.8 (2022).

  8. Zou, W., Wolchok, J. D. & Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 8, 328rv324 (2016).

    Article  Google Scholar 

  9. Khalil, D. N., Smith, E. L., Brentjens, R. J. & Wolchok, J. D. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat. Rev. Clin. Oncol. 13, 273–290 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Reina-Campos, M. et al. Metabolic programs of T cell tissue residency empower tumour immunity. Nature 621, 179–187 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Barry, M. & Bleackley, R. C. Cytotoxic T lymphocytes: all roads lead to death. Nat. Rev. Immunol. 2, 401–409 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Golstein, P. & Griffiths, G. M. An early history of T cell-mediated cytotoxicity. Nat. Rev. Immunol. 18, 527–535 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Wang, W. et al. CD8(+) T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Liao, P. et al. CD8(+) T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell 40, 365–378.e366 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Yang, W. S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bell, H. N., Stockwell, B. R. & Zou, W. Ironing out the role of ferroptosis in immunity. Immunity 57, 941–956 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mao, C. et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 593, 586–590 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bersuker, K. et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575, 688–692 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Doll, S. et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Lei, G., Zhuang, L. & Gan, B. Targeting ferroptosis as a vulnerability in cancer. Nat. Rev. Cancer 22, 381–396 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Stockwell, B. R. Ferroptosis turns 10: emerging mechanisms, physiological functions, and therapeutic applications. Cell 185, 2401–2421 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Xia, Y. et al. The mevalonate pathway is a druggable target for vaccine adjuvant discovery. Cell 175, 1059–1073.e1021 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Kim, S. et al. Blocking myristoylation of Src Inhibits its kinase activity and suppresses prostate cancer progression. Cancer Res. 77, 6950–6962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lao, Y. et al. Glutaryl-CoA dehydrogenase suppresses tumor progression and shapes an anti-tumor microenvironment in hepatocellular carcinoma. J. Hepatol. https://doi.org/10.1016/j.jhep.2024.05.034 (2024).

  26. Chen, B., Sun, Y., Niu, J., Jarugumilli, G. K. & Wu, X. Protein lipidation in cell signaling and diseases: function, regulation, and therapeutic opportunities. Cell Chem. Biol. 25, 817–831 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen, S. et al. Palmitoylation-dependent activation of MC1R prevents melanomagenesis. Nature 549, 399–403 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ko, P. J. & Dixon, S. J. Protein palmitoylation and cancer. EMBO Rep. 19, e46666. (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Qu, M., Zhou, X., Wang, X. & Li, H. Lipid-induced S-palmitoylation as a vital regulator of cell signaling and disease development. Int. J. Biol. Sci. 17, 4223–4237 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Jin, J., Zhi, X., Wang, X. & Meng, D. Protein palmitoylation and its pathophysiological relevance. J. Cell. Physiol. 236, 3220–3233 (2021).

    Article  CAS  PubMed  Google Scholar 

  31. Jiang, Y. et al. STAT3 palmitoylation initiates a positive feedback loop that promotes the malignancy of hepatocellular carcinoma cells in mice. Sci. Signal. 16, eadd2282 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Mukai, K. et al. Activation of STING requires palmitoylation at the Golgi. Nat. Commun. 7, 11932 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Haag, S. M. et al. Targeting STING with covalent small-molecule inhibitors. Nature 559, 269–273 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Wang, L. et al. Palmitoylation prevents sustained inflammation by limiting NLRP3 inflammasome activation through chaperone-mediated autophagy. Mol. Cell 83, 281–297.e210 (2023).

    Article  CAS  PubMed  Google Scholar 

  35. Yu, T. et al. NLRP3 Cys126 palmitoylation by ZDHHC7 promotes inflammasome activation. Cell Rep. 43, 114070 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kim, Y. C. et al. Toll-like receptor mediated inflammation requires FASN-dependent MYD88 palmitoylation. Nat. Chem. Biol. 15, 907–916 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Lu, Y. et al. Palmitoylation of NOD1 and NOD2 is required for bacterial sensing. Science 366, 460–467 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Zhou, L. et al. Palmitoylation restricts SQSTM1/p62-mediated autophagic degradation of NOD2 to modulate inflammation. Cell Death Differ. 29, 1541–1551 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhang, G. et al. CPT1A induction following epigenetic perturbation promotes MAVS palmitoylation and activation to potentiate antitumor immunity. Mol. Cell 83, 4370–4385.e4379 (2023).

