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:

Tumor-induced reshuffling of lipid composition on the endoplasmic reticulum membrane sustains macrophage survival and pro-tumorigenic activity

Abstract

Tumor-associated macrophages (TAMs) display pro-tumorigenic phenotypes for supporting tumor progression in response to microenvironmental cues imposed by tumor and stromal cells. However, the underlying mechanisms by which tumor cells instruct TAM behavior remain elusive. Here, we uncover that tumor-cell-derived glucosylceramide stimulated unconventional endoplasmic reticulum (ER) stress responses by inducing reshuffling of lipid composition and saturation on the ER membrane in macrophages, which induced IRE1-mediated spliced XBP1 production and STAT3 activation. The cooperation of spliced XBP1 and STAT3 reinforced the pro-tumorigenic phenotype and expression of immunosuppressive genes. Ablation of XBP1 expression with genetic manipulation or ameliorating ER stress responses by facilitating LPCAT3-mediated incorporation of unsaturated lipids to the phosphatidylcholine hampered pro-tumorigenic phenotype and survival in TAMs. Together, we uncover the unexpected roles of tumor-cell-produced lipids that simultaneously orchestrate macrophage polarization and survival in tumors via induction of ER stress responses and reveal therapeutic targets for sustaining host antitumor immunity.

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: The TME promotes lipid accumulation in pro-tumorigenic TAMs.
Fig. 2: The TME activates IRE1/sXBP1 in pro-tumorigenic TAMs.
Fig. 3: Tumor cells drive pro-tumorigenic polarization in BMDMs via IRE1.
Fig. 4: Deletion of XBP1 in TAMs suppresses tumor growth.
Fig. 5: Activation of IRE1–STAT3 signal supports CM-induced polarization.
Fig. 6: Mincle-dependent glucosylceramide sensing pathway tailors macrophage activation.
Fig. 7: CM causes reshuffling of lipid composition and saturation of ER membrane.
Fig. 8: LXR agonist reduces tumor burden and hampers TAM survival in a LPCAT3-dependent manner.

Similar content being viewed by others

Data availability

RNA-seq results are available in the Gene Expression Omnibus database under accession code (GSE166735). Other data are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Lopez-Yrigoyen, M., Cassetta, L. & Pollard, J.W. Macrophage targeting in cancer. Ann. N. Y. Acad. Sci. 1499, 18–41 (2021).

    Article  PubMed  Google Scholar 

  2. Anfray, C., Ummarino, A., Andon, F.T. & Allavena, P. Current strategies to target tumor-associated-macrophages to improve anti-tumor immune responses. Cells 9, 46 (2019).

    Article  PubMed Central  CAS  Google Scholar 

  3. Loke, P. & Allison, J. P. PD-L1 and PD-L2 are differentially regulated by Th1 and Th2 cells. Proc. Natl Acad. Sci. USA 100, 5336–5341 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Laoui, D. et al. Tumor hypoxia does not drive differentiation of tumor-associated macrophages but rather fine-tunes the M2-like macrophage population. Cancer Res. 74, 24–30 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Qian, B. Z. & Pollard, J. W. Macrophage diversity enhances tumor progression and metastasis. Cell 141, 39–51 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wenes, M. et al. Macrophage metabolism controls tumor blood vessel morphogenesis and metastasis. Cell Metab. 24, 701–715 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Casazza, A. et al. Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell 24, 695–709 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Pyonteck, S. M. et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 19, 1264–1272 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Topper, M. J. et al. Epigenetic therapy ties MYC depletion to reversing immune evasion and treating lung cancer. Cell 171, 1284–1300 e1221 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Liu, Y. et al. Tumor-repopulating cells induce PD-1 expression in CD8(+) T cells by transferring kynurenine and AhR activation. Cancer Cell 33, 480–494 e487 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Li, X. et al. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat. Rev. Clin. Oncol. 16, 425–441 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Vriens, K. et al. Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity. Nature 566, 403–406 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Chen, M. et al. An aberrant SREBP-dependent lipogenic program promotes metastatic prostate cancer. Nat. Genet. 50, 206–218 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wang, H. et al. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat. Immunol. 21, 298–308 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Herber, D. L. et al. Lipid accumulation and dendritic cell dysfunction in cancer. Nat. Med. 16, 880–886 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Veglia, F. et al. Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature 569, 73–78 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Di Conza, G. & Ho, P.C. ER stress responses: an emerging modulator for innate immunity. Cells 9, 695 (2020).

