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:

Creatine kinase B suppresses ferroptosis by phosphorylating GPX4 through a moonlighting function

Abstract

Activation of receptor protein kinases is prevalent in various cancers with unknown impact on ferroptosis. Here we demonstrated that AKT activated by insulin-like growth factor 1 receptor signalling phosphorylates creatine kinase B (CKB) T133, reduces metabolic activity of CKB and increases CKB binding to glutathione peroxidase 4 (GPX4). Importantly, CKB acts as a protein kinase and phosphorylates GPX4 S104. This phosphorylation prevents HSC70 binding to GPX4, thereby abrogating the GPX4 degradation regulated by chaperone-mediated autophagy, alleviating ferroptosis and promoting tumour growth in mice. In addition, the levels of GPX4 are positively correlated with the phosphorylation levels of CKB T133 and GPX4 S104 in human hepatocellular carcinoma specimens and associated with poor prognosis of patients with hepatocellular carcinoma. These findings reveal a critical mechanism by which tumour cells counteract ferroptosis by non-metabolic function of CKB-enhanced GPX4 stability and underscore the potential to target the protein kinase activity of CKB for cancer treatment.

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: CKB T133 phosphorylation increases GPX4 stability.
Fig. 2: CKB-mediated GPX4 S104 phosphorylation stabilizes GPX4.
Fig. 3: GPX4 phosphorylation blocks CMA-mediated GPX4 degradation.
Fig. 4: GPX4 S104 phosphorylation suppresses ferroptosis.
Fig. 5: GPX4 S104 phosphorylation promotes tumour growth.
Fig. 6: GPX4 pS104 predicts clinical aggressiveness of HCC.

Similar content being viewed by others

Data availability

The human HCC data were derived from the TCGA Research Network. The dataset derived from this resource that supports the findings of this study is available at the following link: https://tcga.xenahubs.net/download/TCGA.LIHC.sampleMap/HiSeqV2.gz. Mass spectrometry data have been deposited in ProteomeXchange with the accession code PXD040322. UniProt protein database (EMBL-EBI) was used for protein identification. Source data are provided with this paper.

References

  1. Stockwell, B. R. et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 171, 273–285 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. 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 

  3. Jiang, X., Stockwell, B. R. & Conrad, M. Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 22, 266–282 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  4. 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 

  5. Ingold, I. et al. Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell 172, 409–422.e21 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Yant, L. J. et al. The selenoprotein GPX4 is essential for mouse development and protects from radiation and oxidative damage insults. Free Radic. Biol. Med 34, 496–502 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Stockwell, B. R. Dawn of a new era of targeted antioxidant therapies. Cell Chem. Biol. 26, 1483–1485 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hangauer, M. J. et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature 551, 247–250 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zou, Y. et al. A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nat. Commun. 10, 1617 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Li, X., Egervari, G., Wang, Y., Berger, S. L. & Lu, Z. Regulation of chromatin and gene expression by metabolic enzymes and metabolites. Nat. Rev. Mol. Cell Biol. 19, 563–578 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Li, X., Qian, X. & Lu, Z. Fructokinase A acts as a protein kinase to promote nucleotide synthesis. Cell Cycle 15, 2689–2690 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Li, X., Zheng, Y. & Lu, Z. PGK1 is a new member of the protein kinome. Cell Cycle 15, 1803–1804 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Xu, D. et al. The evolving landscape of noncanonical functions of metabolic enzymes in cancer and other pathologies. Cell Metab. 33, 33–50 (2021).

    Article  CAS  PubMed  Google Scholar 

  14. Xu, D. et al. The protein kinase activity of fructokinase A specifies the antioxidant responses of tumor cells by phosphorylating p62. Sci. Adv. 5, eaav4570 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lu, Z. & Hunter, T. Metabolic kinases moonlighting as protein kinases. Trends Biochem. Sci. 43, 301–310 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lu, S. & Wang, Y. Nonmetabolic functions of metabolic enzymes in cancer development. Cancer Commun. 38, 63 (2018).

    Article  Google Scholar 

  17. Jiang, H., Zhu, L., Xu, D. & Lu, Z. A newly discovered role of metabolic enzyme PCK1 as a protein kinase to promote cancer lipogenesis. Cancer Commun. 40, 389–394 (2020).