    Article  CAS  PubMed  Google Scholar 

  40. Bu, L. et al. Targeting APT2 improves MAVS palmitoylation and antiviral innate immunity. Mol. Cell 84, 3513–3529.e3515 (2024).

    Article  CAS  PubMed  Google Scholar 

  41. Wang, L. et al. Palmitoylation acts as a checkpoint for MAVS aggregation to promote antiviral innate immune responses. J. Clin. Invest. 134, e177924 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yao, H. et al. A peptidic inhibitor for PD-1 palmitoylation targets its expression and functions. RSC Chem. Biol. 2, 192–205 (2021).

    Article  CAS  PubMed  Google Scholar 

  43. Yao, H. et al. Inhibiting PD-L1 palmitoylation enhances T-cell immune responses against tumours. Nat. Biomed. Eng. 3, 306–317 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Yang, Y. et al. Palmitoylation stabilizes PD-L1 to promote breast tumor growth. Cell Res. 29, 83–86 (2019).

    Article  PubMed  Google Scholar 

  45. Zhang, N. et al. A palmitoylation-depalmitoylation relay spatiotemporally controls GSDMD activation in pyroptosis. Nat. Cell Biol. 26, 757–769 (2024).

    Article  PubMed  Google Scholar 

  46. Du, G. et al. ROS-dependent S-palmitoylation activates cleaved and intact gasdermin D. Nature https://doi.org/10.1038/s41586-024-07373-5 (2024).

  47. Balasubramanian, A. et al. The palmitoylation of gasdermin D directs its membrane translocation and pore formation during pyroptosis. Sci. Immunol. 9, eadn1452 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bu, L. et al. High-fat diet promotes liver tumorigenesis via palmitoylation and activation of AKT. Gut 73, 1156–1168 (2024).

    Article  CAS  PubMed  Google Scholar 

  49. Zhang, Z. et al. Palmitoylation of TIM-3 promotes immune exhaustion and restrains antitumor immunity. Sci. Immunol.9, eadp7302 (2024).

    Article  CAS  PubMed  Google Scholar 

  50. Wang, Z. et al. AMPKα1-mediated ZDHHC8 phosphorylation promotes the palmitoylation of SLC7A11 to facilitate ferroptosis resistance in glioblastoma. Cancer Lett. 584, 216619 (2024).

    Article  CAS  PubMed  Google Scholar 

  51. Meng, Q. J. et al. Entrainment of disrupted circadian behavior through inhibition of casein kinase 1 (CK1) enzymes. PNAS 107, 15240–15245 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Janovska, P. et al. Casein kinase 1 is a therapeutic target in chronic lymphocytic leukemia. Blood 131, 1206–1218 (2018).

    Article  CAS  PubMed  Google Scholar 

  53. Rana, M. S. et al. Fatty acyl recognition and transfer by an integral membrane S-acyltransferase. Science 359, eaao6326 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Lee, C. J. et al. Bivalent recognition of fatty acyl-CoA by a human integral membrane palmitoyltransferase. PNAS 119, eaao6326 (2022).

    Article  Google Scholar 

  55. Yang, A. et al. Regulation of RAS palmitoyltransferases by accessory proteins and palmitoylation. Nat. Struct. Mol. Biol. 31, 436–446 (2024).

    Article  CAS  PubMed  Google Scholar 

  56. Meng, X. et al. FBXO38 mediates PD-1 ubiquitination and regulates anti-tumour immunity of T cells. Nature 564, 130–135 (2018).

    Article  CAS  PubMed  Google Scholar 

  57. Friedmann Angeli, J. P., Krysko, D. V. & Conrad, M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat. Rev. Cancer 19, 405–414 (2019).

    Article  CAS  PubMed  Google Scholar 

  58. Broadfield, L. A., Pane, A. A., Talebi, A., Swinnen, J. V. & Fendt, S. M. Lipid metabolism in cancer: new perspectives and emerging mechanisms. Dev. Cell 56, 1363–1393 (2021).