    Article  PubMed Central  CAS  Google Scholar 

  18. Chen, X. & Cubillos-Ruiz, J.R. Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat. Rev. Cancer 21, 71–88 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Song, M. et al. IRE1alpha-XBP1 controls T cell function in ovarian cancer by regulating mitochondrial activity. Nature 562, 423–428 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ma, X. et al. Cholesterol induces CD8(+) T cell exhaustion in the tumor microenvironment. Cell Metab. 30, 143–156 e145 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Cubillos-Ruiz, J. R. et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell 161, 1527–1538 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Mohamed, E. et al. The unfolded protein response mediator PERK governs myeloid cell-driven immunosuppression in tumors through inhibition of STING signaling. Immunity 52, 668–682 e667 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Batista, A. et al. IRE1alpha regulates macrophage polarization, PD-L1 expression, and tumor survival. PLoS Biol. 18, e3000687 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Heinzer, S., Worz, S., Kalla, C., Rohr, K. & Weiss, M. A model for the self-organization of exit sites in the endoplasmic reticulum. J. Cell Sci. 121, 55–64 (2008).

    Article  CAS  PubMed  Google Scholar 

  25. Zhang, L. et al. Single-cell analyses inform mechanisms of myeloid-targeted therapies in colon cancer. Cell 181, 442–459 e429 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Lambrechts, D. et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat. Med. 24, 1277–1289 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Maynard, A. et al. Heterogeneity and targeted therapy-induced adaptations in lung cancer revealed by longitudinal single-cell RNA sequencing. Preprint at bioRxiv https://doi.org/10.1101/2019.12.08.868828 (2019).

  28. Sanderson, S. M. et al. The Na++/K++ ATPase regulates glycolysis and defines immunometabolism in tumors. Preprint at bioRxiv https://doi.org/10.1101/2020.03.31.018739 (2020).

  29. Liu, P. S. et al. Alpha-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat. Immunol. 18, 985–994 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Etzerodt, A. et al. Specific targeting of CD163(+) TAMs mobilizes inflammatory monocytes and promotes T cell-mediated tumor regression. J. Exp. Med. 216, 2394–2411 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Movahedi, K. et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 70, 5728–5739 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Yan, D., Wang, H. W., Bowman, R. L. & Joyce, J. A. STAT3 and STAT6 signaling pathways synergize to promote cathepsin secretion from macrophages via IRE1alpha activation. Cell Rep. 16, 2914–2927 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Herbeuval, J. P., Lelievre, E., Lambert, C., Dy, M. & Genin, C. Recruitment of STAT3 for production of IL-10 by colon carcinoma cells induced by macrophage-derived IL-6. J. Immunol. 172, 4630–4636 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Liu, Y. et al. Role for the endoplasmic reticulum stress sensor IRE1alpha in liver regenerative responses. J. Hepatol. 62, 590–598 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Erbay, E. et al. Reducing endoplasmic reticulum stress through a macrophage lipid chaperone alleviates atherosclerosis. Nat. Med. 15, 1383–1391 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Rong, X. et al. LXRs regulate ER stress and inflammation through dynamic modulation of membrane phospholipid composition. Cell Metab. 18, 685–697 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Huang, S. C. et al. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nat. Immunol. 15, 846–855 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Su, P. et al. Enhanced lipid accumulation and metabolism are required for the differentiation and activation of tumor-associated macrophages. Cancer Res. 80, 1438–1450 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lee, W. B. et al. Mincle-mediated translational regulation is required for strong nitric oxide production and inflammation resolution. Nat. Commun. 7, 11322 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Li, C. et al. The Mincle/Syk/NF-kappaB signaling circuit is essential for maintaining the protumoral activities of tumor-associated macrophages. Cancer Immunol. Res. 8, 1004–1017 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Clement, M. et al. Necrotic cell sensor Clec4e promotes a proatherogenic macrophage phenotype through activation of the unfolded protein response. Circulation 134, 1039–1051 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Tanaka, M. et al. C-type lectin Mincle mediates cell death-triggered inflammation in acute kidney injury. J. Exp. Med. 217, e20192230. (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Williams, S. J. Sensing lipids with Mincle: structure and function. Front. Immunol. 8, 1662 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Halbleib, K. et al. Activation of the unfolded protein response by lipid bilayer stress. Mol. Cell 67, 673–684 e678 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Kast, H. R., Nguyen, C. M., Anisfeld, A. M., Ericsson, J. & Edwards, P. A. CTP:phosphocholine cytidylyltransferase, a new sterol- and SREBP-responsive gene. J. Lipid Res. 42, 1266–1272 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Hashidate-Yoshida, T. et al. Fatty acid remodeling by LPCAT3 enriches arachidonate in phospholipid membranes and regulates triglyceride transport. eLife 4, e06328 (2015).