    Article  Google Scholar 

  18. Jiang, H. et al. Fructose and fructose kinase in cancer and other pathologies. J. Genet. Genomics 48, 531–539 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Kazak, L. & Cohen, P. Creatine metabolism: energy homeostasis, immunity and cancer biology. Nat. Rev. Endocrinol. 16, 421–436 (2020).

    Article  PubMed  Google Scholar 

  20. Loo, J. M. et al. Extracellular metabolic energetics can promote cancer progression. Cell 160, 393–406 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Nagpal, L., Kornberg, M. D., Albacarys, L. K. & Snyder, S. H. Inositol hexakisphosphate kinase-2 determines cellular energy dynamics by regulating creatine kinase-B. Proc. Natl Acad. Sci. USA 118, e2020695118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rahbani, J. F. et al. Creatine kinase B controls futile creatine cycling in thermogenic fat. Nature 590, 480–485 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Papalazarou, V. et al. The creatine-phosphagen system is mechanoresponsive in pancreatic adenocarcinoma and fuels invasion and metastasis. Nat. Metab. 2, 62–80 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Kazak, L. et al. A creatine-driven substrate cycle enhances energy expenditure and thermogenesis in beige fat. Cell 163, 643–655 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Breuhahn, K. & Schirmacher, P. Reactivation of the insulin-like growth factor-II signaling pathway in human hepatocellular carcinoma. World J. Gastroenterol. 14, 1690–1698 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xu, D. et al. The gluconeogenic enzyme PCK1 phosphorylates INSIG1/2 for lipogenesis. Nature 580, 530–535 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Zhang, Y. et al. mTORC1 couples cyst(e)ine availability with GPX4 protein synthesis and ferroptosis regulation. Nat. Commun. 12, 1589 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Liu, R. et al. Choline kinase alpha 2 acts as a protein kinase to promote lipolysis of lipid droplets. Mol. Cell 81, 2722–2735.e9 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Li, X. et al. Mitochondria-translocated PGK1 functions as a protein kinase to coordinate glycolysis and the TCA cycle in tumorigenesis. Mol. Cell 61, 705–719 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lin, L., Perryman, M. B., Friedman, D., Roberts, R. & Ma, T. S. Determination of the catalytic site of creatine kinase by site-directed mutagenesis. Biochim. Biophys. Acta 1206, 97–104 (1994).

    Article  CAS  PubMed  Google Scholar 

  31. Wu, Z. et al. Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc. Natl Acad. Sci. USA 116, 2996–3005 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Circu, M. L. & Aw, T. Y. Redox biology of the intestine. Free Radic. Res. 45, 1245–1266 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Droge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 82, 47–95 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Yang, Y. & Yee, D. IGF-I regulates redox status in breast cancer cells by activating the amino acid transport molecule xC. Cancer Res. 74, 2295–2305 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hodgson, N., Trivedi, M., Muratore, C., Li, S. & Deth, R. Soluble oligomers of amyloid-beta cause changes in redox state, DNA methylation, and gene transcription by inhibiting EAAT3 mediated cysteine uptake. J. Alzheimers Dis. 36, 197–209 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Takahashi, S., Hisatsune, A., Kurauchi, Y., Seki, T. & Katsuki, H. Insulin-like growth factor 1 specifically up-regulates expression of modifier subunit of glutamate-cysteine ligase and enhances glutathione synthesis in SH-SY5Y cells. Eur. J. Pharmacol. 771, 99–106 (2016).

    Article  CAS  PubMed  Google Scholar 

  37. Hayes, J. D. & Dinkova-Kostova, A. T. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem. Sci. 39, 199–218 (2014).

    Article  CAS  PubMed  Google Scholar 

  38. Ghoneum, A., Abdulfattah, A. Y., Warren, B. O., Shu, J. & Said, N. Redox homeostasis and metabolism in cancer: a complex mechanism and potential targeted therapeutics. Int. J. Mol. Sci. 21, 3100 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Canli, O. et al. Glutathione peroxidase 4 prevents necroptosis in mouse erythroid precursors. Blood 127, 139–148 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Koppula, P. et al. A targetable CoQ-FSP1 axis drives ferroptosis- and radiation-resistance in KEAP1 inactive lung cancers. Nat. Commun. 13, 2206 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Xue, J. et al. The Nrf2/GCH1/BH4 axis ameliorates radiation-induced skin injury by modulating the ROS cascade. J. Invest. Dermatol. 137, 2059–2068 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Imai, H. et al. Failure of the expression of phospholipid hydroperoxide glutathione peroxidase in the spermatozoa of human infertile males. Biol. Reprod. 64, 674–683 (2001).

