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:

Cholesterol mobilization regulates dendritic cell maturation and the immunogenic response to cancer

Abstract

Maturation of conventional dendritic cells (cDCs) is crucial for maintaining tolerogenic safeguards against auto-immunity and for promoting immunogenic responses to pathogens and cancer. The subcellular mechanism for cDC maturation remains poorly defined. We show that cDCs mature by leveraging an internal reservoir of cholesterol (harnessed from extracellular cell debris and generated by de novo synthesis) to assemble lipid nanodomains on cell surfaces of maturing cDCs, enhance expression of maturation markers and stabilize immune receptor signaling. This process is dependent on cholesterol transport through Niemann–Pick disease type C1 (NPC1) and mediates homeostatic and Toll-like receptor (TLR)-induced maturation. Importantly, we identified the receptor tyrosine kinase AXL as a regulator of the NPC1-dependent construction of lipid nanodomains. Deleting AXL from cDCs enhances their maturation, thus improving anti-tumor immunity. Altogether, our study presents new insights into cholesterol mobilization as a fundamental basis for cDC maturation and highlights AXL as a therapeutic target for modulating cDCs.

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: A cellular reservoir of free cholesterol is essential for DC maturation.
Fig. 2: Cholesterol mobilization is essential for TLR-induced cDC maturation.
Fig. 3: Cholesterol is mobilized to construct cell surface lipid nanodomains that are essential for DC maturation.
Fig. 4: AXL limits cholesterol mobilization, lipid nanodomain assembly and immunogenic receptor function.
Fig. 5: The AXL–NPC1 axis in cDC maturation regulates T cell immunity.
Fig. 6: Cholesterol-dependent cDC maturation influences anti-tumor immunity.
Fig. 7: The effects of cholesterol mobilization on cDC maturation are conserved in humans.

Similar content being viewed by others

Data availability

The scRNA-seq data generated for this study are available using GEO accession number GSE282849.

Code availability

No new code was developed for the completion of this study.

References

  1. Inaba, K. et al. Granulocytes, macrophages, and dendritic cells arise from a common major histocompatibility complex class II-negative progenitor in mouse bone marrow. Proc. Natl Acad. Sci. USA 90, 3038–3042 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Miller, J. C. et al. Deciphering the transcriptional network of the dendritic cell lineage. Nat. Immunol. 13, 888–899 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Steinman, R. M., Hawiger, D. & Nussenzweig, M. C. Tolerogenic dendritic cells. Annu. Rev. Immunol. 21, 685–711 (2003).

    CAS  PubMed  Google Scholar 

  4. Vestre, K. et al. Rab7b regulates dendritic cell migration by linking lysosomes to the actomyosin cytoskeleton. J. Cell Sci. 134, jcs259221 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Randolph, G. J., Angeli, V. & Swartz, M. A. Dendritic-cell trafficking to lymph nodes through lymphatic vessels. Nat. Rev. Immunol. 5, 617–628 (2005).

    CAS  PubMed  Google Scholar 

  6. Del Prete, A. et al. Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cell. Mol. Immunol. 20, 432–447 (2023).

    PubMed  PubMed Central  Google Scholar 

  7. Park, M. D. et al. On the biology and therapeutic modulation of macrophages and dendritic cells in cancer. Annu. Rev. Cancer Biol. 7, 291–311 (2023).

    Google Scholar 

  8. Tordesillas, L. et al. PDL2+ CD11b+ dermal dendritic cells capture topical antigen through hair follicles to prime LAP+ Tregs. Nat. Commun. 9, 5238 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Zhang, Y. et al. Regulation of T cell activation and tolerance by PDL2. Proc. Natl Acad. Sci. USA 103, 11695–11700 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Oh, S. A. et al. PD-L1 expression by dendritic cells is a key regulator of T-cell immunity in cancer. Nat. Cancer 1, 681–691 (2020).

    CAS  PubMed  Google Scholar 

  11. Peng, Q. et al. PD-L1 on dendritic cells attenuates T cell activation and regulates response to immune checkpoint blockade. Nat. Commun. 11, 4835 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Garris, C. S. et al. Successful anti-PD-1 cancer immunotherapy requires T cell–dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity 49, 1148–1161 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Martínez-López, M., Iborra, S., Conde-Garrosa, R. & Sancho, D. Batf3-dependent CD103+ dendritic cells are major producers of IL-12 that drive local TH1 immunity against Leishmania major infection in mice. Eur. J. Immunol. 45, 119–129 (2015).

    PubMed  Google Scholar 

  14. Pulendran, B. et al. Distinct dendritic cell subsets differentially regulate the class of immune response in vivo. Proc. Natl Acad. Sci. USA 96, 1036–1041 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Reis e Sousa, C. et al. In vivo microbial stimulation induces rapid CD40 ligand-independent production of interleukin 12 by dendritic cells and their redistribution to T cell areas. J. Exp. Med. 186, 1819–1829 (1997).

    CAS  PubMed  Google Scholar 

  16. Maier, B. et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 580, 257–262 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Bosteels, V. et al. LXR signaling controls homeostatic dendritic cell maturation. Sci. Immunol. 8, eadd3955 (2023).

    CAS  PubMed  Google Scholar 

  18. Binnewies, M. et al. Unleashing type-2 dendritic cells to drive protective antitumor CD4+ T cell immunity. Cell 177, 556–571 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Bosteels, C. et al. Inflammatory type 2 cDCs acquire features of cDC1s and macrophages to orchestrate immunity to respiratory virus infection. Immunity 52, 1039–1056 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Leader, A. M. et al. Single-cell analysis of human non-small cell lung cancer lesions refines tumor classification and patient stratification. Cancer Cell 39, 1594–1609 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Magen, A. et al. Intratumoral dendritic cell-CD4+ T helper cell niches enable CD8+ T cell differentiation following PD-1 blockade in hepatocellular carcinoma. Nat. Med. 29, 1389–1399 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Pelka, K. et al. Spatially organized multicellular immune hubs in human colorectal cancer. Cell 184, 4734–4752 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Steele, N. G. et al. Multimodal mapping of the tumor and peripheral blood immune landscape in human pancreatic cancer. Nat. Cancer 1, 1097–1112 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhang, Y. et al. Single-cell analyses reveal key immune cell subsets associated with response to PD-L1 blockade in triple-negative breast cancer. Cancer Cell 39, 1578–1593 (2021).

    CAS  PubMed  Google Scholar 

  25. Qian, J. et al. A pan-cancer blueprint of the heterogeneous tumor microenvironment revealed by single-cell profiling. Cell Res. 30, 745–762 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Gerhard, G. M., Bill, R., Messemaker, M., Klein, A. M. & Pittet, M. J. Tumor-infiltrating dendritic cell states are conserved across solid human cancers. J. Exp. Med. 218, e20200264 (2021).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  28. Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Ohno-Iwashita, Y. et al. Perfringolysin O, a cholesterol-binding cytolysin, as a probe for lipid rafts. Anaerobe 10, 125–134 (2004).

    CAS  PubMed  Google Scholar 

  30. Shimada, Y., Maruya, M., Iwashita, S. & Ohno-Iwashita, Y. The C-terminal domain of perfringolysin O is an essential cholesterol-binding unit targeting to cholesterol-rich microdomains. Eur. J. Biochem. 269, 6195–6203 (2002).

    CAS  PubMed  Google Scholar 

  31. Wilhelm, L. P., Voilquin, L., Kobayashi, T., Tomasetto, C. & Alpy, F. Intracellular and plasma membrane cholesterol labeling and quantification using filipin and GFP-D4. Methods Mol. Biol. 1949, 137–152 (2019).

    CAS  PubMed  Google Scholar 

  32. Anderson, H. A. & Roche, P. A. MHC class II association with lipid rafts on the antigen presenting cell surface. Biochim. Biophys. Acta 1853, 775–780 (2015).

    CAS  PubMed  Google Scholar 

  33. Blouin, C. M. et al. Glycosylation-dependent IFN-γR partitioning in lipid and actin nanodomains is critical for JAK activation. Cell 166, 920–934 (2016).

    CAS  PubMed  Google Scholar 

  34. Croce, C. et al. Efficient cholesterol transport in dendritic cells defines optimal exogenous antigen presentation and Toxoplasma gondii proliferation. Front. Cell Dev. Biol. 10, 837574 (2022).

    PubMed  PubMed Central  Google Scholar 

  35. Gassart, A. D. et al. MHC class II stabilization at the surface of human dendritic cells is the result of maturation-dependent MARCH I down-regulation. Proc. Natl Acad. Sci. USA 105, 3491–3496 (2008).

    PubMed  PubMed Central  Google Scholar 

  36. Morana, O. et al. Identification of a new cholesterol-binding site within the IFN-γ receptor that is required for signal transduction. Adv. Sci. 9, e2105170 (2022).

    Google Scholar 

  37. Day, C. A. & Kenworthy, A. K. Functions of cholera toxin B-subunit as a raft cross-linker. Essays Biochem. 57, 135–145 (2015).

    PubMed  PubMed Central  Google Scholar 

  38. Van Heyningen, S. Cholera toxin: interaction of subunits with ganglioside GM1. Science 183, 656–657 (1974).

    Google Scholar 

  39. Lamaze, C., Fujimoto, L. M., Yin, H. L. & Schmid, S. L. The actin cytoskeleton is required for receptor-mediated endocytosis in mammalian cells. J. Biol. Chem. 272, 20332–20335 (1997).

    CAS  PubMed  Google Scholar 

  40. Sarkar, P., Kumar, G. A., Shrivastava, S. & Chattopadhyay, A. Chronic cholesterol depletion increases F-actin levels and induces cytoskeletal reorganization via a dual mechanism. J. Lipid Res. 63, 100206 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Lu, Q. et al. Tyro-3 family receptors are essential regulators of mammalian spermatogenesis. Nature 398, 723–728 (1999).

    CAS  PubMed  Google Scholar 

  42. Mattiuz, R. et al. Type 1 conventional dendritic cells and interferons are required for spontaneous CD4+ and CD8+ T-cell protective responses to breast cancer. Clin. Transl. Immunology 10, e1305 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Marjanovic, N. D. et al. Emergence of a high-plasticity cell state during lung cancer evolution. Cancer Cell 38, 229–246 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Byers, L. A. et al. An epithelial–mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin. Cancer Res. 19, 279–290 (2013).

    CAS  PubMed  Google Scholar 

  45. Cui, Z.-L. et al. YES-associated protein 1 promotes adenocarcinoma growth and metastasis through activation of the receptor tyrosine kinase Axl. Int. J. Immunopathol. Pharmacol. 25, 989–1001 (2012).

    CAS  PubMed  Google Scholar 

  46. Schmid, E. T. et al. AXL receptor tyrosine kinase is required for T cell priming and antiviral immunity. eLife 5, e12414 (2016).

    PubMed  PubMed Central  Google Scholar 

  47. Loschko, J. et al. Absence of MHC class II on cDCs results in microbial-dependent intestinal inflammation. J. Exp. Med. 213, 517–534 (2016).

    PubMed  PubMed Central  Google Scholar 

  48. Balan, S. et al. Large-scale human dendritic cell differentiation revealing Notch-dependent lineage bifurcation and heterogeneity. Cell Rep. 24, 1902–1915 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Rothlin, C. V., Ghosh, S., Zuniga, E. I., Oldstone, M. B. A. & Lemke, G. TAM receptors are pleiotropic inhibitors of the innate immune response. Cell 131, 1124–1136 (2007).

    CAS  PubMed  Google Scholar 

  50. Engelsen, A. S. T. et al. Dissecting the role of AXL in cancer immune escape and resistance to immune checkpoint inhibition. Front. Immunol. 13, 869676 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Goyette, M.-A. et al. Targeting Axl favors an antitumorigenic microenvironment that enhances immunotherapy responses by decreasing Hif-1α levels. Proc. Natl Acad. Sci. USA 118, e2023868118 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Holland, S. J. et al. R428, a selective small molecule inhibitor of Axl kinase, blocks tumor spread and prolongs survival in models of metastatic breast cancer. Cancer Res. 70, 1544–1554 (2010).

    CAS  PubMed  Google Scholar 

  53. Myers, K. V., Amend, S. R. & Pienta, K. J. Targeting Tyro3, Axl and MerTK (TAM receptors): implications for macrophages in the tumor microenvironment. Mol. Cancer 18, 94 (2019).

    PubMed  PubMed Central  Google Scholar 

  54. Tirado-Gonzalez, I. et al. AXL inhibition in macrophages stimulates host-versus-leukemia immunity and eradicates naive and treatment-resistant leukemia. Cancer Discov. 11, 2924–2943 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Lu, F. et al. Identification of NPC1 as the target of U18666A, an inhibitor of lysosomal cholesterol export and Ebola infection. eLife 4, e12177 (2015).

    PubMed  PubMed Central  Google Scholar 

  56. Belabed, M. et al. Kinesin-1 regulates antigen cross-presentation through the scission of tubulations from early endosomes in dendritic cells. Nat. Commun. 11, 1817 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Brinkman, E. K., Chen, T., Amendola, M. & van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42, e168 (2014).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We sincerely thank members of the Merad laboratory for helpful discussions; P. Suri and R. Samstein in the Department of Immunology and Immunotherapy for providing the 4T1 cells; S.D. Kumar and the Advanced Microscopy and Bioimaging Core; and the Mount Sinai Flow Cytometry Core and Human Immune Monitoring Center for technical support. Finally, we thank BerGenBio for generously providing the AXL inhibitor for our in vitro and in vivo studies.

Author information

Authors and Affiliations

Authors

Contributions

M.M. conceived the project. M.B., M.D.P. and M.M. designed the experiments. M.B., M.D.P. and M.M. wrote the manuscript. M.B. performed experiments with support from M.D.P., J.L.B., R.M. and C.M.W. K.J.R., S.B. and A.P. performed experiments with human cDCs, including RNA sequencing. M.D.P. performed computational analyses. C.M.B. generated the D4-GFP cholesterol probe. S.T.C. and N.M.L. contributed to the conceptual development of the project. D.J.P., S.G., C.V.R., C.M.B. and C.L. advised on study design. N.M.L. was supported by the Cancer Research Institute and Bristol Myers Squibb Irvington Postdoctoral Research Fellowship to Promote Racial Diversity (award CRI3931). R.M. was supported by the 2021 AACR–AstraZeneca Immuno-oncology Research Fellowship (grant 21-40-12-MATT). K.J.R. was supported by a Fulbright Future Scholarship and the Mater Foundation. C.M.B. and C.L. were supported by institutional grants from the Curie Institute, INSERM and the Centre National de la Recherche Scientifique and by grants from the Agence Nationale de la Recherche (ANR NanoGammaR-17-CE15-0032) to C.L. and from the Ligue Nationale contre le Cancer and ARC to C.M.B. C.M.B. and C.L. are members of Labex CelTisPhyBio (ANR-10-LBX-0038) and are part of the IDEX PSL (ANR-10-IDEX-0001-02 18796).

Corresponding author

Correspondence to Miriam Merad.

Ethics declarations

Competing interests

M.M. serves on the scientific advisory board of and holds stock from Compugen, Dynavax, Innate Pharma, Morphic Therapeutics, Asher Bio, Dren Bio, Nirogy, Genenta, OncoResponse and Owkin. M.M. serves on the ad hoc scientific advisory board of DBV and Genentech and on the foundation advisory board of Breakthrough Cancer. M.M. receives funding for contracted research from Genentech, Regeneron and Boehringer Ingelheim. M.M. is listed as an inventor on a patent application (16/092576) submitted by the Icahn School of Medicine at Mount Sinai that covers the use of multiplex immunohistochemistry to characterize tumors and treatment responses. The technology is filed through the Icahn School of Medicine at Mount Sinai and is currently unlicensed. This technology was used to evaluate tissue in this study, and the results could impact the value of this technology. T.U.M. has served on advisory and/or data safety monitoring boards for Rockefeller University, Regeneron Pharmaceuticals, AbbVie, Bristol Meyers Squibb, Boehringer Ingelheim, Atara, AstraZeneca, Genentech, Celldex, Chimeric, Glenmark, Simcere, Surface, G1 Therapeutics, NGM Bio, DBV Technologies, Arcus and Astellas and has research grants from Regeneron, Bristol Myers Squibb, Merck and Boehringer Ingelheim. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Jamie D. K. Wilson, in collaboration with the Nature Immunology team. 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 Disrupting cholesterol mobilization impairs cDC maturation.

Flow cytometry-based measurement of cDC maturation markers (that is, IFN-ɣR, CD40, CD80, CD86, PD-L1, PD-L2 or MHC-I, MHC-II) on splenic mature cDCs either treated with (a) the LXRα/β agonist GW3965, (b) supplemental mevalonate, (c) simvastatin, (d) the NPC1 inhibitor U18666A, and (e) wild-type (WT) and Npc1 haploinsufficient (Npc1+/-) mature cDCs. Across all panels, mean values of technical replicates ± SEM of a representative experiment are shown. P-values computed by unpaired, two-tailed t-test.

Extended Data Fig. 2 Quantification of cell surface cholesterol.

Flow cytometric quantification of (a) D4-GFP and (b) CTxB staining of wild-type (WT) and Npc1+/- mature cDCs. (c) Immunofluorescence staining for free cholesterol on the cell surface of 1) cDCs, 2) cDCs supplemented with mevalonate, 3) mature cDCs, 4) mature cDCs treated with the NPC1 inhibitor U18666A, 5) mature cDCs treated with simvastatin, and 6) mature cDCs treated with the LXR agonist GW3965, 7) mature cDCs treated with methyl-β-cyclodextrin, 8) mature cDCs treated with sphingomyelinase. (d) Fluorescence quantification of D4-GFP staining on cDCs and cDCs supplemented with mevalonate, scale 5μM. (e) Fluorescence quantification of D4-GFP staining on groups 3-8 in (c). Across all panels, data represent mean ± SEM of at least three independent replicates or biologically distinct experiments (c, d, e). P-values computed by unpaired, two-tailed t-test.

Extended Data Fig. 3 The phagocytic cell sensor AXL controls cDC maturation.

Flow cytometric measurement (a) of free cholesterol using the D4-GFP probe on WT and KO mature cDCs or (b) of free cholesterol acquired extracellularly by mature cDCs exposed to D4-GFP-stained apoptotic cell debris. (c) Immuno-fluorescence staining and quantification for free cholesterol on the surface of CD11c+ mature cDCs that were fed D4-GFP-stained cell debris, scale 5 μM. (d)-(h) Flow cytometric measurement of cDC maturation markers (that is, CCR7, IFN-ɣR, MHC-I, MHC-II, CD40, CD80, CD86, PD-L1, PD-L2). Across all panels, mean values of technical replicates ± SEM of a representative experiment are shown. P-values computed by ordinary one-way ANOVA t-test.

Extended Data Fig. 4 De novo cholesterol synthesis and transport via NPC1 are regulated by AXL during cDC maturation.

Flow cytometric measurement of cDC maturation markers (that is, CCR7, IFN-ɣR, CD40, CD80, MHC-I, MHC-II, PD-L1, and PD-L2) on WT and AXL KO mature cDCs either left untreated or treated with (a) the NPC1 inhibitor U18666A or (b) simvastatin. Across all panels, mean values of technical replicates ± SEM of a representative experiment are shown. P-values computed by either ordinary one-way ANOVA t-test.

Extended Data Fig. 5 NPC1 haploinsufficiency impairs cDC maturation and T cell activation.

(a) Proliferation and activation of OT-I and OT-II cells following co-culture with wild-type (WT) and Npc1 haploinsufficient (Npc1+/-) mature cDCs. (b)-(e) Flow cytometric measurement of cDC maturation markers on mature cDCs isolated from tumor-bearing lungs of WT and Npc1 haploinsufficient (Npc1+/-) mice. Across all panels, mean values of technical replicates ± SEM of a representative experiment are shown. P-values computed by unpaired, two-tailed t-test.

Extended Data Fig. 6 The therapeutic efficacy of the cDC checkpoint AXL is a T cell-dependent response.

(a) mRNA expression of AXL by KrasG12D/+Trp53-/- tumor cells, according to single-cell RNA-sequencing of tumor cells from the KP GEMM model of lung adenocarcinoma (Marjanovic et al., 2020). (b) Ex vivo flow cytometric staining of CTxB on Axl+/+ (WT) and Axl-/- (KO) mature cDCs from tumor-bearing lungs. (c) Flow cytometric measurement of intracellular IL-12p40 produced by mature cDCs from the tumor-bearing lungs of (left) WT and BCT-treated mice or (right) WT, KO and Zbtb46Cre-Axlfl/fl (AxlΔDC) mice. (d) Frequency of NK cells in the (left) lungs and (right) tumor-draining lymph nodes of WT, KO, and AxlΔDC mice. (e) (Left) histology and (right) quantification of tumor burden in the lungs of WT, KO, and KO mice depleted of CD8 T cells at 21 days post-tumor cell inoculation. Same scale bar as panel (f). Scale bar, 1 mm. (f) (Left) histology and (right) quantification of tumor burden in the lungs of WT, KO, and KO mice depleted of CD4 T cells at 21 days post-tumor cell inoculation. Same scale bar as panel (e). Scale bar, 1 mm. (g) Frequency of total CD8 T cells in the (left) blood and (right) lungs of WT, KO, and KO mice depleted of CD8 T cells. (h) Frequency of total CD4 T cells in the (left) blood and (right) lungs of WT, KO, and KO mice depleted of CD4 T cells. Across all panels, data represent mean ± SEM of at least three biologically distinct experiments. All p-values computed by ordinary one-way ANOVA t-test.

Supplementary information

Reporting Summary

Peer Review File

Supplementary Tables 1–3

Supplementary Table 1. Submodules defined by the identification of highly correlated variable genes across DCs integrated from different scRNA-seq datasets. Supplementary Table 2. Metadata per cell barcode of integrated scRNA-seq analysis, including cell subtype annotation and dataset of origin. Supplementary Table 3. Active metabolic pathways inferred from gene set enrichment analysis of genes enriched in mature cDCs.

Supplementary Video 1

Z-stack recording of pSTAT1 and DAPI staining of WT BMDCs fed with KP-GFP cells.

Supplementary Video 2

Z-stack recording of pSTAT1 and DAPI staining of Axl-KO BMDCs fed with KP-GFP cells.

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

Belabed, M., Park, M.D., Blouin, C.M. et al. Cholesterol mobilization regulates dendritic cell maturation and the immunogenic response to cancer. Nat Immunol 26, 188–199 (2025). https://doi.org/10.1038/s41590-024-02065-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-024-02065-8

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