    Article  PubMed Central  Google Scholar 

  47. Jalil, A. et al. Revisiting the role of LXRs in PUFA metabolism and phospholipid homeostasis. Int. J. Mol. Sci. 20, 3787 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  48. Tavazoie, M. F. et al. LXR/ApoE activation restricts innate immune suppression in cancer. Cell 172, 825–840 e818 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Vitale, I., Manic, G., Coussens, L. M., Kroemer, G. & Galluzzi, L. Macrophages and metabolism in the tumor microenvironment. Cell Metab. 30, 36–50 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. Poli, V. & Camporeale, A. STAT3-mediated metabolic reprograming in cellular transformation and implications for drug resistance. Front. Oncol. 5, 121 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Tcyganov, E. N. et al. Distinct mechanisms govern populations of myeloid-derived suppressor cells in chronic viral infection and cancer. J. Clin. Invest. 131, e145971 (2021).

    Article  CAS  PubMed Central  Google Scholar 

  52. Ackerman, D. & Simon, M. C. Hypoxia, lipids, and cancer: surviving the harsh tumor microenvironment. Trends Cell Biol. 24, 472–478 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Thomas, C. et al. LPCAT3 deficiency in hematopoietic cells alters cholesterol and phospholipid homeostasis and promotes atherosclerosis. Atherosclerosis 275, 409–418 (2018).

    Article  CAS  PubMed  Google Scholar 

  54. Martinez-Lopez, M. et al. Microbiota sensing by Mincle-Syk axis in dendritic cells regulates interleukin-17 and −22 production and promotes intestinal barrier integrity. Immunity 50, 446–461 e449 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Meeth, K., Wang, J. X., Micevic, G., Damsky, W. & Bosenberg, M. W. The YUMM lines: a series of congenic mouse melanoma cell lines with defined genetic alterations. Pigment Cell Melanoma Res. 29, 590–597 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Yu, Y. R. et al. Disturbed mitochondrial dynamics in CD8(+) TILs reinforce T cell exhaustion. Nat. Immunol. 21, 1540–1551 (2020).

    Article  CAS  PubMed  Google Scholar 

  57. Niwa, H., Yamamura, K. & Miyazaki, J. Efficient selection for high-expression transfectants with a novel eukaryotic vector. Gene 108, 193–199 (1991).

    Article  CAS  PubMed  Google Scholar 

  58. Driscoll, W. S., Vaisar, T., Tang, J., Wilson, C. L. & Raines, E. W. Macrophage ADAM17 deficiency augments CD36-dependent apoptotic cell uptake and the linked anti-inflammatory phenotype. Circ. Res. 113, 52–61 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ho, P. C. et al. Immune-based antitumor effects of BRAF inhibitors rely on signaling by CD40L and IFNgamma. Cancer Res. 74, 3205–3217 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ho, P. C. et al. Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell 162, 1217–1228 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Cox, B. & Emili, A. Tissue subcellular fractionation and protein extraction for use in mass-spectrometry-based proteomics. Nat. Protoc. 1, 1872–1878 (2006).

    Article  CAS  PubMed  Google Scholar 

  62. Tsugawa, H., Kanazawa, M., Ogiwara, A. & Arita, M. MRMPROBS suite for metabolomics using large-scale MRM assays. Bioinformatics 30, 2379–2380 (2014).

    Article  CAS  PubMed  Google Scholar 

  63. Checa, A. et al. Hexosylceramides as intrathecal markers of worsening disability in multiple sclerosis. Mult. Scler. 21, 1271–1279 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Durinck, S. et al. BioMart and Bioconductor: a powerful link between biological databases and microarray data analysis. Bioinformatics 21, 3439–3440 (2005).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank L. Glimcher and G. Hotamisligil for providing XBP1flox/flox mice and D. Sancho for providing Mincle-deficient bone marrows. We thank T. Shimizu and J.-i. Miyazaki for providing plasmid LPCAT3-pCNX2. P.-C.H. is funded by the Swiss Institute for Experiment Cancer Research (ISREC 26075483), European Research Council Staring Grant (802773-MitoGuide), SNSF project grants (31003A_163204 and 31003A_182470), UNIL Interdisciplinary Grant, the Cancer Research Institute (CLIP investigator award and Lloyd J. Old STAR award) and Ludwig Cancer Research. J.W.L. is supported by NIH R01CA1932556. P.P. is supported by the New York University Abu Dhabi Research Enhancement Fund Swedish Research Council (Vetenskapsrådet), Swedish Cancer Society (Cancerfonden). D.M. is supported by French government grants managed by the French National Research Agency under the references ANR-11-LABX-0021, ANR-15-IDEX-0003 and ANR-19-CE14-0020. S.C.-C.H. is supported by Cancer Research Institute CLIP Investigator Award, the VeloSano Pilot Award, the Case Comprehensive Cancer Center American Cancer Society Pilot Grants and the Cleveland Digestive Diseases Research Core Center Pilot Grant (IRG91-022-19, IRG-16-186-21, 1P30DK097948). L.N.R. is supported by Immunology T32 Training Program (AI089474). J.I. is supported by SNSF R’Equip 316030_183377. F.M. is supported by Swiss Cancer league KFS-4230-08-2017.

Author information

Authors and Affiliations

Authors

Contributions

G.D.C. and P.-C.H. designed the research. G.D.C, Y.-R.Y. and C.-H.T. performed in vivo experiments. G.D.C., Y.-R.Y. and X.L. performed in vitro experiments. H.G.-A. and J.I. performed lipidomic experiments and analysis. F.F. performed electron microscopy analyses. L.Z., M.F. and L.N.R. performed western blots. X.X. and P.P. performed computational analysis of RNA sequencing. A.J. and D.M. provided donor mice and bone marrow for bone marrow transplantation experiments. Z.X. and J.W.L. performed computational analysis of single-cell RNA sequencing of human and murine tumor cohorts. S.C.-C.H. and F.M. provided feedback and advice. G.D.C. and P.-C.H. wrote the manuscript.

Corresponding author

Correspondence to Ping-Chih Ho.

Ethics declarations

Competing interests

P.-C.H. is scientific advisory for Elixiron Immunotherapeutics, Acepodia and Novartis. P.-C.H. also receives research support from Roche and Elixiron. J.W.L. is a paid advisor to Restoration Foodworks. The remaining authors declare no competing interests.

Additional information

Peer review information: Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 TAMs display high lipid content and ER stress responses in the inducible Braf/Pten melanoma model.

a, Representative histogram (left) and quantitative plot (right) of BODIPY C12 staining in splenic macrophages (n = 10) or tumor-associated macrophages (TAMs) (n = 11) from YUMM1.7 melanoma bearing mice. Data are pooled from two independent experiment. bh, Analysis of Braf/Pten tumor-bearing mice seven weeks after tamoxifen administration. b, c, Representative histogram (left) and quantitative plot (right) of BODIPY staining (b) and BODIPY C12 staining (c) in splenic macrophages or TAMs isolated from Braf/Pten tumor-bearing mice (n = 5 per group). d, Representative histogram (left) and quantitative plots (right) of BODIPY staining in TAMs gated based on ARG1 expression (n = 10 per group). e, qPCR analysis of mRNA expression of the indicated genes in splenic macrophages and TAMs isolated from Braf/Pten tumor-bearing mice (n = 4 per group). f, Representative histogram (left) and quantitative plot of the abundance (right) of sXBP1+ subset among splenic macrophages and TAMs in Braf/Pten tumor-bearing mice (n = 10 per group). g, Representative histogram (left) and quantitative plot (right) of BODIPY staining in TAMs gated based on sXBP1 expression (n = 10 per group). h, Representative histogram (left) and quantitative plot of the abundance (right) of sXBP1+ cells among ARG1+ and ARG1 TAMs (n = 10 per group). i, Gating strategy applied to define ARG1 + macrophages from spleen and tumor of YUMM1.7 tumor-bearing mice. jl, Representative histogram (j) and quantitative plot of BODIPY staining (k) or sXBP1 (l) in skin-resident macrophages or TAMs from Braf/Pten tumor-bearing mice (n = 10 per group). m, Representative histogram (left) and quantitative plot (right) of pPERK staining in splenic macrophages or tumor-associated macrophages (TAMs) (n = 9 per group) of YUMM1.7 melanoma bearing mice. Data are pooled from two independent experiments. n, qRT-PCR of the indicated genes from sorted splenic macrophages and TAMs (n = 8 per group) isolated from YUMM1.7 tumor-bearing mice. Data are representative of two independent experiments (b, c, e). Data are pooled from two independent experiment (a, d, fh, jn). Each symbol represents one individual. All data are mean ± s.e.m and were analyzed by two-tailed, unpaired Student’s t-test or paired t-test (d, g, h).

Source data

Extended Data Fig. 2 Tumor cells reinforce protumorigenic polarization in macrophages via an IL-4/IL-13 independent manner.

a, Representative histogram (left) and quantitative plot (right) of BODIPY FL C12 staining in BMDM cultured in DMEM (Ctrl) or YUMM1.7 CM (n = 4 per group). Data are representative of two independent experiments. b, c, Representative histogram (left) and quantitative plot (right) of ARG1 (b) and MRC1 (c) expression in BMDMs cultured in DMEM or CM (n = 3 per group). Data are representative of three independent experiments. d, qPCR analysis of mRNA expression of the indicated genes in BMDMs cultured in DMEM or CM for 18 h (n = 3 per group). Data are representative of three independent experiments. e, Proliferation of CFSE-labeled T cells activated with anti-CD3 and anti-CD28 alone or in co-culture with BMDM Naïve or previously exposed to CM in a ratio 2:1 (n = 6). Data are pooled of two independent experiments. f, qPCR analysis of Arg1 mRNA expression in BMDMs treated with IL-4 and IL-13 (10 ng/ml) in the absence or presence of 0.25μg/ml anti-IL-4 and 0.25μg/ml anti-IL-13 neutralizing antibodies for 18 h (n = 3 per group). g, qPCR analysis of mRNA expression of indicated genes in BMDMs treated with CM in the absence or presence of 0.25μg/ml anti-IL-4 and 0.25μg/ml anti-IL-13 neutralizing antibody for 18 h (n = 3 per group). Data are representative results of three independent experiments. h, Multiplex cytokine array was used to determine the concentration of IL-13 (left) and IL-4 (right) in CM from YUMM1.7 and MEF cells. Stand0 to Stand7 show the increased concentration detected by the standard provided by the kit (n = 4). Data are representative of two independent experiments. ik, Representative histogram (left) and quantitative plot (right) of BODIPY staining (i), and protein expression of ARG1 (j), and MRC1 (k) in BMDMs stimulated with regular culture medium (Ctrl) or CM from YUMM1.7 (CM) or MEF (CM MEF) (n = 3 per group). Data are representative results of three independent experiments. l, qPCR analysis of indicated genes in BMDMs exposed to CM or Tunicamycin for 18 h (n = 3 per group). Data are representative results of three independent experiments. m, Quantification of BODIPY staining in BMDMs treated with CM in the absence or presence of 50μM STF083010; Ctrl (n = 7), CM (n = 7), CM-STF (n = 6). Data are pooled from three independent experiments. n, Immunoblots of indicated proteins in BMDM transduced with retrovirus expressing scramble or IRE1-targeting gRNAs. o, qPCR analysis of BIP and sXBP1 mRNA expression in BMDMs treated with 1μγ/ml of Tunicamycin, 1μM thapsigargin and CM for 16 h (n = 3 per group). Data are representative results of three independent experiments. Data are mean ± s.e.m. were analyzed by two-tailed, unpaired Student’s t-test or one-way ANOVA with Tukey’s multiple comparison test (e).

Source data

Extended Data Fig. 3 XBP1 supports protumorigenic polarization in response to cancer-derived stimuli.

a, Immunoblots of indicated proteins in control or XBP1-deficient BMDMs stimulated with or without 1μM thapsigargin for 6 h. Data are representative results of two independent experiments. b, Quantification of BODIPY staining in BMDMs generated from WT (XBP1wt) or KO (XBP1cKO) mice cultured stimulated with regular culture medium (Ctrl) or YUMM1.7 CM (n = 12 per group). Data are pooled from three independent experiments. c, Percentages of TAMs (F4/80+ CD11b+ Gr1-) among CD45+ cells in melanomas from tumor-bearing XBP1wt (n = 5) and XBP1cKO mice (n = 6). Data are representative results of three independent experiments. d, e, Tumor growth (d) and tumor weight (e) of YUMM1.7-OVA melanoma from WT and XBP1cKO mice treated with PBS or with anti-CSF1R as indicated in the methods; XBP1wt PBS (n = 13), XBP1cKO PBS mice (n = 12), XBP1wt αCSF1R (n = 12), XBP1cKO αCSF1R (n = 13). Data are pooled from three independent experiments. f, percentages of TAMs (F4/80+ cells gated on CD11b + Gr1-) among CD45 + cells in the experiment showed in d, e (n = 8 per group for XBP1wt PBS, XBP1cKO PBS, XBP1wt αCSF1R; n = 9 for XBP1cKO αCSF1R). Data are pooled from two independent experiments. g, Representative plots of iTAMs and mTAMs populations in tumor and spleen of YUMM1.7 tumor-bearing. hk, Representative histograms (up) and quantitative plots (down) of PDL1 (h), MHCII (i), sXBP1 (j) and ARG1 (k) expression in iTAMs and mTAMs from YUMM1.7 tumor-bearing mice (n = 6). Data are pooled from two independent experiments. l, Representative plots of iTAMs and mTAMs populations in tumor and spleen of Braf/Pten melanoma-bearing mice. mp, Representative histograms (up) and quantitative plots (down) of PDL1 (m), MHCII (n), sXBP1 (o) and ARG1 (p) expression in iTAMs and mTAMs from Braf/Pten melanoma-bearing mice (n = 5). Data are representative of two independent experiments. Each symbol represents one individual. qt, Tumor growth (q) and tumor weight (r) of B16-OVA melanoma and tumor growth (s) and tumor weight (t) of MC38-OVA colon adenocarcinoma in XBP1wt (n = 9 for B16-OVA and n = 8 for MC38-OVA) or XBP1cKO (n = 10 for B16-OVA and n = 7 for MC38-OVA) mice. Data are pooled from two independent experiments. Data are mean ± s.e.m. were analyzed by two-tailed, unpaired Student’s t-test.

Source data

Extended Data Fig. 4 STAT3 is required for CM-induced protumorigenic polarization.

a, Immunoblots of indicated proteins in BMDMs expressing scramble or STAT3-targeting gRNAs treated with 10 ng/ml IL-6 for 6 h. Data are representative results of two independent experiments. b, c, Immunoblots of indicated proteins (b) and qPCR analysis of mRNA expression of indicated genes (c) in BMDMs treated with CM in the presence of vehicle (CM) or 10μM Stattic (CM + Stattic) (n = 6 for Ctrl and n = 5 for CM and CM + Stattic). Data are representative results of three independent experiments. d, Multiplex cytokine array was used to determine the concentration of IL-10 (left) and IL-6 (right) in CM from YUMM1.7 and MEF cells. Stand0 to Stand7 show the increased concentration detected by the standard provided by the kit (n = 4). Data are representative results of two independent experiments. e, Immunoblot of indicated proteins in BMDMs treated with control media (Ctrl), cancer cell conditioned media (CM) or CM plus 50μM STF081030 for 18 h. Data are representative of two independent experiments. f, Immunoblot of BMDMs treated with control vehicle (Ctrl), cancer cell conditioned media (CM) or tunicamycin (1μg/ml; Tuni.) for the indicated time points. Data are representative of two independent experiments. All data are mean ± s.e.m and were analyzed by two-tailed, unpaired Student’s t-test.

Source data

Extended Data Fig. 5 β-glucosylceramide, rather than cholesterol, is released by tumor cells to mediate the protumorigenic polarization in macrophages.

a, b, Quantification results of Cholesterol (a) (n = 6 per group) and fatty acids (b) (n = 4 per group) on YUMM1.7 CM prior (Yumm1.7) and after treatment with lipid removal reagent (Y1.7 w.o. Lipids). Data are pooled from two independent experiments. c, Immunoblot of YUMM1.7 cells stably transduced with lentivirus expressing short hairpin RNA against scramble or HMGCR sequence. d, qPCR analysis of indicated genes in BMDMs exposed to CM isolated from YUMM1.7 shCTRL or from YUMM1.7 shHMGCR (n = 3). Data are representative of three independent experiments. e, qPCR analysis of mRNA expression of indicated genes in BMDMs treated with CM in the absence or presence of 1μg/ml α-CD36 antibody (n = 3 per group). Data are representative results of two independent experiments. f, Quantification of BODIPY staining in BMDM cultured with regular culture medium (Ctrl) or with YUMM1.7 CM in the absence or presence of 5μg/ml α-Mincle antibody; Ctrl (n = 9), CM (n = 9), CMα-Mincle (n = 8). Data are pooled from three independent experiments. g, h, Immunoblot and quantification of the indicated proteins (g) (n = 4 per group) and qPCR analysis of mRNA expression of ARG1 and MRC1 (h) of WT or Mincle-KO BMDMs exposed to regular culture medium (Ctrl) or or with YUMM1.7 CM for 18 h (n = 9 per group). Data are pooled from three independent experiments. i, Proliferation of CFSE-labeled T cells activated with anti-CD3 and anti-CD28 alone or co-cultured with WT or Mincle-KO BMDMs previously treated with CM in a ratio 2:1 for 72 h; T cells (n = 6), WT (n = 8), ΚΟ (n = 9). Data are pooled from three independent experiments. j, Quantification result of indicated β-glucosylceramide levels from CM derived from YUMM1.7 shCTRL and YUMM1.7 shUGCG cells (n = 3 per group). k, qPCR analysis of the indicated genes in BMDMs treated with Ctrl or CM derived from YUMM1.7 shCTRL and YUMM1.7 shUGCG cells alone or in presence of αMincle antibody (5μg/ml) (n = 3). Data are representative of two independent experiments. lm, Quantification result of indicated β-glucosylceramide levels in serum and tumor interstitital fluid (TIF) isolated from YUMM1.7 melanoma-bearing mice (l) or Braf/Pten melanoma-bearing mice (m) (n = 5 per group). All data are mean ± s.e.m and were analyzed by two-tailed, unpaired Student’s t-test (af), paired Student’s t-test (g, j, lm), one-way ANOVA with Sidak’s multiple comparison test (i), one-way ANOVA with Tukey’s multiple comparison test (k).

Source data

Extended Data Fig. 6 Macrophage-specific ablation of LPCAT3 abolishes GW3965 anti-tumor responses.

a, b, qPCR analysis of mRNA expression of sXBP1 (a) and immunoblot and quantification (b) of indicated proteins in WT or LPCAT3-KO BMDMs exposed to regular culture medium (Ctrl) and YUMM1.7 CM in absence or presence of 3μM GW3965 (n = 9 per group for qPCR and n = 3 per group for immunoblots). Data are pooled from three independent experiments c, Proliferation of CFSE-labeled T cells activated with anti-CD3 and anti-CD28 alone or co-cultured with WT or LPCAT3-KO BMDMs previously treated with YUMM1.7 CM in the absence or presence of GW3965 in a ratio 2:1 for 72 h (n = 3 per group). Data are representative of three independent experiments. d, Illustration of experimental design for bone marrow transplantation. e, qPCR analysis of exon 3 of LPCAT3 gene in LPCAT3fl/fl (WT) and LysM-Cre LPCAT3fl/fl (KO) mice (n = 15). f, Bone marrow was isolated from WT and KO chimeric tumor-bearing mice and the abundance of indicated immune cells was measured by flow cytometry (n = 4). g, Percentage of mTAMs among CD11b+ tumor-infiltrating myeloid cells from YUMM1.7-OVA melanoma treated with either control vehicle or GW3965 in mice transplanted with BM cells from LPCAT3fl/fl (WT) and LysM-Cre LPCAT3fl/fl (KO) mice (WT + Vehicle: n = 9; WT + GW3965: n = 10; KO + Vehicle: n = 10; KO + GW3965: n = 9). Data are pooled from two independent experiments. Each symbol represents one individual. All data are mean ± s.e.m and were analyzed by two-tailed, unpaired Student’s t-test (a, e-g), RM one-way ANOVA with Bonferroni’s multiple comparison test (b), and ordinary one-way ANOVA with Tukey’s multiple comparison test (c).

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1 and 2

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. 3

Uncropped Western Blot

Source Data Fig. 4

Statistical source data

Source Data Fig. 5

Statistical source data

Source Data Fig. 5

Uncropped Western Blot

Source Data Fig. 6

Statistical source data

Source Data Fig. 6

Uncropped Western Blot

Source Data Fig. 7

Statistical source data

Source Data Fig. 7

Uncropped Western Blot

Source Data Fig. 8

Statistical source data

Source Data Fig. 8

Uncropped Western Blot

Source Data Extended Data Fig. 1

Statistical source data

Source Data Extended Data Fig. 2

Statistical source data

Source Data Extended Data Fig. 2

Uncropped Western Blot

Source Data Extended Data Fig. 3

Statistical source data

Source Data Extended Data Fig. 3

Uncropped Western Blot

Source Data Extended Data Fig. 4

Statistical source data

Source Data Extended Data Fig. 4

Uncropped Western Blot

Source Data Extended Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 5

Uncropped Western Blot

Source Data Extended Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 6

Uncropped Western Blot

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Di Conza, G., Tsai, CH., Gallart-Ayala, H. et al. Tumor-induced reshuffling of lipid composition on the endoplasmic reticulum membrane sustains macrophage survival and pro-tumorigenic activity. Nat Immunol 22, 1403–1415 (2021). https://doi.org/10.1038/s41590-021-01047-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01047-4

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