    Article  CAS  PubMed  Google Scholar 

  43. Imai, H. et al. Depletion of selenoprotein GPx4 in spermatocytes causes male infertility in mice. J. Biol. Chem. 284, 32522–32532 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Dasgupta, S. et al. Metabolic enzyme PFKFB4 activates transcriptional coactivator SRC-3 to drive breast cancer. Nature 556, 249–254 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Guo, D. et al. Aerobic glycolysis promotes tumor immune evasion by hexokinase2-mediated phosphorylation of IκBα. Cell Metab. 34, 1312–1324.e6 (2022).

    Article  CAS  PubMed  Google Scholar 

  46. Li, X. et al. A splicing switch from ketohexokinase-C to ketohexokinase-A drives hepatocellular carcinoma formation. Nat. Cell Biol. 18, 561–571 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Du, L. et al. β-Catenin induces transcriptional expression of PD-L1 to promote glioblastoma immune evasion. J. Exp. Med. 217, e20191115 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Qian, X. et al. KDM3A senses oxygen availability to regulate PGC-1α-mediated mitochondrial biogenesis. Mol. Cell 76, 885–895.e7 (2019).

    Article  CAS  PubMed  Google Scholar 

  49. Qian, X. et al. PTEN suppresses glycolysis by dephosphorylating and inhibiting autophosphorylated PGK1. Mol. Cell 76, 516–527.e7 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. Li, X. et al. Nucleus-translocated ACSS2 promotes gene transcription for lysosomal biogenesis and autophagy. Mol. Cell 66, 684–697.e9 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. 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 

  52. Wu, J. et al. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature 572, 402–406 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Xu, D. Q. et al. PAQR3 controls autophagy by integrating AMPK signaling to enhance ATG14L-associated PI3K activity. EMBO J. 35, 496–514 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Xu, D. et al. PAQR3 modulates cholesterol homeostasis by anchoring Scap/SREBP complex to the Golgi apparatus. Nat. Commun. 6, 8100 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Yang, W. et al. Nuclear PKM2 regulates β-catenin transactivation upon EGFR activation. Nature 480, 118–122 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lee, J. H. et al. EGFR-phosphorylated platelet isoform of phosphofructokinase 1 promotes PI3K activation. Mol. Cell 70, 197–210.e7 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported by grants from the Ministry of Science and Technology of the People’s Republic of China (2021YFA0805600, D.X.; 2020YFA0803300, Z.L.), the National Natural Science Foundation of China (82188102 and 82030074, Z.L.; 92157113 and 82072630, D.X.; 82173114, Z.W.; 82072903 and 82272872, T.L.; 82002811, M.Y.; 82030065 and 81873932, Q.Z.), the Zhejiang Natural Science Foundation Key Project (LD22H160002, D.X.; LD21H160003, Z.L.), Zhejiang Natural Science Foundation Discovery Project (LQ22H160023, Z.W.), Chinese Academy of Sciences Intramural Research Program (QTJC20220010-A, Q.X.) and the Leading Innovative and Entrepreneur Team Introduction Program of Zhejiang (2019R01001, Z.L.). Z.L. is the Kuancheng Wang Distinguished Chair. The authors received no specific funding for this work.

Author information

Authors and Affiliations

Authors

Contributions

D.X., Z.L. and Q.Z. conceived and designed the study and wrote the paper. K.W., M.Y., T.L., Z.W., Y.D., G.J., Y.S., L.W., L.L., P.Z., B.D., X.Y., H.S., T.W., J.Z., J.Y. and Y.D. performed the experiments and analysed the data. Y.X., Q.W. and L.X. reviewed and edited the paper. X.Q., L.M. and J.F. provided the technical support in gene editing and interpretation of the data.

Corresponding authors

Correspondence to Qin Zhou, Zhimin Lu or Daqian Xu.

Ethics declarations

Competing interests

Z.L. owns shares in Signalway Biotechnology (Pearland, TX), which supplied rabbit antibodies that recognize CKB pT133 and GPX4 pS104. Z.L.’s interest in this company had no bearing on its being chosen to supply these reagents. The other authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Lawrence Kazak and the other, anonymous, reviewer(s) 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 CKB T133 phosphorylation upregulates GPX4 stability.

(b, d-h, l,m, r) Immunoprecipitation and immunoblotting analyses were performed with the indicated antibodies. (a, b, q-s) Data are the mean ± SD (n = 6). N.S., not significant (a, q, s); **P < 0.001 (r); ***P < 0.0001(b) by two-tailed Student’s t-test. (a, m, q-s) Huh7 and HCCLM3 were treated with IGF1 for the indicated time (a). The indicated cells expressing CKB shRNA with reconstituted expression of the indicated CKB proteins were harvested (m, s), or treated with IGF1 for 12 h (q), or treated with CHX for the indicated time (r). The mRNA expression levels of Gpx4 gene were measured using qPCR (a, q, s). (b) Huh7 and HCCLM3 cells were treated with CHX for the indicated time in the presence of IGF1. The quantification of GPX4 protein levels relative to initial protein levels is shown. (c) Selected mass spectrometry identified peptide hits are shown. (d) Huh7 cells were pretreated with or without indicated inhibitors for 30 min before treatment with or without IGF1 for 1 h. (e) Huh7 cells transfected with or without the indicated plasmid were treated with or without Tris DBA (10 μM) for 12 h. (f) Huh7 cells were treated with or without IGF1 (100 ng/ml) for 1 h. (g) A GST pull-down assay was performed as indicated. (h) In vitro kinase assays were performed in the presence of [γ-32P] ATP. Autoradiography were performed. (i) Purified GST-CKB proteins were incubated with His-AKT1 for an in vitro kinase assay. Mass-spectrometric analysis was performed. (j) Alignment of protein sequences spanning CKB T133 from different species. (k, l) Huh7 cells expressing Flag-CKB were treated with or without IGF1 for 1 h (l). IHC analyses of human HCC samples (k) or immunoblotting (l) were performed with the indicated antibodies and a CKB pT133-blocking peptide. (n-p) Genomic DNA was extracted from two individual clones of indicated cells with knock-in expression of CKB T133A. PCR products amplified from the indicated DNA fragments were shown (n, o) and sequenced (p).

Source data

Extended Data Fig. 2 GPX4 S104 phosphorylation stabilizes GPX4.

(a,b, h, j, k, p, t-x) Immunoprecipitation and immunoblotting analyses were performed with the indicated antibodies. (a-e, o, q-s, v-x) Data are the mean ± SD (n = 6). **P < 0.001 (v-x); ***P < 0.0001 (a-e, o, q-s, v-x); N.S., not significant (b, c, r, s) by two-tailed Student’s t-test. (a) The indicated cells treated with IGF1 for 1 h were harvested. (b-f) The indicated purified His-CKB on Ni-NTA agarose beads were incubated with active GST-AKT1 for an in vitro kinase assay. The binding affinity of CKB to creatine (b, d) and the relative CKB activity (c, e) were measured. AKT-phosphorylated CKB conjugated on Ni-NTA beads were washed and incubated with His-GPX4 proteins for an in vitro kinase assay. Mass-spectrometric analysis was performed (f). (g) Alignment of protein sequences spanning GPX4 S104. (h) Purified His-CKB were incubated with GST-GPX4 in the presence of [γ-32P] ATP for in vitro kinase assay. Autoradiography was performed. (i, j) Huh7 cells expressing Flag-GPX4 were treated with IGF1 for 1 h (j). IHC analyses of human HCC samples (i) or immunoblotting (j) were performed with the indicated antibodies and GPX4 pS104-blocking peptide. (k) Huh7 cells expressing Flag-GPX4 were transfected with HA-myr-AKT1. (l-n) Genomic DNA was extracted from the indicated cells. Amplified PCR products were shown (l, m) and sequenced (n). (o, p) Huh7 cells were pretreated with NAC (5 mM) for 30 min before IGF1 incubation for 12 h. The intercellular ROS levels were measured (o). (q-s) Relative CKB activity (q), binding affinity of the His-CKB proteins to creatine (r) and ATP (s) were measured. (t, u) Huh7 or HCCLM3 cells expressing CKB shRNA with reconstituted expression of the indicated CKB proteins were treated with or without IGF1 for 1 h . (v-x) The indicated cells expressing CKB shRNA with reconstituted expression of the indicated CKB proteins (v) or with knock-in expression of GPX4 mutants (w, x) were treated with CHX for the indicated time. The quantification of GPX4 protein levels relative to initial protein levels is shown.