    Article  CAS  PubMed  Google Scholar 

  59. Kim, R. et al. Ferroptosis of tumour neutrophils causes immune suppression in cancer. Nature 612, 338–346 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ma, X. et al. CD36-mediated ferroptosis dampens intratumoral CD8(+) T cell effector function and impairs their antitumor ability. Cell Metab. 33, 1001–1012.e1005 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zheng, Y. et al. Modulation of virus-induced neuroinflammation by the autophagy receptor SHISA9 in mice. Nat. Microbiol. 8, 958–972 (2023).

    Article  CAS  PubMed  Google Scholar 

  62. Liu, D. et al. Targeting the TRIM14/USP14 axis enhances immunotherapy efficacy by inducing autophagic degradation of PD-L1. Cancer Res. 84, 2806–2819 (2024).

    Article  CAS  PubMed  Google Scholar 

  63. Brigidi, G. S. & Bamji, S. X. Detection of protein palmitoylation in cultured hippocampal neurons by immunoprecipitation and acyl-biotin exchange (ABE). J. Vis. Exp. https://doi.org/10.3791/50031 (2013).

  64. Zhou, B. et al. Low-background acyl-biotinyl exchange largely eliminates the coisolation of non-S-acylated proteins and enables deep S-acylproteomic analysis. Anal. Chem. 91, 9858–9866 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yan, W., Hwang, D. & Aebersold, R. Quantitative proteomic analysis to profile dynamic changes in the spatial distribution of cellular proteins. Methods Mol. Biol. 432, 389–401 (2008).

    Article  CAS  PubMed  Google Scholar 

  66. Nishimura, T. & Mizushima, N. The ULK complex initiates autophagosome formation at phosphatidylinositol synthase-enriched ER subdomains. Autophagy 13, 1795–1796 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Li, J. et al. Tumor-specific GPX4 degradation enhances ferroptosis-initiated antitumor immune response in mouse models of pancreatic cancer. Sci. Transl. Med. 15, eadg3049 (2023).

    Article  CAS  PubMed  Google Scholar 

  68. Roveri, A., Maiorino, M. & Ursini, F. Enzymatic and immunological measurements of soluble and membrane-bound phospholipid-hydroperoxide glutathione peroxidase. Methods Enzymol. 233, 202–212 (1994).

    Article  CAS  PubMed  Google Scholar 

  69. Stolwijk, J. M., Falls-Hubert, K. C., Searby, C. C., Wagner, B. A. & Buettner, G. R. Simultaneous detection of the enzyme activities of GPx1 and GPx4 guide optimization of selenium in cell biological experiments. Redox Biol. 32, 101518 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Frazão, C. et al. Crystallization and preliminary diffraction data analysis of both single and pseudo-merohedrally twinned crystals of rubredoxin oxygen oxidoreductase from Desulfovibrio gigas. Acta Crystallogr. D Biol. Crystallogr. 55, 1465–1467 (1999).

    Article  PubMed  Google Scholar 

  71. Honorato, R. V. et al. Structural biology in the clouds: the WeNMR-EOSC ecosystem. Front. Mol. Biosci. 8, 729513 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Chu, M. et al. Elucidation of potential targets of san-miao-san in the treatment of osteoarthritis based on network pharmacology and molecular docking analysis. eCAM 2022, 7663212 (2022).

    PubMed  PubMed Central  Google Scholar 

  73. Wang, X. et al. TOX promotes the exhaustion of antitumor CD8(+) T cells by preventing PD1 degradation in hepatocellular carcinoma. J. Hepatol. 71, 731–741 (2019).

    Article  CAS  PubMed  Google Scholar 

  74. Peng, L. et al. Hippo-signaling-controlled MHC class I antigen processing and presentation pathway potentiates antitumor immunity. Cell Rep. 43, 114003 (2024).

    Article  CAS  PubMed  Google Scholar 

  75. Efimova, I. et al. Vaccination with early ferroptotic cancer cells induces efficient antitumor immunity. J. Immunother. Cancer 8, e001369 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Wiernicki, B. et al. Cancer cells dying from ferroptosis impede dendritic cell-mediated anti-tumor immunity. Nat. Commun. 13, 3676 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhang, H. L. et al. PKCβII phosphorylates ACSL4 to amplify lipid peroxidation to induce ferroptosis. Nat. Cell Biol. 24, 88–98 (2022).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Y. Ao in Shenzhen University for providing the MEF cell line. This work was supported by the National Natural Science Foundation of China (82341047 to J.C. and 32170913 and 32370982 to B.H.), the Guangdong Basic and Applied Basic Research Foundation (2024B1515040009, 2023B1212060028 and 2023B1515120085 to J.C. and 2023B1515120083 to B.H.), the Guizhou Provincial Science and Technology Projects (Qian Kehe Basic-ZK (2024) General 594 to X.Z.), the Shenzhen Medical Research Fund (B2302008 to B.H.), Changzhou High-Level Health Talents Training Project (2022260 to F.Z.), Changzhou Science and Technology Project (CJ20230062 to F.Z.) and the Fundamental Research Funds for the Central Universities (23yxqntd001 to J.C.). The funders had no role in the study design, data collection and analysis, decision to publish or preparation of the paper.

Author information

Authors and Affiliations

Contributions

J.C. conceived the project and designed the experiments. L.Z. and G.L. performed the experiments. T.Z., Z.C., S.Y., W.L., L.C., Y.Y., M.H., J.L., Q.D., D.L., B.H., F.Z. and X.Z. provided technical help. L.Z. and G.L. analyzed the data. J.C. provided resources, and directed the research. L.Z., G.L. and J.C. wrote the paper. All authors read and approved the final paper.

Corresponding author

Correspondence to Jun Cui.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Inhibition of palmitoylation increases the ferroptosis sensitivity of various kind of tumor cells.

a, The cell viability of HT-1080 cells treated with RSL3 (0.05 μM) in conjunction with or without 2-BP (10 μM) and DFO (5 μM), Liproxstatin-1 (5 μM), Z-VAD-FMK (10 μM) or NSA (2.5 μM) for 24 h. b, Lactate dehydrogenase (LDH) release measurement of HT-1080 cells treated with RSL3 (0.05 μM) in conjunction with or without 2-BP (10 μM) and Ferrostatin-1 (Fer-1, 2 μM), DFO (5 μM), Liproxstatin-1 (5 μM), Z-VAD-FMK (10 μM) or NSA (2.5 μM) for 16 h. c, Images of HT-1080 cell colonies by crystal violet staining after treatment with the indicated concentrations of ML162 in conjunction with DMSO or 2-BP (10 μM) for 24 h. d, Propidium iodide (PI) incorporation measurement of HT-1080 cells treated with ML162 (0.05 μM) in combination with DMSO or 2-BP (10 μM) for 12 h. e, Lipid peroxidation assessment of HT-1080 cells treated with ML162 (0.05 μM) in conjunction with DMSO or 2-BP (10 μM) for 10 h. f, The cell viability of HT-1080 cells treated with the indicated concentrations of ML162 in conjunction with or without 2-BP (10 μM) and Ferrostatin-1 (Fer-1, 2 μM) for 24 h. g, Images of HT-1080 cell colonies by crystal violet staining after treatment with the indicated concentrations of IKE in conjunction with DMSO or 2-BP (10 μM) for 24 h. h, PI incorporation measurement of HT-1080 cells treated with IKE (1 μM) in combination with DMSO or 2-BP (10 μM) for 12 h. i, Lipid peroxidation assessment of HT-1080 cells treated with IKE (1 μM) in conjunction with DMSO or 2-BP (10 μM) for 10 h. j, The cell viability of HT-1080 cells treated with the indicated concentrations of IKE in conjunction with or without 2-BP (10 μM) and Fer-1 (2 μM) for 24 h. k, The cell viability of A375 cells treated with the indicated concentrations of RSL3 in conjunction with or without 2-BP (10 μM) and Fer-1 (2 μM) for 24 h. l-n, The cell viability of A375 (l), B16-F10 (m) and MC38 (n) cells treated with the indicated concentrations of ML162 in conjunction with or without 2-BP (10 μM) and Fer-1 (2 μM) for 24 h. Data in c and g are representative of three independent experiments. For b,d,e,h,i, data are presented as mean ± s.d. of n = 3 biological replicates per group. For a,f,j-n, data are presented as mean ± s.d. n = 4 biological replicates per group. Statistical analysis was performed using one-way ANOVA with Dunnett’s test (a,b), two-way ANOVA with Šídák’s test (d,e,h,i) and two-way ANOVA with Tukey’s test (f,j-n).

Source data

Extended Data Fig. 2 Inhibition of GPX4 palmitoylation increased the ferroptosis sensitivity of tumor cells.

a, Immunoblotting analysis of GPX4 palmitoylation by ABE assay in HEK293T cells transfected with plasmids encoding GPX4-HA for 36 h, followed with treatment of 2-BP (10 μM) for 12 h. b, HEK293T cells were transfected with plasmids encoding GPX4-HA for 36 h, followed with treatment of palmitic acid-azide (50 μM) with or without 2-BP (10 μM) for 8 h, then prepared for Click-iT reaction. c, Immunoblotting analysis of GPX4 palmitoylation by ABE assay after the treatment of DMSO, RSL3 (1 μM) or IKE (5 μM) for 10 h in HEK293T cells transfected with plasmid encoding GPX4-HA for 36 h. d, Immunoblotting analysis of GPX4 and Flag-FSP1 in indicated cell lines. e, Immunoblotting analysis of GPX4 in GPX4-KO HT-1080 cells overexpressing HA-tagged wild-type GPX4 or mutant GPX4-C75S. f, Schematic illustration of GPX4-competing peptides. g, Immunoblotting analysis of GPX4 palmitoylation by ABE assay in HT-1080 after the treatment of CPP-P Scr (10 μM), CPP-P Ctrl (10 μM) or CPP-P Comp (10 μM) for 12 h. h, Lipid peroxidation assessment of HT-1080 cells treated with IKE (1 μM) in conjunction with CPP-P Scr (10 μM), CPP-P Ctrl (10 μM) or CPP-P Comp (10 μM) for 10 h. i, The cell viability of HT-1080 cells treated with IKE (1 μM) in conjunction with CPP-P Scr (10 μM), CPP-P Ctrl (10 μM) or CPP-P Comp (10 μM), and Fer-1 (2 μM) for 24 h. j, Lipid peroxidation assessment of HT-1080 cells treated with IFN-γ (20 ng ml−1) and AA (20 μM) in conjunction with CPP-P Scr (10 μM), CPP-P Ctrl (10 μM) or CPP-P Comp (10 μM) for 36 h. k, The cell viability of HT-1080 cells treated with IFN-γ (20 ng ml-1) plus AA (20 μM) in conjunction with CPP-P Scr (10 μM), CPP-P Ctrl (10 μM) or CPP-P Comp (10 μM), and Fer-1 (2 μM) for 48 h. Data in d and e are representative of three independent experiments. For a–c,g, quantification of the indicated protein levels are determined by Image Lab software; data are presented as mean ± s.d. n = 3 independent experiments per group. For h,j, data are presented as mean ± s.d. of n = 3 biological replicates pe group. For i,k, data are presented as mean ± s.d. of n = 4 biological replicates per group. Statistical analysis was performed using two-tailed paired Student’s t-test (a,b), one-way ANOVA with Dunnett’s test (c,g) and two-way ANOVA with Tukey’s test (h-k); ns, not significant.

Source data

Extended Data Fig. 3 zDHHC8 is required for GPX4 palmitoylation.

a,b, Immunoblotting analysis of GPX4 palmitoylation by ABE assay in HEK293T cells transfected with plasmids encoding indicated zDHHCs-Flag for 36 h. c, Immunoblotting analysis of zDHHC8 in mock or ZDHHC8-KO HT-1080 cells. d, Immunoblotting analysis of zDHHC8 in Zdhhc8-knockdown B16-F10 cells. e, The cell viability of mock or ZDHHC8-KO HT-1080 cells treated with RSL3 (0.05 μM) together with or without DFO (5 μM), Liproxstatin-1 (5 μM), Z-VAD-FMK (10 μM) or NSA (2.5 μM) for 24 h. f, LDH release measurement of mock or ZDHHC8-KO HT-1080 cells treated with RSL3 (0.05 μM) together with or without ferrostatin-1 (Fer-1, 2 μM), DFO (5 μM), Liproxstatin-1 (5 μM), Z-VAD-FMK (10 μM) or NSA (2.5 μM) for 16 h. g, Immunoblotting analysis of zDHHC8 in ZDHHC8-knockdown HT-1080 cells, MC38 cells and CRC derived organoid. h, The cell viability of shCtrl or shZDHHC8 HT-1080 cells treated with RSL3 (0.05 μM), ML162 (0.05 μM) or IKE (1 μM), together with or without Fer-1 (2 μM) for 24 h. i,j, The cell viability of Zdhhc8-knockdown (shZdhhc8) B16-F10 cells (i) and MC38 cells (j) treated with RSL3 (0.05 μM), ML162 (0.05 μM) or IKE (1 μM), together with or without Fer-1 (2 μM) for 24 h. k, The cell viability of mock or ZDHHC8-KO HT-1080 cells treated with the indicated concentrations of RSL3 (left) or ML162 (right) in conjunction with DMSO or 2-BP (10 μM) for 24 h. l, Relative GSH level of HT-1080 cells after treated with vehicle, IKE (1 μM) or indicated concentration of 2-BP for 12 h. Data in a-d,g are representative of three independent experiments. For e,h-k data are presented as mean ± s.d. of n = 4 biological replicates per group. For f,l, data are presented as mean ± s.d. of n = 3 biological replicates per group. Statistical analysis was performed using one-way ANOVA with Dunnett’s test (l) and two-way ANOVA with Tukey’s test (e,f,h-k); ns, not significant.

Source data

Extended Data Fig. 4 ZDHHC8 deficiency inhibits colorectal cancer growth in vivo.

a, Analysis of mRNA expression of ZDHHC8 in colon adenocarcinoma (COAD) and rectum adenocarcinoma (READ) using the Cancer Genome Atlas (TCGA) datasets. For COAD, n = 41 samples in normal tissues, and n = 458 samples in tumor tissues. For READ, n = 10 samples in normal tissues, and n = 166 samples in tumor tissues. b, ZDHHC8 mRNA expression level is negatively co-related with the infiltration of CD8+ T cells in COAD and READ based on TCGA database. c,d, mRNA and protein level of ZDHHC8 were detected by real-time PCR (c) or immunoblot analysis (d) in CRC patients. e, Immunoblotting analysis of GPX4 palmitoylation by ABE assay in CRC samples from patients. Adj, adjacent normal tissues; Ca, cancerous tissue. f, The cell viability of control (shCtrl) or Zdhhc8-knockdown (shZdhhc8) MC38 cells. g-i, Tumor images (g), tumor growth curve (h), and tumor weight (i) of shCtrl and shZdhhc8 MC38 subcutaneous tumors. j, Lipid peroxidation assessment of shCtrl and shZdhhc8 MC38 subcutaneous tumors. k, Flow cytometric analysis of CD8+ cells in shCtrl and shZdhhc8 MC38 subcutaneous tumors. l, Representative CD8α immunofluorescence staining images of MC38 tumor sections from shCtrl and shZdhhc8 groups (n = 6 mice per group, 2 sections per mouse). Scale bar, 50 μm. m-o, Flow cytometric analysis of IFNγ+ (m), TNF+ (n) and GZMB+ (o) CD8+ cytotoxic T cells in shCtrl and shZdhhc8 MC38 subcutaneous tumors. For a, the box represents the interquartile range (IQR). The central line indicates the median. The lower and upper bounds of the box represent the first quartile and the third quartile. The whiskers extend to the minima and maxima. Data points outside the range of the whiskers are considered outliers. For b, The Tumor Immune Estimation Resource (TIMER, https://cistrome.shinyapps.io/timer/) accomplished the immune infiltration analysis, and the tumor purity adjusted the results. Spearman’s rank correlation test is used, following a two-sided test without adjustment for multiple comparisons. The partial correlation coefficient (part.cor) and the corresponding p value are shown on the plot. n = 458 tumor samples for COAD, n = 166 tumor samples for READ. For c, data are presented as mean ± s.d. of n = 30 samples in normal group and n = 42 samples in tumor group. For d, quantification of the indicated protein level is determined by Image Lab software; data are presented as mean ± s.d. n = 4 independent samples per group. For e, quantification of the indicated protein level is determined by Image Lab software; data are presented as mean ± s.d. n = 3 independent experiments per group. For f, data are presented as mean ± s.d. of n = 4 independent samples per group. For h-k,m-o, data are presented as mean ± s.d. of n = 6 mice in each group. Statistical analysis was performed using two-tailed paired Student’s t-test (e), two-tailed unpaired Student’s t-test (a,c,d), one-way ANOVA with Dunnett’s test (i-k,m-o) and two-way ANOVA with Tukey’s test (h).

Source data

Extended Data Fig. 5 PF-670462 increases the ferroptosis sensitivity of tumor cells via zDHHC8-GPX4 axis.

a, The cell viability of HT-1080 cells treated with RSL3 (0.05 μM) in conjunction with or without PF-670462 (10 μM) and DFO (5 μM), Lipro-1 (5 μM), Z-VAD-FMK (10 μM) or NSA (2.5 μM) for 24 h. b, LDH release measurement of HT-1080 cells treated with RSL3 (0.05 μM) in conjunction with or without PF-670462 (10 μM) and Fer-1 (2 μM), DFO (5 μM), Lipro-1 (5 μM), Z-VAD-FMK (10 μM) or NSA (2.5 μM) for 16 h. c, The cell viability of HT-1080 cells treated with the indicated concentrations of ML162 or IKE in conjunction with or without PF-670642 (10 μM) and Fer-1 (2 μM) for 24 h. d, The cell viability of various tumor cells treated with the indicated concentrations of RSL3 with PF-670462 (10 μM) for 24 h. e,f, The cell viability of HT-1080 cells treated with the indicated concentrations of RSL3 with SR-3029 (e) or LH846 (f) for 24 h. g, Immunoblotting analysis of CK1ε and CK1δ in control (shCtrl) or CK1δ/ε-knockdown (shCK1δ/ε) HT-1080 cells. h, The cell viability of shCtrl and shCK1δ/ε HT-1080 cells after treatment with the indicated concentration of RSL3. i, Immunoblotting analysis of thermal stability of zDHHC8-Flag protein by CETSA after the treatment of DMSO, LH846 (10 μM) or PF-670462 (10 μM) in HEK293T cells transfected with plasmid encoding zDHHC8-Flag. j, Immunoblotting analysis of thermal stability of zDHHC8 (NTD)-Flag protein by CETSA after the treatment of DMSO or PF-670462 (10 μM) in HEK293T cells transfected with plasmid encoding zDHHC8 (NTD)-Flag. k, Immunoblotting analysis of thermal stability of zDHHC-Flag protein by CETSA after the treatment of DMSO or PF-670462 (10 μM) in HEK293T cells transfected with plasmid encoding zDHHC5-Flag, zDHHC9-Flag or zDHHC20-Flag. l, Real-time PCR analysis of ZDHHC8 mRNA level in HT-1080 cells treated with the indicated concentration of PF-670462 for 24 h. For a,c-f and h, data are presented as mean ± s.d. of n = 4 biological replicates per group. For b,l, data are presented as mean ± s.d. of n = 3 biological replicates per group. Data in g,i-k are representative of three independent experiments. For i, quantification of the indicated protein level is determined by Image Lab software; data are presented as mean ± s.d. n = 3 independent experiments per group. Statistical analysis was performed using one-way ANOVA with Dunnett’s test (a,b,l), two-way ANOVA with Šídák’s test (d) and two-way ANOVA with Tukey’s test (c,i); ns, not significant.

Source data

Extended Data Fig. 6 PF-670462 selectively promotes zDHHC8 degradation.

Immunoblotting analysis of zDHHCs in HEK293T cells. HEK293T cells were transfected with plasmids encoding the indicated Flag-tagged zDHHCs for 24 h, then treated with the indicated concentrations of PF-670462 for 16 h. Cell lysates were harvested for immunoblotting analysis. Data are representative of three independent experiments.

Source data

Extended Data Fig. 7 PF-670462 promotes lysosomal degradation of zDHHC8.

a, Immunoblotting analysis of zDHHC8-Flag in HEK293T cells. HEK293T cells were transfected with plasmid encoding zDHHC8-Flag for 24 h, then treated with PF-670462 (10 μM) for 8 h. Cycloheximide (CHX, 100 mg/mL) was added in all groups at the indicated time points. Cell lysates were harvested for immunoblotting analysis. b, Immunoblotting analysis of zDHHC8-Flag in HEK293T cells. HEK293T cells were transfected with plasmid encoding zDHHC8-Flag for 24 h. Then the cells were treated with MG132 (10 μM) or bafilomycin A1 (Baf A1, 20 nM) for 12 h, after the pre-treatment of PF-670462 (10 μM) for 8 h. c, Immunoblotting analysis of GPX4 palmitoylation by ABE assay in HEK293T cells transfected with plasmid encoding GPX4-HA for 36 h, then treated with PF-670462 (10 μM) for 12 h. d, Relative GSH level of HT-1080 cells after treated with IKE (1 μM) in combination with vehicle or PF-670462 (10 μM) for 10 h. For ac, quantification of the indicated protein level is determined by Image Lab software; data are presented as mean ± s.d. n = 3 independent experiments per group. For d, data are presented as mean ± s.d. n = 3 independent experiments per group. Statistical analysis was performed using two-tailed paired Student’s t-test (c), one-way ANOVA with Dunnett’s test (b) and two-way ANOVA with Tukey’s test (a,d); ns, not significant.

Source data

Extended Data Fig. 8 PF-670462 has no side cytotoxicity in vivo.

a, Alanine aminotransferase (ALT), aspartate aminotransferase (AST), creatinine (CR) and creatine kinase isoenzymes (CK-MB) level measurement from mice serum after the indicated treatment of PF-670462. b, Representative hematoxylin and eosin (H&E) staining in the mice tissues (heart, liver, spleen, lung and kidney) after the indicated treatment of PF-670462. Scale bar, 200 μm. Data in b are representative of three independent mice. For a, data are shown as mean ± s.d. of n = 3 mice in each group. Statistical analysis was performed using one-way ANOVA with Dunnett’s test (a); ns, not significant.

Source data

Extended Data Fig. 9 PF-670462 enhances CD8+ T cell-mediated antitumor immunity.

a, Images of B16-F10 lung metastasis after treatment with vehicle or PF-670462 (10 mg/kg). b, Statistical of surface lung metastasis nodules. c, Hematoxylin-eosin (H&E) staining of B16-F10 metastasis lungs. Scale bar, 500 μm. d, The overall survival of B16-F10 lung metastasis model after treatment with vehicle and PF-670462 (10 mg/kg). n = 10 in each group. e, Tumor images of B16-F10 subcutaneous tumors in Rag2 -/- mice with indicated treatment. n = 5 in each group. f, Treatment schedule for replenishing of naïve CD8+ T cells and naïve B cells in Rag2 -/- mice. g, Tumor weight of B16-F10 subcutaneous tumors in Rag2 -/- mice with indicated treatment. n = 5 in each group. h. MEF-OVA cells were pretreated with PF-670462 (10 μM) for 6 h and then combinate with indicated concentrations of RSL3 for another 3 h, refresh the medium and the cell viability was detected after another 21 h culture. i, Treatment schedule of the vaccination model. j, The overall survival of B16-F10 subcutaneous tumor model with the indicated treatment. n = 12 mice in each group. For b, data are presented as mean ± s.d. of n = 6 mice in each group. For g, data are presented as mean ± s.d. of n = 5 mice in each group. For h, data are presented as mean ± s.d. of n = 3 biological replicates per group. Statistical analysis was performed using two-tailed unpaired Student’s t-test (b), two-way ANOVA with Šídák’s test (h), two-way ANOVA with Tukey’s test (g) and log-rank (Mantel–Cox) test (d,j); ns, not significant.

Source data

Supplementary information

Supplementary Information

Supplementary Fig.1 and its legend.

Reporting Summary

Supplementary Tables 1–5

Supplementary Table 1. Clinical information of patients with melanoma and colorectal cancer. Supplementary Table 2. Primers used in this project. Supplementary Table 3. General information and inhibition effects of bioactive small-molecule compounds. Supplementary Table 4. General information and inhibition effects of AI-screening candidates. Supplementary Table 5. List of antibodies used in this project.

Source data

Statistical source data

Statistical source data in all figures and extended data figures except for Extended Data Fig. 6.

Unprocessed western blots source data

Unprocessed western blots source data in Figs. 2, 3, 5 and 6 and Extended Data Figs. 2–7.

Fluorescent images source data

Fluorescent images source data in Figs. 1, 4, 5 and 7 and Extended Data Fig. 4.

High-resolution hematoxylin and eosin (H&E) images source data

High-resolution H&E staining images source data in Extended Data Figs. 8 and 9.

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

Zhou, L., Lian, G., Zhou, T. et al. Palmitoylation of GPX4 via the targetable ZDHHC8 determines ferroptosis sensitivity and antitumor immunity. Nat Cancer 6, 768–785 (2025). https://doi.org/10.1038/s43018-025-00937-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s43018-025-00937-y

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer