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:

LPCAT1-mediated membrane phospholipid remodelling promotes ferroptosis evasion and tumour growth

Abstract

The mechanisms underlying the dynamic remodelling of cellular membrane phospholipids to prevent phospholipid peroxidation-induced membrane damage and evade ferroptosis, a non-apoptotic form of cell death driven by iron-dependent lipid peroxidation, remain poorly understood. Here we show that lysophosphatidylcholine acyltransferase 1 (LPCAT1) plays a critical role in ferroptosis resistance by increasing membrane phospholipid saturation via the Lands cycle, thereby reducing membrane levels of polyunsaturated fatty acids, protecting cells from phospholipid peroxidation-induced membrane damage and inhibiting ferroptosis. Furthermore, the enhanced in vivo tumour-forming capability of tumour cells is closely associated with the upregulation of LPCAT1 and emergence of a ferroptosis-resistant state. Combining LPCAT1 inhibition with a ferroptosis inducer synergistically triggers ferroptosis and suppresses tumour growth. Therefore, our results unveil a plausible role for LPCAT1 in evading ferroptosis and suggest it as a promising target for clinical intervention in human cancer.

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: Persistent treatment with either RSL3 or erastin induces membrane phospholipid saturation.
Fig. 2: RSL3-/erastin-resistant cells preferentially incorporate SFA-PLs into their membranes.
Fig. 3: LPCAT1 increases the degree of membrane phospholipid saturation.
Fig. 4: Ablation of LPCAT1 sensitizes cells to ferroptosis.
Fig. 5: LPCAT1-mediated ferroptosis inhibition relies on exogenous and endogenous SFAs.
Fig. 6: LPCAT1 promotes tumour development by ferroptosis evasion.
Fig. 7: Inhibition of LPCAT1 suppresses tumour growth.

Similar content being viewed by others

Data availability

RNA-seq data that support the findings of this study have been deposited in the National Center for Biotechnology Information Sequence Read Archive with the accession code PRJNA987832. Clinical data were obtained from the The Cancer Genome Atlas database. The original western blot images have been provided and are publicly available. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. van Meer, G., Voelker, D. R. & Feigenson, G. W. Membrane lipids: where they are and how they behave. Nat. Rev. Mol. Cell Biol. 9, 112–124 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Wang, B. & Tontonoz, P. Phospholipid remodeling in physiology and disease. Annu. Rev. Physiol. 81, 165–188 (2019).

    Article  PubMed  Google Scholar 

  3. Hazel, J. R. & Williams, E. E. The role of alterations in membrane lipid composition in enabling physiological adaptation of organisms to their physical environment. Prog. Lipid Res. 29, 167–227 (1990).

    Article  CAS  PubMed  Google Scholar 

  4. Matsuki, H., Goto, M. & Tamai, N. Membrane states of saturated glycerophospholipids: a thermodynamic study of bilayer phase transitions. Chem. Pharm. Bull. 67, 300–307 (2019).

    Article  CAS  Google Scholar 

  5. Doll, S. et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 13, 91–98 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Dixon, S. J. et al. Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem. Biol. 10, 1604–1609 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yuan, H., Li, X., Zhang, X., Kang, R. & Tang, D. Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochem. Biophys. Res. Commun. 478, 1338–1343 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Friedmann Angeli, J. P. et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat. Cell Biol. 16, 1180–1191 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Yan, B. et al. Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol. Cell 81, 355–369 (2021).

    Article  CAS  PubMed  Google Scholar 

  10. 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 

  11. Tesfay, L. et al. Stearoyl-CoA desaturase 1 protects ovarian cancer cells from ferroptotic cell death. Cancer Res. 79, 5355–5366 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Magtanong, L. et al. Exogenous monounsaturated fatty acids promote a ferroptosis-resistant cell state. Cell Chem. Biol. 26, 420–432 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Liang, D. et al. Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones. Cell 186, 2748–2764 (2023).

    Article  CAS  PubMed  Google Scholar 

  14. Levental, I. & Lyman, E. Regulation of membrane protein structure and function by their lipid nano-environment. Nat. Rev. Mol. Cell Biol. 24, 107–122 (2023).