Source data

Extended Data Fig. 3 CKB-mediated GPX4 S104 phosphorylation stabilizes GPX4.

(a, b) Alignment of protein sequences spanning GPX4 S104 and the adjacent CMA-target motif from different species. The structure of GPX4 exhibiting CMA-target motif and S104. (c) Purified His-CKB WT or T133D proteins on Ni-NTA beads were incubated with the indicated GST-GPX4 proteins for an in vitro kinase assay. Then the beads were removed and the remaining GST-GPX4 proteins were incubated with His-HSC70 followed by a GST pulldown assay as indicated. (d, e) Huh7 and HCCLM3 cells expressing GPX4 shRNA with reconstituted expression of the indicated GPX4 proteins were harvested for immunoblotting and mRNA detection using quantitative PCR as indicated. Data are the mean ± SD (n = 6). N.S., not significant (two-tailed Student’s t-test).

Source data

Extended Data Fig. 4 GPX4 S104 phosphorylation suppresses Lipid peroxidation.

(a-d) Parental Huh7 and HCCLM3 cells and the indicated clones with knock-in expression of CKB T133A mutants (upper) or GPX4 S104A (lower) were treated with or without cystine deprivation (a, c) or 20 μM Erastin (b, d) and IGF1 for 24 h. Reduced GSH (a, b) and GSSG (c, d) were measured respectively. Data are the mean ± SD (n = 6), ^P < 0.05; *P < 0.01; **P < 0.001; ***P < 0.0001 by two-tailed Student’s t-test. (e) Parental Huh7 and HCCLM3 cells and the indicated clones with knock-in expression of CKB T133A mutants (left) or GPX4 S104A (right) were treated with or without cystine deprivation or 20 μM Erastin in the presence or absence of IGF1 (100 ng/ml) for 24 h. The cell lysates were harvested for immunoblotting analyses as indicated. (f-k) Parental HCCLM3 (f, g, j,k) or Huh7 (h, i) cells and the indicated clones with knock-in expression of CKB T133A or GPX4 S104A mutants were treated with or without cystine deprivation (f, h, j) or 20 μM Erastin (g, i, k) combined with Fer-1 (2 μM) in the absence or presence of IGF1 for 24 h. Lipid ROS-positive cells were measured. Data are the mean ± SD (n = 3), *P < 0.01 by two-tailed Student’s t-test (f, g). Lipid peroxidation was assessed using BODIPY 581/591 C11 staining followed by FACS analysis.

Source data

Extended Data Fig. 5 GPX4 S104 phosphorylation suppresses ferroptosis.

(a, b, d-k) Data are the mean ± SD, *P < 0.01; **P < 0.001;***P < 0.0001 by two-tailed Student’s t-test. (a, b, d-g) Parental Huh7 cells and the indicated clones with knock-in expression of CKB T133A or GPX4 S104A mutants were treated with or without cystine deprivation (a, d, f) or 20 μM Erastin (b, e, g) in the presence or absence of IGF1 (100 ng/ml) for 24 h. The indicated mRNA expression levels were measured using quantitative PCR. GSSG (d, e) and reduced GSH (f, g) were measured respectively (n = 6). (c) Huh7 cells stably transfected with GLRX and GSTA1 shRNA were harvested for immunoblotting analyses as indicated. (h, i) Parental HCCLM3 cells and the indicated clones with knock-in expression of CKB T133A or GPX4 S104A mutants were treated with or without cystine deprivation (h) or 20 μM Erastin (i) combined with Fer-1 (2 μM) in the absence or presence of IGF1 for 24 h. Cell death were measured respectively (n = 3). (j, k) Parental HCCLM3 cells and the indicated clones with knock-in expression of CKB T133A mutants or GPX4 S104A were treated with different doses of sulfasalazine (SAS) (j) or Erastin (k) for 24 h, cell viability were measured (n = 3). (l) Parental Huh7 and HCCLM3 cells and the indicated clones with knock-in expression of CKB T133A mutants were treated with or without IGF1 for 12 h. Immunoblotting analyses were performed with the indicated antibodies.

Source data

Extended Data Fig. 6 CKB suppresses ferroptosis in a GPX4 pS104 dependent manner.

(a-l) Data are the mean ± SD (n = 3), *P < 0.01; **P < 0.001; N.S., not significant by two-tailed Student’s t-test. (a-d) Huh7 and HCCLM3 cells expressing GPX4 shRNA with reconstituted expression of the indicated GPX4 proteins were treated with or without cystine deprivation (a, c) or 20 μM Erastin (b, d) for 24 h. Lipid ROS-positive cells (a, b) and cell death (c, d) were measured respectively. (e, f) Huh7 and HCCLM3 cells expressing GPX4 shRNA with reconstituted expression of the indicated GPX4 proteins were treated with different doses of sulfasalazine (SAS) (e) or Erastin (f) for 24 h, cell viability were measured. (g-l) Parental Huh7 cells and the indicated clones with knock-in expression of GPX4 S104A mutants were stably transfected with CKB shRNA and reconstituted with indicated CKB proteins. The cells were treated with or without cystine deprivation (g, i) or 20 μM Erastin (h, j) for 24 h. Lipid ROS-positive cells (g, h) and cell death (i, j) were measured respectively. The cells were treated with different doses of sulfasalazine (SAS) (k) or Erastin (l) for 24 h, cell viability were measured.

Source data

Extended Data Fig. 7 CKB suppresses ferroptosis through its protein kinase activity.

(a-l) Data are the mean ± SD (n = 3), ^ P < 0.05; *P < 0.01; **P < 0.001; N.S., not significant by two-tailed Student’s t-test. (a-d) Huh7 and HCCLM3 cells expressing IGF1R-CA and CKB shRNA with reconstituted expression of the indicated CKB proteins were treated with or without cystine deprivation (a, c) or 20 μM Erastin (b, d) for 24 h. Lipid ROS-positive cells (a, b) and cell death (c, d) were measured respectively. (e, f) Huh7 and HCCLM3 cells expressing IGF1R-CA and CKB shRNA with reconstituted expression of the indicated CKB proteins were treated with different doses of sulfasalazine (SAS) (e) or Erastin (f) for 24 h, cell viability were measured. (g-j) Parental Huh7 and HCCLM3 cells and the indicated clones with knock-in expression of CKB T133A (g, i) or GPX4 S104A mutants (h, j) were treated with or without 2 μM RSL3 for 8 h. Lipid ROS-positive cells (g, h) and cell death (i, j) were measured respectively. (k, l) Parental Huh7 and HCCLM3 cells and the indicated clones with knock-in expression of CKB T133A (k) and GPX4 S104A mutants (l) were treated with different doses of RSL3 for 8 h, cell viability were measured.

Source data

Extended Data Fig. 8 CKB phosphorylates GPX4 to inhibit ferroptosis.

(a, b) MDA-MB-231, A549 and HCT116 cells expressing CKB shRNA with reconstituted expression of the indicated CKB proteins were treated with or without IGF1 for 12 h. Immunoblotting analyses were performed with the indicated antibodies. (c, d) MDA-MB-231, A549 and HCT116 cells expressing CKB shRNA with reconstituted expression of the indicated CKB proteins were treated with or without 20 μM Erastin in the absence or presence of IGF1 for 24 h. Lipid ROS-positive cells (c) and cell death (d) were measured respectively. Data are the mean ± SD (n = 3), ^ P < 0.05; *P < 0.01; **P < 0.001 by two-tailed Student’s t-test. (e-g) Genomic DNA was extracted from parental MDA-MB-231, A549 and HCT116 cells and the indicated clones with knock-in expression of GPX4 S104A. PCR products amplified from the indicated DNA fragments were shown (e, f) and sequenced (g). (h) Parental MDA-MB-231, A549 and HCT116 cells and the indicated clones with knock-in expression of GPX4 S104A mutants were treated with or without IGF1 for 12 h. Immunoblotting analyses were performed with the indicated antibodies. (i, j) Parental MDA-MB-231, A549 and HCT116 cells and the indicated clones with knock-in expression of GPX4 S104A mutants were treated with or without 20 μM Erastin for 24 h. Lipid ROS-positive cells (i) and cell death (j) were measured respectively. Data are the mean ± SD (n = 3), *P < 0.01; **P < 0.001 by two-tailed Student’s t-test.

Source data

Extended Data Fig. 9 AKT-CKB-GPX4 axis suppresses ferroptosis.

(a) The whole cell lysates of LN18, LN229, U87 and U251 were harvested for immunoblotting analyses as indicated. (b, c) LN18, LN229, U87 and U251 cells were treated with or without 20 μM Erastin combined with Fer-1 (2 μM) for 24 h. Lipid ROS-positive cells (b) and cell death (c) were measured respectively. Data are the mean ± SD (n = 3), *P < 0.01; **P < 0.001; *** P < 0.0001 by two-tailed Student’s t-test. (d) U251 and U87 cells were treated with or without MK-2206 (5 μM) for 12 h. Immunoblotting analyses were performed with the indicated antibodies. (e, f) Parental U251 and U87 cells were treated with or without 20 μM Erastin in the absence or presence of MK-2206 (5 μM) for 24 h. Lipid ROS-positive cells (e) and cell death (f) were measured respectively. Data are the mean ± SD (n = 3), *P < 0.01; **P < 0.001 by two-tailed Student’s t-test.

Source data

Extended Data Fig. 10 CKB-mediated GPX4 phosphorylation promotes tumour growth.

(a, b, e-i, k, l, n, o, q) Data are the mean ± SD, ^P < 0.05; *P < 0.01; **P < 0.001; ***P < 0.0001; N.S., not significant by two-tailed Student’s t-test (a, e, g, h, i, l, o) and by two-tailed Mann-Whitney U test (b, f, k, n, q). (b, f, j, m, p) IHC analyses of the indicated xenograft tumors from nude mice were performed with the indicated antibodies. Representative staining images and staining scores are shown. (a, b) Huh7 cells were subcutaneously injected into 6-week-old male athymic nude mice. When the tumor reached 50 mm3, the mice were assigned randomly into different treatment groups. Sulfasalazine (SAS) and Liproxstatin-1 (Lip-1) were intraperitoneally injected daily at a dose of 100 mg/kg and 10 mg/kg respectively until the endpoint at Day 28. Tumor volume and weight were analyzed (n = 7) (a). (c, r) The whole cell lysates of WT and GPX4 knockout Huh7 cells (c) and L02, THLE-2, Huh7, HCCLM3 (r) were harvested for immunoblotting analyses as indicated. (d-f) WT and GPX4 knockout Huh7 cells were subcutaneously injected into the left or right flanks of 6-week-old male athymic nude mice with and without daily Lip-1 intraperitoneal injection from the 4th day. The resulting tumors were resected 28 days after injection (d). Tumor volume and weight were analyzed (n = 6) (e). (g-q) IGF1R CA-expressing parental Huh7 cells and the indicated clones with knock-in expression of CKB T133A (g) and GPX4 S104A (h) or Huh7 cells expressing CKB shRNA with reconstituted expression of the indicated CKB proteins (i-k) or Parental Huh7 cells and the indicated clones with knock-in expression of GPX4 S104D mutants stably transfected with CKB shRNA and reconstituted with indicated CKB proteins (l-q) were intrahepatically (g, h) or subcutaneously (i-q) injected into 6-week-old male athymic nude mice (n = 7). When the tumor reached 50 mm3, the mice were assigned randomly into different treatment groups. SAS was intraperitoneally injected daily at a dose of 100 mg/kg until the endpoint at Day 28. Tumor volume and weight were analyzed.

Source data

Supplementary information

Source data

Source Data Fig. 1

Unprocessed WBs.

Source Data Fig. 2

Unprocessed WBs.

Source Data Fig. 3

Unprocessed WBs.

Source Data Extended Data Fig. 1

Unprocessed WBs.

Source Data Extended Data Fig. 2

Unprocessed WBs.

Source Data Extended Data Fig. 3

Unprocessed WBs.

Source Data Extended Data Fig. 4

Unprocessed WBs.

Source Data Extended Data Fig. 5

Unprocessed WBs.

Source Data Extended Data Fig. 8

Unprocessed WBs.

Source Data Extended Data Fig. 9

Unprocessed WBs.

Source Data Extended Data Fig. 10

Unprocessed WBs.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Statistical source data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical source data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical source data for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Statistical source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Statistical source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical source data for Extended Data Fig. 9.

Source Data Extended Data Fig. 10

Statistical source data for Extended Data Fig. 10.

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

Wu, K., Yan, M., Liu, T. et al. Creatine kinase B suppresses ferroptosis by phosphorylating GPX4 through a moonlighting function. Nat Cell Biol 25, 714–725 (2023). https://doi.org/10.1038/s41556-023-01133-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01133-9

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing