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:

The receptor DNGR-1 signals for phagosomal rupture to promote cross-presentation of dead-cell-associated antigens

A Publisher Correction to this article was published on 02 February 2021

This article has been updated

Abstract

Type 1 conventional dendritic (cDC1) cells are necessary for cross-presentation of many viral and tumor antigens to CD8+ T cells. cDC1 cells can be identified in mice and humans by high expression of DNGR-1 (also known as CLEC9A), a receptor that binds dead-cell debris and facilitates XP of corpse-associated antigens. Here, we show that DNGR-1 is a dedicated XP receptor that signals upon ligand engagement to promote phagosomal rupture. This allows escape of phagosomal contents into the cytosol, where they access the endogenous major histocompatibility complex class I antigen processing pathway. The activity of DNGR-1 maps to its signaling domain, which activates SYK and NADPH oxidase to cause phagosomal damage even when spliced into a heterologous receptor and expressed in heterologous cells. Our data reveal the existence of innate immune receptors that couple ligand binding to endocytic vesicle damage to permit MHC class I antigen presentation of exogenous antigens and to regulate adaptive 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: DNGR-1 has a dedicated role in XP of ligand-associated antigens.
Fig. 2: DNGR-1 marks an early MHC I+ nondegradative phagosome population.
Fig. 3: DNGR-1 signaling causes phagosomal damage resulting in rupture.
Fig. 4: Phagosomal rupture results in P2C transfer and XP.
Fig. 5: SYK signaling promotes phagosomal rupture and XP.
Fig. 6: Phagosomal ROS promotes damage and rupture of phagosomes.
Fig. 7: NOX2 in cDC1 is necessary for cross-presentation of dead-cell-associated antigens.

Similar content being viewed by others

Data availability

Source data are provided with this paper. All other data supporting the findings can be requested from the corresponding author.

Change history

  • 02 February 2021

    A Correction to this paper has been published: https://doi.org/10.1038/s41590-021-00885-6.

References

  1. Huang, A. Y. et al. Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. Science 264, 961–965 (1994).

    CAS  PubMed  Google Scholar 

  2. Sigal, L. J., Crotty, S., Andino, R. & Rock, K. L. Cytotoxic T-cell immunity to virus-infected non-haematopoietic cells requires presentation of exogenous antigen. Nature 398, 77–80 (1999).

    CAS  PubMed  Google Scholar 

  3. Iborra, S. et al. The DC receptor DNGR-1 mediates cross-priming of CTLs during vaccinia virus infection in mice. J. Clin. Invest. 122, 1628–1643 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Smed-Sörensen, A. et al. Influenza A virus infection of human primary dendritic cells impairs their ability to cross-present antigen to CD8 T cells. PLoS Pathog. 8, e1002572 (2012).

    PubMed  PubMed Central  Google Scholar 

  5. Alloatti, A. et al. Critical role for Sec22b-dependent antigen cross-presentation in antitumor immunity. J. Exp. Med. 214, 2231–2241 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Theisen, D. J. et al. WDFY4 is required for cross-presentation in response to viral and tumor antigens. Science 362, 694–699 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Cruz, F. M., Colbert, J. D., Merino, E., Kriegsman, B. A. & Rock, K. L. The biology and underlying mechanisms of cross-presentation of exogenous antigens on MHC-I molecules. Annu Rev. Immunol. 35, 149–176 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gros, M. & Amigorena, S. Regulation of antigen export to the cytosol during cross-presentation. Front Immunol. 10, 1283–1289 (2019).

    Google Scholar 

  9. Colbert, J. D., Cruz, F. M. & Rock, K. L. Cross-presentation of exogenous antigens on MHC I molecules. Curr. Opin. Immunol. 64, 1–8 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Grotzke, J. E. & Cresswell, P. Are ERAD components involved in cross-presentation? Mol. Immunol. 68, 112–115 (2015).

    CAS  PubMed  Google Scholar 

  11. Dingjan, I. et al. Lipid peroxidation causes endosomal antigen release for cross-presentation. Sci. Rep. 6, 22064 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Dingjan, I. et al. VAMP8-mediated NOX2 recruitment to endosomes is necessary for antigen release. Eur. J. Cell Biol. 96, 705–714 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Reis e Sousa, C. & Germain, R. N. Major histocompatibility complex class I presentation of peptides derived from soluble exogenous antigen by a subset of cells engaged in phagocytosis. J. Exp. Med. 182, 841–851 (1995).

    CAS  PubMed  Google Scholar 

  14. Germain, R. N. et al. Processing and presentation of endocytically acquired protein antigens by MHC class II and class I molecules. Immunol. Rev. 151, 5–30 (1996).

    CAS  Google Scholar 

  15. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8α+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Savina, A. et al. The small GTPase Rac2 controls phagosomal alkalinization and antigen crosspresentation selectively in CD8+ dendritic cells. Immunity 30, 544–555 (2009).

    CAS  PubMed  Google Scholar 

  17. Weimershaus, M. et al. Conventional dendritic cells require IRAP-Rab14 endosomes for efficient cross-presentation. J. Immunol. 188, 1840–1846 (2012).

    CAS  PubMed  Google Scholar 

  18. Kretzer, N. M. et al. RAB43 facilitates cross-presentation of cell-associated antigens by CD8α+ dendritic cells. J. Exp. Med. 213, 2871–2883 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Han, D. et al. Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells. Nature 566, 270–274 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Ou, P. et al. Thioesterase PPT1 balances viral resistance and efficient T cell crosspriming in dendritic cells. J. Exp. Med. 216, 2091–2112 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Hanč, P. et al. In C-Type Lectin Receptors in Immunity (Ed. Yamasaki, S.) 65–81 (Springer, 2016).

  22. Schulz, O. et al. Myosin II synergizes with F-actin to promote DNGR-1-dependent cross-presentation of dead cell-associated antigens. Cell Rep. 24, 419–428 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Sancho, D. et al. Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature 458, 899–903 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Zelenay, S. et al. The dendritic cell receptor DNGR-1 controls endocytic handling of necrotic cell antigens to favor cross-priming of CTLs in virus-infected mice. J. Clin. Invest. 122, 1615–1627 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Iborra, S. et al. Optimal generation of tissue-resident but not circulating memory T cells during viral infection requires crosspriming by DNGR-1+ dendritic cells. Immunity 45, 847–860 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Hanč, P. et al. A pH- and ionic strength-dependent conformational change in the neck region regulates DNGR-1 function in dendritic cells. EMBO J. 35, 2484–2497 (2016).

    PubMed  PubMed Central  Google Scholar 

  27. Grotzke, J. E. et al. Sec61 blockade by mycolactone inhibits antigen cross-presentation independently of endosome-to-cytosol export. Proc. Natl Acad. Sci. USA 114, E5910–E5919 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Kozik, P. et al. Small molecule enhancers of endosome-to-cytosol import augment anti-tumor immunity. Cell Rep. 32, 107905 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Savina, A., Vargas, P., Guermonprez, P., Lennon, A.-M. & Amigorena, S. Measuring pH, ROS production, maturation, and degradation in dendritic cell phagosomes using cytofluorometry-based assays. Methods Mol. Biol. 595, 383–402 (2010).

    CAS  PubMed  Google Scholar 

  30. Böttcher, J. P. et al. Oncogenic transformation of dendritic cells and their precursors leads to rapid cancer development in mice. J. Immunol. 195, 5066–5076 (2015).

    PubMed  PubMed Central  Google Scholar 

  31. Thurston, T. L. M., Wandel, M. P., von Muhlinen, N., Foeglein, Á. & Randow, F. Galectin 8 targets damaged vesicles for autophagy to defend cells against bacterial invasion. Nature 482, 414–418 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Ellison, C. J., Kukulski, W., Boyle, K. B., Munro, S. & Randow, F. Transbilayer movement of sphingomyelin precedes catastrophic breakage of enterobacteria-containing vacuoles. Curr. Biol. 30, 2974–2983.e6 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Cebrian, I. et al. Sec22b regulates phagosomal maturation and antigen crosspresentation by dendritic cells. Cell 147, 1355–1368 (2011).

    CAS  PubMed  Google Scholar 

  34. Lin, M. L. et al. Selective suicide of cross-presenting CD8+ dendritic cells by cytochrome c injection shows functional heterogeneity within this subset. Proc. Natl Acad. Sci. USA 105, 3029–3034 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Dersh, D., Yewdell, J. W. & Wei, J. In Antigen Processing Methods in Molecular Biology Vol. 1988 (Ed. van Endert, P.) 109–122 (Humana, 2019).

  36. Zhang, J.-G. et al. The dendritic cell receptor Clec9A binds damaged cells via exposed actin filaments. Immunity 36, 646–657 (2012).

    CAS  PubMed  Google Scholar 

  37. Lee, S. B., Bae, I. H., Bae, Y. S. & Um, H.-D. Link between mitochondria and NADPH oxidase 1 isozyme for the sustained production of reactive oxygen species and cell death. J. Biol. Chem. 281, 36228–36235 (2006).

    CAS  PubMed  Google Scholar 

  38. Palmowski, M. J. et al. Role of immunoproteasomes in cross-presentation. J. Immunol. 177, 983–990 (2006).

    CAS  PubMed  Google Scholar 

  39. Ackerman, A. L., Giodini, A. & Cresswell, P. A role for the endoplasmic reticulum protein retrotranslocation machinery during crosspresentation by dendritic cells. Immunity 25, 607–617 (2006).

    CAS  PubMed  Google Scholar 

  40. Zehner, M. et al. Mannose receptor polyubiquitination regulates endosomal recruitment of p97 and cytosolic antigen translocation for cross-presentation. Proc. Natl Acad. Sci. USA 108, 9933–9938 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Ackerman, A. L., Kyritsis, C., Tampé, R. & Cresswell, P. Early phagosomes in dendritic cells form a cellular compartment sufficient for cross presentation of exogenous antigens. Proc. Natl Acad. Sci. USA 100, 12889–12894 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Guermonprez, P. et al. ER–phagosome fusion defines an MHC class I cross-presentation compartment in dendritic cells. Nature 425, 397–402 (2003).

    CAS  PubMed  Google Scholar 

  43. Houde, M. et al. Phagosomes are competent organelles for antigen cross-presentation. Nature 425, 402–406 (2003).

    CAS  PubMed  Google Scholar 

  44. Accapezzato, D. et al. Chloroquine enhances human CD8+ T cell responses against soluble antigens in vivo. J. Exp. Med. 202, 817–828 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Belizaire, R. & Unanue, E. R. Targeting proteins to distinct subcellular compartments reveals unique requirements for MHC class I and II presentation. Proc. Natl Acad. Sci. USA 106, 17463–17468 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Turk, B. & Turk, V. Lysosomes as ‘suicide bags’ in cell death: myth or reality? J. Biol. Chem. 284, 21783–21787 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Graham, D. B. et al. An ITAM-signaling pathway controls cross-presentation of particulate but not soluble antigens in dendritic cells. J. Exp. Med. 204, 2889–2897 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Savina, A. et al. NOX2 controls phagosomal pH to regulate antigen processing during crosspresentation by dendritic cells. Cell 126, 205–218 (2006).

    CAS  PubMed  Google Scholar 

  49. Nair-Gupta, P. et al. TLR signals induce phagosomal MHC-I delivery from the endosomal recycling compartment to allow cross-presentation. Cell 158, 506–521 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Alloatti, A. et al. Toll-like receptor 4 engagement on dendritic cells restrains phago-lysosome fusion and promotes cross-presentation of antigens. Immunity 43, 1087–1100 (2015).

    CAS  PubMed  Google Scholar 

  51. Fuertes Marraco, S. A. et al. Novel murine dendritic cell lines: a powerful auxiliary tool for dendritic cell research. Front. Immunol. 3, 331 (2012).

    PubMed  PubMed Central  Google Scholar 

  52. Karttunen, J., Sanderson, S. & Shastri, N. Detection of rare antigen-presenting cells by the lacZ T-cell activation assay suggests an expression cloning strategy for T-cell antigens. Proc. Natl Acad. Sci. USA 89, 6020–6024 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Dhomen, N. et al. Oncogenic Braf induces melanocyte senescence and melanoma in mice. Cancer Cell 15, 294–303 (2009).

    CAS  PubMed  Google Scholar 

  54. Boes, M. et al. T cells induce extended class II MHC compartments in dendritic cells in a Toll-like receptor-dependent manner. J. Immunol. 171, 4081–4088 (2003).

    CAS  PubMed  Google Scholar 

  55. Schraml, B. U. et al. Genetic tracing via DNGR-1 expression history defines dendritic cells as a hematopoietic lineage. Cell 154, 843–858 (2013).

    CAS  PubMed  Google Scholar 

  56. Deerinck, T., Bushong, E. A., Lev-Ram, V. & Shu, X. Enhancing serial block-face scanning electron microscopy to enable high resolution 3D nanohistology of cells and tissues. Microsc. Microanal. 16, 1138–1139 (2010).

    CAS  Google Scholar 

  57. Cardona, A. et al. TrakEM2 software for neural circuit reconstruction. PLoS ONE 7, e38011 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Bogovich, J. A., Hanslovsky, P., Wong, A. & Saalfeld, S. Robust registration of calcium images by learned contrast synthesis. IEEE International Symposium on Biomedical Imaging https://doi.org/10.1109/ISBI.2016.7493463 (2016).

  59. Belevich, I., Joensuu, M., Kumar, D., Vihinen, H. & Jokitalo, E. Microscopy image browser: a platform for segmentation and analysis of multidimensional datasets. PLoS Biol. 14, e1002340 (2016).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Immunobiology Laboratory for helpful discussions and suggestions. We thank the Crick BRF and Flow Cytometry STPs for their support throughout this project. We are grateful to P. Faull and B. Snijders for helpful comments. This work was supported by the Francis Crick Institute, which receives core funding from Cancer Research UK (FC001136), the UK Medical Research Council (FC001136) and the Wellcome Trust (FC001136), by an ERC Advanced Investigator grant (AdG 268670), by a Wellcome Investigator Award (WT106973MA) and by a prize from the Louis-Jeantet Foundation. C.M.H. was supported by a FEBS Long-Term Fellowship and a Marie Skłodowska-Curie Individual Fellowship under Horizon 2020 from the European Union. M.D.B. was supported by an EMBO LTF (ALTF 1096-2018) and a Marie Skłodowska-Curie Individual Fellowship under Horizon 2020 from the European Union. F.R. is supported by the MRC (U105170648) and the Wellcome Trust (WT104752MA).

Author information

Authors and Affiliations

Authors

Contributions

J.C., C.M.H. and H.B. performed most of the experiments. O.S. and H.R. helped with the sorting of single bead MuTuDCs. C.M.H. and M.D.B. performed in vivo experiments. O.S. performed initial experiments and provided key reagents. E.C. helped with confocal experiments. N.C.R. helped with mouse breeding and genotyping. H.R. helped with ImageStream analysis. S.Z. generated the DNGR-1::Dectin-1 chimeras. L.C. and M.C.D. performed electron microscopy. A.A. and S.A. provided advice and training for the PhagoFACS experiments. V.P. provided the Cybb−/− mouse strain. D.C.T. provided reagents that contributed to this study. C.J.E. and F.R. provided the lysenin and galectin probes. H.B., J.C., C.M.H. and C.R.S. interpreted the data and wrote the manuscript. C.R.S. designed and supervised the study.

Corresponding author

Correspondence to Caetano Reis e Sousa.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Peer reviewer reports are available. Zoltan Fehervari was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team. 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 Bead uptake, CD4+ T cell stimulation and DNGR-1 mediated XP by the cytosolic pathway.

a, OVA and FM-OVA beads stained with anti-ovalbumin, anti-actin or phalloidin analysed by flow cytometry. b, WT MuTuDCs were pulsed with OVA or FM-OVA beads, fixed, stained with anti-OVA to mark uninternalized beads (‘out. beads’) and imaged by confocal microscopy (scale bar 5 µm). Image representative of one experiment. c, FM-OVA bead internalisation by WT, KO and KO-WT MuTuDCs analysed by flow cytometry. MFI from one of two experiments (n = 2). de, WT, KO and KO-WT MuTuDCs pulsed with OVA or FM-OVA beads for 4 hrs were co-cultured with OT-I cells. IFN-γ was assessed by ELISA. One of three experiments is shown. Plotted as mean (±s.d.) of experimental duplicates. P values determined using two-way ANOVA. f, ImageStream analysis of WT MuTuDCs pulsed with yellow-green FM-OVA or OVA beads for 4 hrs and sorted for single bead+ MuTuDCs. g, Single bead+ WT MuTuDCs were co-cultured with OT-II cells. CD69+CD4+ T cell frequency was determined by flow cytometry. One of three experiments is shown (n = 3). hn, WT MuTuDCs were incubated with FM-OVA beads (blue, 10:1, beads:DCs), UV-irradiated bm1 OVA MEFs (red, 3:1, dead cells:DCs) or hen egg white (grey, 1 mg/ml) in the presence of 250 μM leupeptin, 250 μM pepstatin, 10 μM lactacystin, 20 μM lactacystin, 50 μM chloroquine or 500 μM E64. After 4 hrs, OT-I cells were added overnight. OT-I cells alone were also incubated with SIINFEKL peptide (black, 1 nM) in the presence of the inhibitors. IFN-γ was assessed by ELISA, plotted as mean (±s.d.) of experimental duplicates and representative of two independent experiments (n = 2). P values determined using an unpaired t test. n.s., not significant; *P ≤ 0.05; **P ≤ 0.01; ****P ≤ 0.00001.

Source data

Extended Data Fig. 2 DNGR-1+ non-degradative phagosome in RAW264.7 macrophages ectopically expressing DNGR-1.

ag, WT and KO MuTuDCs (ae), KID (f) or RAW264.7 (g) cells were pulsed with FM-OVA beads for the indicated times. Phagosomes were isolated and stained with anti-DNGR-1, anti-LAMP-2, anti-OVA and anti-MHC I antibodies. Analysis was performed by flow cytometry. Data represented as mean (± s.d.) and from one of two experiments are shown (n = 2). h, Confocal microscopy image of RAW264.7 cells transiently expressing H-2Kb were pulsed with IgG-beads for 2 hrs (scale bar = 6 μm). Each dot represents a single image out of 10 images per condition (n = 10). Data from one representative experiment of two independent experiments (n = 2).

Source data

Extended Data Fig. 3 Recruitment of galectin-8 to DNGR-1+ phagosomes containing FM-OVA beads, phagocytosis by cells expressing chimeric receptors and sulforhodimine B release from phagosomes.

a, PhagoFACS of phagosomes isolated from WT MuTuDCs pulsed with FM-OVA beads for 4 hrs and stained with anti-DNGR-1, anti-LAMP-2 and anti-galectin-8 antibodies. Frequency of phagosomes is plotted as mean (± s.d.) for three independent experiments (n = 3). P values were determined by unpaired t test, Welch’s test. b, WT MuTuDCs expressing mCherry::galectin-3 were incubated with OVA or FM-OVA beads (5:1, beads to DCs) for 4 hrs. Cells were fixed, uninternalised beads (‘out. bead’) marked by anti-OVA staining and cells were imaged by confocal microscopy (scale bar = 5 μm). Images are a representative image from 18 FM-OVA and 36 OVA images. c, HEK293T cells expressing cytosolic GFP and C7, C9::C7 or C9(Y7F)::C7 receptors and stained for Dectin-1. Images show a single representative image from 3 similar images. d, HEK293T cells expressing the indicated receptors were pulsed with biotinylated zymosan for 1 hr before fixation and labelling of uninternalised zymosan with fluorescent streptavidin. Internalised zymosan particles were enumerated from replicate confocal images (n = 3 images for empty vector and C7 tail-less, 15 images for C7, C9::C7 and C9(Y7F)::C7 and 5 images for C7 R406 and C9::C7 R406). P values were determined using an unpaired t test. e, HEK293T cells expressing the indicated receptors were challenged with zymosan pre-soaked in sulforhodamine B (SRB). SRB in the cytosol was quantified and normalized to phagosomal fluorescence. Data and images are representative of three independent repeats (n = 3) and n = 40 cells for C9::C7 and 28 cells for C9(Y7F)::C7. Data (d,e) represented as mean (± s.e.m.). P values were determined using an unpaired t test, Welch’s test. n.s., not significant; **P ≤ 0.01; ***P ≤ 0.001.

Source data

Extended Data Fig. 4 Generation of HEK293T cell lines able to present OVA on H-2Kb.

a, Confocal microscope images of HEK293T cells stably expressing cytosolic GFP, H-2Kb and either C7, C9::C7 or C9(Y7F)::C7 receptors (anti-Dectin-1 staining). Nuclei were stained with DAPI. Images are representative of 3 similar images (scale bars 5 μm). b, As for (a) but using flow cytometry c, IL-2 from B3Z hybridoma cells cultured with the above H-2Kb-expressing HEK293T cells in the presence of the indicated concentrations of SIINFEKL peptide. Data are plotted as mean (± s.d.) of experimental triplicates from one of three independent experiments (n = 3). P values were determined by two-way ANOVA. d, Confocal images of HEK293T cells stably transfected with empty vector or plasmid encoding either C7, C9::C7 or C9(Y7F)::C7 receptors and pulsed with biotinylated zymosan for 1 hr before fixation and labelling of uninternalised zymosan with fluorescent streptavidin (scale bars 10 μm).. The number of internalised zymosan particles (right) was enumerated from the images (left). Data represented as mean (± s.e.m.) and each dot represents an independent experiment. e, RAW264.7 cells expressing H-2Kb and either C9::C7 or C9(Y7F)::C7 receptors were pulsed with zymosan-OVA for 4 hrs before fixation with 0.5 % PFA and incubated with OT-I cells. IFN-γ was assessed by ELISA. f, LLOMe (1 mM) was added during zymosan-OVA pulse of RAW264.7 cells expressing H-2Kb and C9(Y7F)::C7 receptor. All data in (e,f) are plotted as mean (± s.e.m.) of experimental triplicates. n.s., not significant; **P ≤ 0.01; ****P ≤ 0.00001.

Source data

Extended Data Fig. 5 SYK is required for GFP influx into phagosomes.

a, Western blot and confocal microscope images of SYK protein expression in HEK293T SYK KO cells, complemented with mouse mCherry::SYK or mCherry::SYK K396R kinase dead (KD). Bottom panel: mCherry expression of the same micrographs depicted as in Fig. 5c lower panel (scale bar 2 μm). b, Flow cytometry plots measuring phagocytosis of HEK293T WT and SYK KO cells incubated with zymosan-pHrodo-Red particles over 30 min timecourse. Data are representative of two independent experiments (n = 2).

Source data

Extended Data Fig. 6 DNGR signalling promotes phagosomal ROS production.

ab, Confocal images of RAW264.7 cells transfected with empty vector or plasmid encoding C9::C7 or C9(Y7F)::C7 receptors and pulsed with zymosan (a) or dead sRBCs (b) in the presence of Nitroblue tetrazolium (NBT) (Scale bar 10 μm). Quantification of ROS+ phagosomes. Data represented as mean (± s.e.m.) (a) or (± s.d.) (b) and are representative of two independent determinations (n = 2). P values determined by one-way ANOVA. c, RAW264.7 stably expressing C9 or C9(Y7F) receptors were pulsed with CellTracker DeepRed (CTDR)-labelled FP-sRBCs for 2 hrs. Percentage of CTDR+ RAW264.7 cells was quantified by flow cytometry. Data represented as mean (± s.d.) and are representative of two independent experiments (n = 2). d, Confocal images of RAW264.7 stably expressing C9 or C9(Y7F) receptors pulsed with dead cells in the presence of NBT for 2 hrs (scale bars 10 μm). Image is a representative image of three similar images. e, RT-PCR of NADPH oxidase subunits in HEK293T. Representative of two experiments (n = 2). f, HEK293T cells stably expressing C9::C7 were challenged with zymosan-Oxyburst in the presence or absence of DPI for 1 hr. Oxyburst+ positive phagosomes were quantified across 5 fields of view (n > 100 phagosomes). Data represented as mean (± s.e.m.). P values were calculated by unpaired parametric test, Mann-Whitney and are representative of two independent experiments (n = 2). g, HEK293T C9::C7 cells were pulsed with zymosan-Ova (left) or transfected with plasmid encoding VENUS-SIINFEKL (right) in the presence or absence of DPI (10 μM) for 4 hrs before fixing and adding of OT-I Rag1-/- T-cells. IFN-γ was assessed by ELISA, plotted as mean (± s.d.) of an experimental triplicate. n.s., not significant; *P ≤ 0.05; **P ≤ 0.01.

Source data

Extended Data Fig. 7 Generation of DNGR-1 and NOX2-deficient cDC1 chimeras.

a, Flow cytometry gating strategy to define and quantify chimeric T cell populations in blood. b, Quantification of percentage chimerism in the blood 8 weeks post adoptive transfer across the indicated genotypes. Each dot is an individual mouse sample (Batf3-/-: Batf3-/- n = 7, Batf3-/-: WT n = 8, Batf3-/-: Clec9acre/cre n = 8, Batf3-/-: Cybb-/- n = 9).

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–5.

Reporting Summary

Peer Review Information

Supplementary Video 1

SBF SEM 3D reconstruction of damaged phagosome. HEK293T cells expressing mCherry::lysenin and the C9::C7 receptor were incubated with zymosan for 4 h. The 3D reconstruction of a lysenin+ phagosome was carried out as described in the Methods, and the video was prepared using the IMARIS software.

Source data

Source Data Fig. 1

Numerical data used for plots.

Source Data Fig. 2

Numerical data used for plots.

Source Data Fig. 3

Numerical data used for plots.

Source Data Fig. 4

Numerical data used for plots.

Source Data Fig. 5

Unprocessed immunoblots.

Source Data Fig. 5

Numerical data used for plots.

Source Data Fig. 6

Unprocessed immunoblots.

Source Data Fig. 6

Numerical data used for plots.

Source Data Fig. 7

Numerical data used for plots.

Source Data Extended Data Fig. 1

Numerical data used for plots.

Source Data Extended Data Fig. 2

Numerical data used for plots.

Source Data Extended Data Fig. 3

Numerical data used for plots.

Source Data Extended Data Fig. 4

Numerical data used for plots.

Source Data Extended Data Fig. 5

Unprocessed immunoblots.

Source Data Extended Data Fig. 6

Numerical data used for plots.

Source Data Extended Data Fig. 6

Unprocessed gels.

Source Data Extended Data Fig. 7

Numerical data used for plots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Canton, J., Blees, H., Henry, C.M. et al. The receptor DNGR-1 signals for phagosomal rupture to promote cross-presentation of dead-cell-associated antigens. Nat Immunol 22, 140–153 (2021). https://doi.org/10.1038/s41590-020-00824-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-00824-x

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