    Article  CAS  PubMed  Google Scholar 

  15. MacDonald, J. I. & Sprecher, H. Phospholipid fatty acid remodeling in mammalian cells. Biochim. Biophys. Acta 1084, 105–121 (1991).

    Article  CAS  PubMed  Google Scholar 

  16. Panov, A. V. & Dikalov, S. I. Cardiolipin, perhydroxyl radicals, and lipid peroxidation in mitochondrial dysfunctions and aging. Oxid. Med. Cell. Longev. 2020, 1323028 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  18. 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 

  19. Kraft, V. A. N. et al. GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent. Sci. 6, 41–53 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. 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 

  21. Lands, W. E. & Hart, P. Metabolism of glycerolipids. VI. Specificities of acyl coenzyme A: phospholipid acyltransferases. J. Biol. Chem. 240, 1905–1911 (1965).

    Article  CAS  PubMed  Google Scholar 

  22. Dolma, S., Lessnick, S. L., Hahn, W. C. & Stockwell, B. R. Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell 3, 285–296 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Perez, M. A., Magtanong, L., Dixon, S. J. & Watts, J. L. Dietary lipids induce ferroptosis in Caenorhabditis elegans and human cancer cells. Dev. Cell 54, 447–454 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Das, U. N. Saturated fatty acids, MUFAs and PUFAs regulate ferroptosis. Cell Chem. Biol. 26, 309–311 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Rysman, E. et al. De novo lipogenesis protects cancer cells from free radicals and chemotherapeutics by promoting membrane lipid saturation. Cancer Res. 70, 8117–8126 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Du, Y. et al. Lysophosphatidylcholine acyltransferase 1 upregulation and concomitant phospholipid alterations in clear cell renal cell carcinoma. J. Exp. Clin. Cancer Res. 36, 66 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Morita, Y. et al. Lysophosphatidylcholine acyltransferase 1 altered phospholipid composition and regulated hepatoma progression. J. Hepatol. 59, 292–299 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Mansilla, F. et al. Lysophosphatidylcholine acyltransferase 1 (LPCAT1) overexpression in human colorectal cancer. J. Mol. Med. 87, 85–97 (2009).

    Article  CAS  PubMed  Google Scholar 

  29. 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 

  30. Yi, J., Zhu, J., Wu, J., Thompson, C. B. & Jiang, X. Oncogenic activation of PI3K–AKT–mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc. Natl Acad. Sci. USA 117, 31189–31197 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chen, H. et al. Aspirin promotes RSL3-induced ferroptosis by suppressing mTOR/SREBP-1/SCD1-mediated lipogenesis in PIK3CA-mutant colorectal cancer. Redox Biol. 55, 102426 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhang, Y. et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem. Biol. 26, 623–633 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lei, G. et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30, 146–162 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lee, H. et al. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat. Cell Biol. 22, 225–234 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Abbott, S. K., Else, P. L., Atkins, T. A. & Hulbert, A. J. Fatty acid composition of membrane bilayers: importance of diet polyunsaturated fat balance. Biochim. Biophys. Acta 1818, 1309–1317 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Niki, E., Yoshida, Y., Saito, Y. & Noguchi, N. Lipid peroxidation: mechanisms, inhibition, and biological effects. Biochem. Biophys. Res. Commun. 338, 668–676 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Reed, A. et al. LPCAT3 inhibitors remodel the polyunsaturated phospholipid content of human cells and protect from ferroptosis. ACS Chem. Biol. 17, 1607–1618 (2022).

    Article  CAS  PubMed  Google Scholar 

  39. Sun, W. Y. et al. Phospholipase iPLA2β averts ferroptosis by eliminating a redox lipid death signal. Nat. Chem. Biol. 17, 465–476 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bi, J. et al. Oncogene amplification in growth factor signaling pathways renders cancers dependent on membrane lipid remodeling. Cell Metab. 30, 525–538 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Abdelzaher, E. & Mostafa, M. F. Lysophosphatidylcholine acyltransferase 1 (LPCAT1) upregulation in breast carcinoma contributes to tumor progression and predicts early tumor recurrence. Tumour Biol. 36, 5473–5483 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Uehara, T. et al. Overexpression of lysophosphatidylcholine acyltransferase 1 and concomitant lipid alterations in gastric cancer. Ann. Surg. Oncol. 23, S206–S213 (2016).

    Article  PubMed  Google Scholar 

  43. Liu, S. et al. Combination RSL3 treatment sensitizes ferroptosis- and EGFR-inhibition-resistant HNSCCs to cetuximab. Int. J. Mol. Sci. 23, 9014 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Yao, F. et al. A targetable LIFR–NF-κB–LCN2 axis controls liver tumorigenesis and vulnerability to ferroptosis. Nat. Commun. 12, 7333 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. 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

This work was supported by the National Natural Science Foundation of China (grant numbers 82030078 and 82330082 to J.L., and U23A20455 to L.S.), the Shenzhen Medical Research Fund (grant number B2302046 to J.L.), the National Key Research and Development Program of China (grant number 2020YFA0509400 to L.S.) and the Fundamental Research Funds for the Central Universities (grant number 23ykcxqt001 to J.L.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Project planning was done by J.L. and Z.L. Z.L., Y.H. and H.Z. performed most experiments, analysed data and wrote the paper. M.L., Y.L., R.F., M.Y. and X. Li. performed the animal experiments. S.Z., M.T., X. Liao, R.Y. and Y.X. performed the cell biology experiments. S.C., W.Q. and Q.Z. performed the lipidomic analyses. D.T. discussed experiments and edited the manuscript. B.L., L.S. and J.L. conceived the idea, designed and discussed experiments, supervised progress and extensively edited and communicated regarding the manuscript.

Corresponding authors

Correspondence to Bo Li, Libing Song or Jun Li.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Reviewer Recognition

Nature Cell Biology thanks Boyi Gan, Graeme Lancaster and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Persistent RSL3 or erastin induces membrane PL saturation.

a, Viability analysis of the indicated cells treated with RSL3 or erastin. b–e, Relative cellular levels of GSH (b, d), DCFH-DA (c), and Ferro-Orange (e) were examined in the indicated RSL3- or erastin-treated cells. f, IB analysis of GPX4, FSP1, GCH1, DHODH, LPCAT3 and ACSL4 expression in the indicated cells. GAPDH served as a loading control. Data are presented as mean ± SD, n = 3 biologically independent samples in a, f, n = 6 biologically independent samples in b-e. Statistical analysis was performed using one-way ANOVA followed by Dunnett’s test in b-e, two-way ANOVA followed by Tukey’s test in a. NS, not significant.

Source data

Extended Data Fig. 2 The PL composition is rapidly modulated by extracellular FFAs.

a–h, Volcano plots of lipidomics analysis of the indicated cells treated with PA (5 μM, 10 h), or OA (5 μM, 10 h), or ALA (5 μM, 10 h), or AA (5 μM, 10 h). Cutoff: FC threshold = 2, P < 0.05. i-j, Volcano plots of lipidomics analysis of the indicated cells treated with mixed PA (5 μM, 10 h), OA (5 μM, 10 h), ALA (5 μM, 10 h), and AA (5 μM, 10 h). Cutoff: FC threshold = 2, P < 0.05. Data are presented as mean ± SD, n = 3 biologically independent samples in a-j. Statistical analysis was performed using an unpaired two-tailed Student’s t-test in a-j.

Extended Data Fig. 3 RSL3-/erastin-resistant cells prefer SFA into membrane PL.

a, Heatmaps showing the metabolite content as detected by metabolomics. b–d, Representative images (b, c) and quantification (d) of membrane azide-fluor 488 fluorescence intensity in the indicated cells treated with PA-alkyne (5 μM, 10 h), or AA-alkyne (5 μM, 10 h), or ALA-alkyne (5 μM, 10 h), or AA-alkyne (5 μM, 10 h) normalized against integrin β1 fluorescence intensity. Scale bar, 10 μm or 2 μm. e-g, Quantification of RSL3- or erastin-induced membrane Liperfluo fluorescence intensity in the indicated cells treated with different FAs normalized against Dil intensity. h, Viability analysis of the indicated cells treated with different FFAs plus RSL3 or erastin for 24h. Data are presented as mean ± SD, n = 3 biologically independent samples in a, h, n = 6 biologically independent samples in b-g. Statistical analysis was performed using an unpaired two-tailed Student’s t-test in h, and one-way ANOVA followed by Dunnett’s test in d-h. NS, not significant.

Source data

Extended Data Fig. 4 LPCAT1 is upregulated in RSL3-/erastin-resistant cells.

a, Quantitative real-time (RT)-PCR analysis of expression of LPCAT1, ATRX, SPAG4, FGF2, MTMR11, S100A16, CD68, HSF4, TCIRG1, and UPP1 in the cells transfected with the corresponding siRNA. b, IB analysis of LPCAT1 expression in the indicated cells. GAPDH served as a loading control. c, Heatmaps showing the integration of FFA-alkyne into the membrane of the indicated cells. d, e, Scatter plots showing the changes in levels of phospholipids in the indicated cells vs. parental cells. FC, fold change. Cutoff: FC threshold = 2, P < 0.05. f–i, Repartition (left) and levels (right) of SFA, MUFA, and PUFA in phospholipids in the indicated cells. j, Representative images (left) and quantification (right) of membrane azide-fluor 488 fluorescence intensity in the indicated cells treated with PA-alkyne (5 μM, 10 h) or AA-alkyne (5 μM, 10 h) normalized against integrin β1 fluorescence intensity. Scale bar, 10 μm or 2 μm. Data are presented as mean ± SD, n = 3 biologically independent samples in a-i, n = 6 biologically independent samples in j. Statistical analysis was performed using an unpaired two-tailed Student’s t-test in d-i, one-way ANOVA followed by Dunnett’s test in a, j. NS, not significant.

Source data

Extended Data Fig. 5 LPCAT1 increases the degree of saturation of membrane PL.

a, Representative images of membrane azide-fluor 488 and integrin β1 fluorescence intensity in the indicated cells treated with ALA-alkyne (5 μM, 10 h) or OA-alkyne (5 μM, 10 h) normalized against integrin β1 fluorescence intensity. Scale bar, 10 μm or 2 μm. b-c, Quantification of membrane azide-fluor 488 fluorescence intensity in the indicated cells treated with PA-alkyne (5 μM, 10 h), or OA-alkyne (5 μM, 10 h), or ALA-alkyne (5 μM, 10 h), or AA-alkyne (5 μM, 10 h) normalized against integrin β1 fluorescence intensity. d-f, Relative membrane PA-488 intensity was examined in RSL3 or erastin-treated cells at the indicated times normalized against integrin β1 fluorescence intensity. Data are presented as mean ± SD, n = 6 biologically independent samples in a-f. Statistical analysis was performed using one-way ANOVA followed by Dunnett’s test in b-c. NS, not significant.

Source data

Extended Data Fig. 6 LPCAT1-mediated ferroptosis independent of other pathways.

a, Quantification of membrane Liperfluo fluorescence intensity in the indicated cells treated with RSL3 or erastin for 10 h normalized against Dil intensity. b, IB analysis of GPX4, FSP1, GCH1, DHODH, LPCAT3 and ACSL4 expression in the indicated cells. GAPDH served as a loading control. Protein quantified against controls, set as 1.0. c–h, Relative cellular levels of DCFH-DA (c, d), Ferro-Orange (e, f), and GSH (g, h) were examined in the indicated cells. Data are presented as mean ± SD, n = 3 biologically independent samples in b, n = 6 biologically independent samples in a and c-h. Statistical analysis was performed using an unpaired two-tailed Student’s t-test in a, d, f, h, one-way ANOVA followed by Dunnett’s test in c, e, g. NS, not significant.

Source data

Extended Data Fig. 7 Ablation of LPCAT1 sensitizes cells to ferroptosis.

a, b, Cell death was quantified by PI staining coupled with flow cytometry in the indicated cells treated with RSL3 or erastin for 24 h. c, d, Left: Viability analysis of the indicated cells treated with erastin for 24 h; Right: IB analysis of LPCAT1 expression in the indicated cells. GAPDH served as a loading control. e, Representative images of the indicated cells treated with erastin for 24 h. f, Transmission electron microscopy images showing mitochondrial crest in cells treated with erastin for 24 h. Scale bar, 400 nm (left) and 50 nm (right). g, h, Viability analysis of the cells treated with the indicated ferroptosis inducers. i-k, Viability analysis of the indicated cells treated with RSL3 or erastin plus the indicated cell death inhibitors. Data are presented as mean ± SD, n = 3 biologically independent samples in a-k. Statistical analysis was performed using one-way ANOVA followed by Dunnett’s test in a-b, g-k, and two-way ANOVA followed by Tukey’s test in c, d. NS, not significant.

Source data

Extended Data Fig. 8 LPCAT1 inhibits ferroptosis rely exogenous or endogenous SFA.

a-b, left: Viability analysis of the indicated cells treated with RSL3 for 24 h. Right: IB analysis of LPCAT1 and LPCAT3 expression in the indicated cells. GAPDH served as a loading control. c-d, Viability analysis of the indicated cells, which were pretreated with the indicated concentration of TOFA for 24 h, followed by treatment with erastin or erastin plus ferrostatin-1 for 24 h, in the DFBS medium. e, Viability analysis of the indicated cells, which were pretreated with the indicated concentration of CAY10566 for 24 h, followed by treatment with erastin or erastin plus ferrostatin-1 for 24 h, in the DFBS medium. f, Viability analysis of the indicated cells, which were pretreated with the indicated concentration of palmitic acid and TOFA for 24 h, followed by erastin treatment for 24 h, in the DFBS medium. g, IB analysis of LPCAT1 expression in the indicated cells. h–k, Effects of ferroptosis inducer erastin on the growth of 3D tumour spheroids formed by the indicated cells treated with vehicle or with PA (50 mM) at the indicated time. Data are presented as mean ± SD, n = 3 biologically independent samples in a-k. Statistical analysis was performed using one-way ANOVA followed by Dunnett’s test in c-f, h-k, and two-way ANOVA followed by Tukey’s test in a-b. NS, not significant.

Source data

Extended Data Fig. 9 LPCAT1 promotes tumour development by ferroptosis evasion.

a, IB analysis of LPCAT1 expression in the indicated cancer cells. GAPDH served as a loading control. Protein quantified against controls, set as 1.0. b, Viability analysis of the indicated cells treated with RSL3 for 24 h. c, Heatmaps showing the knockdown efficiency of LPCAT1 in the indicated cells. d, Viability analysis of the indicated cells treated with RSL3 for 24 h. e, Viability analysis of the indicated cells treated with IKE (2 μM) or IKE (2 μM) plus Fer-1 (4 μM) for 24 h. f–h, Upper: IB analysis of LPCAT1 expression in the indicated tumour cells. GAPDH served as a loading control. Lower: Tumour growth curves of the indicated xenografts. n = 5 animals per group. Data are presented as mean ± SD, n = 3 biologically independent samples in a-h. Statistical analysis was performed using one-way ANOVA followed by Dunnett’s test in b, e-h. NS, not significant.

Source data

Extended Data Fig. 10 Inhibition of LPCAT1 suppresses tumour growth.

a, Representative IHC images from the indicated xenograft tumours (Scale bar: 40 μM). b, Staining scores of LPCAT1 and cleaved-caspase 3. Data are presented as mean ± SD, n = 6 biologically independent samples in a-b. Statistical analysis was performed using a two-tailed Student’s t-test and one-way ANOVA followed by Dunnett’s test in b. NS, not significant.

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table 1

Primers and oligonucleotides used in this study.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

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 Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Z., Hu, Y., Zheng, H. et al. LPCAT1-mediated membrane phospholipid remodelling promotes ferroptosis evasion and tumour growth. Nat Cell Biol (2024). https://doi.org/10.1038/s41556-024-01405-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41556-024-01405-y

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