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.

cDC1 prime and are licensed by CD4+ T cells to induce anti-tumour immunity

Abstract

Conventional type 1 dendritic cells (cDC1)1 are thought to perform antigen cross-presentation, which is required to prime CD8+ T cells2,3, whereas cDC2 are specialized for priming CD4+ T cells4,5. CD4+ T cells are also considered to help CD8+ T cell responses through a variety of mechanisms6,7,8,9,10,11, including a process whereby CD4+ T cells ‘license’ cDC1 for CD8+ T cell priming12. However, this model has not been directly tested in vivo or in the setting of help-dependent tumour rejection. Here we generated an Xcr1Cre mouse strain to evaluate the cellular interactions that mediate tumour rejection in a model requiring CD4+ and CD8+ T cells. As expected, tumour rejection required cDC1 and CD8+ T cell priming required the expression of major histocompatibility class I molecules by cDC1. Unexpectedly, early priming of CD4+ T cells against tumour-derived antigens also required cDC1, and this was not simply because they transport antigens to lymph nodes for processing by cDC2, as selective deletion of major histocompatibility class II molecules in cDC1 also prevented early CD4+ T cell priming. Furthermore, deletion of either major histocompatibility class II or CD40 in cDC1 impaired tumour rejection, consistent with a role for cognate CD4+ T cell interactions and CD40 signalling in cDC1 licensing. Finally, CD40 signalling in cDC1 was critical not only for CD8+ T cell priming, but also for initial CD4+ T cell activation. Thus, in the setting of tumour-derived antigens, cDC1 function as an autonomous platform capable of antigen processing and priming for both CD4+ and CD8+ T cells and of the direct orchestration of their cross-talk that is required for optimal anti-tumour immunity.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: CD4+ T cells are required during primary and secondary tumour responses.
Fig. 2: cDC1 are superior at presenting cell-associated material.
Fig. 3: cDC1 expression of major histocompatibility class II is required for tumour rejection.
Fig. 4: cDC1 expression of CD40 is required for tumour rejection.

Data availability

The microarray data generated during the course of this study have been deposited and are available at the Gene Expression Omnibus (GEO) database. The microarrays used in Fig. 4b and Extended Data Fig. 7b can be accessed with the accession number GSE152196. All other primary data and materials that support the findings of this study are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Guilliams, M. et al. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat. Rev. Immunol. 14, 571–578 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  3. Cancel, J. C., Crozat, K., Dalod, M. & Mattiuz, R. Are conventional type 1 dendritic cells critical for protective antitumor immunity and how? Front. Immunol. 10, 9 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Dudziak, D. et al. Differential antigen processing by dendritic cell subsets in vivo. Science 315, 107–111 (2007).

    Article  PubMed  ADS  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Bennett, S. R., Carbone, F. R., Karamalis, F., Miller, J. F. & Heath, W. R. Induction of a CD8+ cytotoxic T lymphocyte response by cross-priming requires cognate CD4+ T cell help. J. Exp. Med. 186, 65–70 (1997).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Bennett, S. R. et al. Help for cytotoxic-T-cell responses is mediated by CD40 signalling. Nature 393, 478–480 (1998).

    Article  PubMed  ADS  CAS  Google Scholar 

  8. Schoenberger, S. P., Toes, R. E., van der Voort, E. I., Offringa, R. & Melief, C. J. T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature 393, 480–483 (1998).

    Article  PubMed  ADS  CAS  Google Scholar 

  9. Bourgeois, C., Rocha, B. & Tanchot, C. A role for CD40 expression on CD8+ T cells in the generation of CD8+ T cell memory. Science 297, 2060–2063 (2002).

    Article  PubMed  ADS  CAS  Google Scholar 

  10. Janssen, E. M. et al. CD4+ T-cell help controls CD8+ T-cell memory via TRAIL-mediated activation-induced cell death. Nature 434, 88–93 (2005).

    Article  PubMed  ADS  CAS  Google Scholar 

  11. Williams, M. A., Tyznik, A. J. & Bevan, M. J. Interleukin-2 signals during priming are required for secondary expansion of CD8+ memory T cells. Nature 441, 890–893 (2006).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  12. Borst, J., Ahrends, T., Bąbała, N., Melief, C. J. M. & Kastenmüller, W. CD4+ T cell help in cancer immunology and immunotherapy. Nat. Rev. Immunol. 18, 635–647 (2018).

    Article  PubMed  CAS  Google Scholar 

  13. Ridge, J. P., Di Rosa, F. & Matzinger, P. A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell. Nature 393, 474–478 (1998).

    Article  PubMed  ADS  CAS  Google Scholar 

  14. Smith, C. M. et al. Cognate CD4+ T cell licensing of dendritic cells in CD8+ T cell immunity. Nat. Immunol. 5, 1143–1148 (2004).

    Article  PubMed  CAS  Google Scholar 

  15. Eickhoff, S. et al. Robust anti-viral immunity requires multiple distinct T Cell-dendritic cell interactions. Cell 162, 1322–1337 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Hor, J. L. et al. Spatiotemporally distinct interactions with dendritic cell subsets facilitates CD4+ and CD8+ T cell activation to localized viral infection. Immunity 43, 554–565 (2015).

    Article  PubMed  CAS  Google Scholar 

  17. Lehmann, C. H. K. et al. DC subset-specific induction of T cell responses upon antigen uptake via Fcγ receptors in vivo. J. Exp. Med. 214, 1509–1528 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Bajaña, S., Roach, K., Turner, S., Paul, J. & Kovats, S. IRF4 promotes cutaneous dendritic cell migration to lymph nodes during homeostasis and inflammation. J. Immunol. 189, 3368–3377 (2012).

    Article  PubMed  CAS  Google Scholar 

  19. Schlitzer, A. et al. IRF4 transcription factor-dependent CD11b+ dendritic cells in human and mouse control mucosal IL-17 cytokine responses. Immunity 38, 970–983 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Williams, J. W. et al. Transcription factor IRF4 drives dendritic cells to promote Th2 differentiation. Nat. Commun. 4, 2990 (2013).

    Article  PubMed  ADS  CAS  Google Scholar 

  21. Valdez, Y. et al. Major histocompatibility complex class II presentation of cell-associated antigen is mediated by CD8α+ dendritic cells in vivo. J. Exp. Med. 195, 683–694 (2002).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  23. Sun, J. C. & Bevan, M. J. Defective CD8 T cell memory following acute infection without CD4 T cell help. Science 300, 339–342 (2003).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  24. Sun, J. C., Williams, M. A. & Bevan, M. J. CD4+ T cells are required for the maintenance, not programming, of memory CD8+ T cells after acute infection. Nat. Immunol. 5, 927–933 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Matsushita, H. et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 482, 400–404 (2012).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  26. Alspach, E. et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature 574, 696–701 (2019).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  27. Bos, R. & Sherman, L. A. CD4+ T-cell help in the tumor milieu is required for recruitment and cytolytic function of CD8+ T lymphocytes. Cancer Res. 70, 8368–8377 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Ferris, S. T. et al. A minor subset of Batf3-dependent antigen-presenting cells in islets of Langerhans is essential for the development of autoimmune diabetes. Immunity 41, 657–669 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Theisen, D. J. et al. Batf3-dependent genes control tumor rejection induced by dendritic cells independently of cross-presentation. Cancer Immunol. Res. 7, 29–39 (2019).

    PubMed  CAS  Google Scholar 

  30. Durai, V. et al. Cryptic activation of an Irf8 enhancer governs cDC1 fate specification. Nat. Immunol. 20, 1161–1173 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4 (2001).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Bern, M. D. et al. Inducible down-regulation of MHC class I results in natural killer cell tolerance. J. Exp. Med. 216, 99–116 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Hashimoto, K., Joshi, S. K. & Koni, P. A. A conditional null allele of the major histocompatibility IA-beta chain gene. Genesis 32, 152–153 (2002).

    Article  PubMed  CAS  Google Scholar 

  34. Archambault, A. S. et al. Cutting edge: conditional MHC class II expression reveals a limited role for B cell antigen presentation in primary and secondary CD4 T cell responses. J. Immunol. 191, 545–550 (2013).

    Article  PubMed  CAS  Google Scholar 

  35. Laidlaw, B. J., Craft, J. E. & Kaech, S. M. The multifaceted role of CD4+ T cells in CD8+ T cell memory. Nat. Rev. Immunol. 16, 102–111 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Ardouin, L. et al. Broad and largely concordant molecular changes characterize tolerogenic and immunogenic dendritic cell maturation in thymus and periphery. Immunity 45, 305–318 (2016).

    Article  PubMed  CAS  Google Scholar 

  37. Pinschewer, D. D. et al. FTY720 immunosuppression impairs effector T cell peripheral homing without affecting induction, expansion, and memory. J. Immunol. 164, 5761–5770 (2000).

    Article  PubMed  CAS  Google Scholar 

  38. Ahrends, T. et al. CD4+ T cell help confers a cytotoxic T cell effector program including coinhibitory receptor downregulation and increased tissue invasiveness. Immunity 47, 848–861 (2017).

    Article  PubMed  CAS  Google Scholar 

  39. Kamphorst, A. O., Guermonprez, P., Dudziak, D. & Nussenzweig, M. C. Route of antigen uptake differentially impacts presentation by dendritic cells and activated monocytes. J. Immunol. 185, 3426–3435 (2010).

    Article  PubMed  CAS  Google Scholar 

  40. Huysamen, C., Willment, J. A., Dennehy, K. M. & Brown, G. D. CLEC9A is a novel activation C-type lectin-like receptor expressed on BDCA3+ dendritic cells and a subset of monocytes. J. Biol. Chem. 283, 16693–16701 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  42. Lesley, R., Kelly, L. M., Xu, Y. & Cyster, J. G. Naive CD4 T cells constitutively express CD40L and augment autoreactive B cell survival. Proc. Natl Acad. Sci. USA 103, 10717–10722 (2006).

    Article  PubMed  ADS  CAS  PubMed Central  Google Scholar 

  43. Pasqual, G. et al. Monitoring T cell-dendritic cell interactions in vivo by intercellular enzymatic labelling. Nature 553, 496–500 (2018).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  44. Satpathy, A. T. et al. Zbtb46 expression distinguishes classical dendritic cells and their committed progenitors from other immune lineages. J. Exp. Med. 209, 1135–1152 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Kohyama, M. et al. Role for Spi-C in the development of red pulp macrophages and splenic iron homeostasis. Nature 457, 318–321 (2009).

    Article  PubMed  ADS  CAS  Google Scholar 

  46. Wu, X. et al. Mafb lineage tracing to distinguish macrophages from other immune lineages reveals dual identity of Langerhans cells. J. Exp. Med. 213, 2553–2565 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Lybarger, L., Wang, X., Harris, M. R., Virgin, H. W., IV & Hansen, T. H. Virus subversion of the MHC class I peptide-loading complex. Immunity 18, 121–130 (2003).

    Article  PubMed  CAS  Google Scholar 

  48. Sedy, J. R. et al. B and T lymphocyte attenuator regulates T cell activation through interaction with herpesvirus entry mediator. Nat. Immunol. 6, 90–98 (2005).

    Article  PubMed  CAS  Google Scholar 

  49. Toebes, M. et al. Design and use of conditional MHC class I ligands. Nat. Med. 12, 246–251 (2006).

    Article  PubMed  CAS  Google Scholar 

  50. Andersen, R. S. et al. Parallel detection of antigen-specific T cell responses by combinatorial encoding of MHC multimers. Nat. Protocols 7, 891–902 (2012).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

K.M.M. was supported by the Howard Hughes Medical Institute and the US National Institutes of Health (R01AI150297); R.D.S. by grants from the National Institutes of Health (R01CA190700) and The Parker Institute for Cancer Immunotherapy, and a Stand Up To Cancer–Lustgarden Foundation Pancreatic Cancer Foundation Convergence Team Translational Research Grant; W.M.Y. by a grant from the National Institutes of Health (R01AI129545); M.D.B. and V.D. by fellowship grants from the National Institutes of Health (F30DK112466 and F30DK108498, respectively); S.T.F. by a postdoctoral training grant from the National Institutes of Health (T32CA95473); G.F.W., D.J.T. and J.T.D. by the National Institutes of Health (R01NS106289, T32 AI007163 and T32CA009621, respectively); and P.B. by the US National Science Foundation (DGE-1143954). We thank the Genome Technology Access Center, Department of Genetics, Washington University School of Medicine in St Louis, for help with genomic analysis. The Center is supported by Cancer Center Support Grant P30 CA91842 from the US National Cancer Institute and by Institute of Clinical and Translational Sciences/Clinical and Translational Science Award UL1 TR000448 from the US National Center for Research Resources. Aspects of studies including tetramer production were performed with assistance by the Immunomonitoring Laboratory, which is supported by the Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs and the Alvin J. Siteman Comprehensive Cancer Center that, in turn, is supported by the National Cancer Institute of the National Institutes of Health Cancer Center Support Grant (P30CA91842) and the Washington University Rheumatic Diseases Research Resource-based Center Grant (P30AR073752).

Author information

Authors and Affiliations

Authors

Contributions

S.T.F., R.W. and V.D. conceived and designed the experiments, collected the data, and performed and interpreted the analyses. S.T.F. and R.W. wrote the manuscript. J.T.D., P.B., D.J.T., T.L., L.L. and C.G.B. collected and analysed data. J.P.W. and D.J.T. helped generate the 1956-mOVA fibrosarcoma cell line. M.D.B. provided the β2mfl/fl mouse and interpreted the analyses. G.F.W., W.M.Y., W.E.G., T.L.M. and R.D.S. provided assistance with experimental design. K.M.M. conceived experiments, interpreted data, and wrote the manuscript.

Corresponding author

Correspondence to Kenneth M. Murphy.

Ethics declarations

Competing interests

R.D.S. is a cofounder, scientific advisory board member, stockholder and royalty recipient of Jounce Therapeutics and Neon Therapeutics and is a scientific advisory board member for A2 Biotherapeutics, BioLegend, Codiak Biosciences, Constellation Pharmaceuticals, NGM Biopharmaceuticals and Sensei Biotherapeutics.

Additional information

Peer review information Nature thanks Cornelis Melief, Sebastian Amigorena and the other, anonymous, reviewer(s) 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 figures and tables

Extended Data Fig. 1 cDC1 are required to prime CD4+ T cells during the tumour immune response.

a, 1956-EV and 1956-mOVA were stained with antibodies against (left) Thy1.1 and (right) OVA. b, Tumour growth curves of B6 wild-type (WT) mice injected with 106 (left)1956-EV or (right)1956-mOVA. c, Tumour growth curves of (left) B6 WT or (right) Irf8 +32–/– mice injected with 106 1956-mOVA. d, B6 WT or Irf8 +32–/– mice were subcutaneously injected with 106 1956-EV or 106 1956-mOVA. Spleens were isolated and stained for presence of SIINFEKL-Kb-tetramer+ CD8+ T cells. (Left) Representative flow plots of percentage of tetramer+ CD8+ T cells. (Right) Graph of tetramer+ CD8+ T cells as a percentage of all T cells. Data are pooled biologically independent samples from two independent experiments (n = 3 for WT EV, n = 4 for WT 1956-mOVA, n = 5 for Irf8 +32–/– 1956-mOVA).

Source data

Extended Data Fig. 2 cDC1 are required to prime CD4+ T cells during the immune response to B16F10 melanoma.

a, B16F10-EV and B16F10-mOVA were stained with antibodies against (left) Thy1.1 and (right) OVA. b, B6 WT or Irf8 +32–/– mice were subcutaneously injected with 106 B16F10-EV or 106 B16F10-mOVA. (Left) Representative flow plots of OT-II T cells 3 days after transfer. (Right) Graph of per cent proliferated OT-II transferred. Data are pooled biologically independent samples from two independent experiments (n = 4 for WT B16F10-EV, n = 6 for all other groups).*P = 0.04 (unpaired, two-tailed Mann–Whitney test).

Source data

Extended Data Fig. 3 Validation of mCherry expression and lineage tracing of Xcr1Cre mouse.

a, Schematic diagrams of the mouse Xcr1 WT allele, the targeting vector (IRES-mCherry-T2a-hCRE with FRT flanked pGK-Neo cassettes), and the targeted allele. Filled and open boxes denote coding and noncoding exons of Xcr1, respectively. b, Southern blot analysis of Xcr1+/+ and Xcr1Cre/+. Genomic DNAs were isolated from mice tails, digested with SalI, electrophoresed, and hybridized with the 5′-radiolabelled probe indicated in a. Southern blot gave a 10.6 and a 6.9 kbp band for WT and targeted allele, respectively. For Southern blot source data, see Supplementary Fig. 1. c, Southern blot analysis of Xcr1+/+ and Xcr1Cre/+. Genomic DNAs were isolated from mice tails, digested with SalI, electrophoresed, and hybridized with the 3′ radiolabelled probe indicated in a. Southern blot gave a 10.6 and a 8.2 kbp band for WT and targeted allele, respectively. For Southern blot source data, see Supplementary Fig. 1. d, Gating strategy to delineate splenic cell populations. e, FACs histograms of mCherry expression from subpopulations in d isolated from Xcr1Cre/+ and Xcr1+/+ mice. f, Graph of mCherry expression in antigen-presenting cell populations gated from d isolated from Xcr1Cre/+ and Xcr1+/+ mice. Data are pooled biologically independent samples from two independent experiments (n = 5 in all groups). g, FACs histograms for YFP expression from subpopulations in d isolated from Xcr1Cre/+ R26LSLYFP/+ and Xcr1+/+ R26LSLYFP/+ mice. h, Graph of YFP expression in splenic cell populations (Mo, monocytes; N, neutrophils; RPM, red pulp macrophages). Data are pooled biologically independent samples from two independent experiments (n = 6 for cDC1, cDC2, plasmacytoid dendritic cells (pDC) and B cells from Xcr1+/+ R26LSLYFP/+ mice, n = 7 for cDC1, cDC2, pDC and B cells from Xcr1Cre/+ R26LSLYFP/+ mice, and n = 4 for all other groups). i, (Top) Gating strategy to delineate SDLN cell populations. (Bottom) FACs histograms for YFP expression in cDC1 and cDC2 isolated from Xcr1Cre/+ R26LSLYFP/+ and Xcr1+/+ R26LSLYFP/+ mice. j, Graph of YFP expression from subpopulations in i isolated from Xcr1Cre/+ R26LSLYFP/+ and Xcr1+/+ R26LSLYFP/+ mice. Data are pooled biologically independent samples from two independent experiments (n = 3 in all groups).

Source data

Extended Data Fig. 4 Proliferation of OT-I in Xcr1Cre/+ β2mfl/fl mice receiving soluble or cell-associated OVA.

a, Representative FACs analysis and histograms of CFSE dilution of proliferated OT-I on day 3 after transfer into (left) Xcr1+/+ β2mfl/fl and (right) Xcr1Cre/+ β2mfl/fl immunized with soluble OVA. b, Graph of per cent proliferation of transferred OT-I in mice immunized with soluble OVA. Data are pooled biologically independent samples from two independent experiments (n = 5 for all groups). c, Representative FACs analysis and histograms of CFSE dilution of proliferated OT-I on day 3 after transfer into (left) Xcr1+/+ β2mfl/fl and (right) Xcr1Cre/+ β2mfl/fl immunized with cell-associated OVA. d, Graph of per cent proliferation of transferred OT-I in mice immunized with cell-associated OVA. Data are pooled biologically independent samples from two independent experiments (n = 5 Xcr1+/+ β2mfl/fl –OVA, n = 6 for Xcr1Cre/+ β2mfl/fl +OVA, n = 7 for Xcr1+/+ β2mfl/fl +OVA, and n = 8 for Xcr1Cre/+ β2mfl/fl +OVA).

Source data

Extended Data Fig. 5 Proliferation of OT-II in Xcr1Cre /+ MHCIIfl/fl and Xcr1Cre /+ MHCIILSL/– mice immunized with soluble and cell-associated OVA.

a, Representative FACS analysis of splenic CD4+ T cell percentage in WT B6, Xcr1+/+ MHCIILSL/–, and Xcr1Cre/+ MHCIILSL/– mice at steady state. b, (Left) Representative FACS analysis of splenic Treg percentage in Xcr1+/+ MHCIIfl/fl and Xcr1Cre/+ MHCIIfl/fl at steady state. (Right) Graph of splenic Treg percentage as a percentage of all CD4+ T cells. Data are pooled biologically independent samples from two independent experiments (n = 5 for Xcr1+/+ MHCIIfl/fl, n = 4 for Xcr1Cre/+ MHCIIfl/fl). c, Representative FACs analysis and histograms of CFSE dilution of proliferated OT-II on day 3 after transfer into Xcr1+/+ MHCIIfl/fl, Xcr1Cre/+ MHCIIfl/fl, and Xcr1Cre/+ MHCIILSL/– immunized with soluble OVA. d, Representative FACs analysis and histograms of CFSE dilution of proliferated OT-II on day 3 after transfer into Xcr1+/+ MHCIIfl/fl, Xcr1Cre/+ MHCIIfl/fl, and Xcr1Cre/+ MHCIILSL/– immunized with cell-associated OVA.

Source data

Extended Data Fig. 6 Analysis of cDC1 in conditionally deleted mice.

a, Graph of splenic cDC1 percentage in Xcr1+/+ β2mfl/fl and Xcr1Cre/+ β2mfl/fl. Data are pooled biologically independent samples from two independent experiments (n = 5 for all groups). P = NS (unpaired, two-tailed Mann–Whitney test). b, Graph of splenic cDC1 percentage in Xcr1+/+ MHCIIfl/fl and Xcr1Cre/+ MHCIIfl/fl. Data are pooled biologically independent samples from two independent experiments (n = 5 for all groups). P = NS (unpaired, two-tailed Mann–Whitney test). c, Graph of absolute numbers of transferred OT-I in soluble OVA treated Xcr1+/+ β2mfl/fl and Xcr1Cre/+ β2mfl/fl mice. Data are pooled biologically independent samples from two independent experiments (n = 5 for all groups). d, Graph of absolute numbers of transferred OT-II in soluble OVA treated Xcr1+/+ MHCIIfl/fl and Xcr1Cre/+ MHCIIfl/fl mice. Data are pooled biologically independent samples from two independent experiments (n = 5 for all groups). e, Graph of per cent proliferated OT-I in cell-associated treated Xcr1+/+ MHCIIfl/fl and Xcr1Cre/+ MHCIIfl/fl mice. Data are pooled biologically independent samples from two independent experiments (n = 6 for Xcr1Cre/+ MHCIIfl/fl OVA and OVA+ and n = 5 for all other groups). f, Graph of per cent proliferation of OT-I after 72 h coculture with ex vivo migratory cDC2 or cDC1 collected from tumour-draining lymph nodes of Xcr1+/+ MHCIIfl/fl or Xcr1Cre/+ MHCIIfl/fl mice injected 6 days earlier with 106 1956-mOVA cells. Cells were cultured at a ratio of 10:1 naive OT-I:cDC. Data are pooled independent samples from two independent experiments (n = 4 for all groups). g, Graph of absolute number of proliferated OT-I per well after 72 h coculture with ex vivo migratory cDC2 or cDC1 collected from tumour-draining lymph nodes of Xcr1+/+ MHCIIfl/fl, Xcr1Cre/+ MHCIIfl/fl mice six days after injection with 106 1956-mOVA. Cells were cultured at 10:1 ratio of naive OT-I:cDC. Data are pooled independent samples from two independent experiments (n = 4 for all groups).

Source data

Extended Data Fig. 7 CD40 deficiency does not affect cDC1 development.

a, SDLN flow cytometry gating for cDC1 expression of CD40. Migratory cDC1 (CD11cint MHCIIhi; Red) were overlaid for expression with resident cDC1 (CD11chi MHCIIint; Blue). (Top) Xcr1+/+ CD40fl/fl and (bottom) Xcr1Cre/+ CD40fl/fl SDLN and splenic antigen-presenting cells stained for CD40 expression. b, Gene-expression data from Xcr1+/+ CD40fl/fl and Xcr1Cre/+ CD40fl/fl cDC1 from spleens and SDLN. Green lines indicate 2-fold changes. c, Graph of per cent proliferation of OT-I after 72 h coculture with ex vivo migratory cDC2 or cDC1 collected from tumour-draining lymph nodes of Xcr1+/+ CD40fl/fl or Xcr1Cre/+ CD40fl/fl mice injected 6 days earlier with 106 1956-mOVA cells. Cells were cultured at a ratio of 10:1 naive OT-I:cDC. Data are pooled independent samples from two independent experiments (n = 3 for Xcr1+/+ CD40fl/fl cDC2 and n = 4 for all other groups). d, Graph of per cent proliferation of OT-I in tumour-draining lymph node of tumour-bearing mice 3 days after transfer. Xcr1+/+ CD40fl/fl and Xcr1Cre/+ CD40fl/fl mice were injected with 106 1956-EV or 106 1956-mOVA. Data are pooled independent samples from two independent experiments (n = 5 for Xcr1Cre/+ CD40fl/fl1956-mOVA, n = 3 for all other groups). e, Graph of per cent proliferation of transferred OT-II in soluble OVA treated Xcr1+/+ CD40fl/fl and Xcr1Cre/+ CD40fl/fl mice. Data are pooled biologically independent samples from two independent experiments (n = 2 for 0 mg Xcr1+/+ CD40fl/fl and Xcr1Cre/+ CD40fl/fl and n = 4 for all other groups). f, Graph of per cent proliferation of OT-II in tumour-draining lymph node of tumour-bearing mice 3 days after transfer. Xcr1+/+ CD40fl/fl and Xcr1Cre/+ CD40fl/fl mice were injected with 106 1956-EV or 106 1956-mOVA. Data are pooled biologically independent samples from three independent experiments (n = 4 for 1956-EV, n = 6 for 1956-mOVA Xcr1+/+ CD40fl/fl, and n = 9 for 1956-mOVA Xcr1Cre/+ CD40fl/fl). g, Gating strategy to delineate day 6 1956-mOVA tumour immune cell antigen-presenting cell populations. h, FACs histogram of CD40 expression on gated antigen-presenting cell populations from e.

Source data

Extended Data Fig. 8 T cells are required at tumour site to induce memory.

a, Schematic of FTY720 injection during primary tumour response. b, Schematic of FTY720 injection during secondary tumour response. c, Peripheral blood CD4+ and CD8+ T cell percentage in control and FTY720 treated mice. Data represent mean ± s.d. pooled biologically independent samples from two independent experiments (n = 2 control, n = 5 FTY720). d, Tumour growth curves of mice injected with FTY720 during the primary or secondary 1956 tumour implantation. Data represent mean ± s.d. pooled biologically independent samples from two independent experiments (n = 4 for FTY720 1º and n = 5 for all other groups). e, Individual mouse tumour growth curves of control or FTY720 injected mice. f, (Left) Tumour growth curves of Xcr1+/+ MHCIIfl/fl and Xcr1Cre/+ MHCIIfl/fl mice during primary and secondary 1956 tumour implantation. Individual mouse tumour growth curves of (middle) Xcr1+/+ MHCIIfl/fl or (right) Xcr1Cre/+ MHCIIfl/fl mice during primary and secondary 1956 tumour implantation. Data represent mean ± s.d. pooled biologically independent samples from two independent experiments (n = 7 for Xcr1+/+ MHCIIfl/fl and n = 6 for Xcr1Cre/+ MHCIIfl/fl). g, (Left) Tumour growth curves of Xcr1+/+ CD40fl/fl and Xcr1Cre/+ CD40fl/fl mice during primary and secondary 1956 tumour implantation. Individual mouse tumour growth curves of (middle) Xcr1+/+ CD40fl/fl or (right) Xcr1Cre/+ CD40fl/fl mice during primary and secondary 1956 tumour implantation. Data represent mean ± s.d. pooled biologically independent samples from two independent experiments (n = 4 for Xcr1+/+ CD40fl/fl and n = 7 for Xcr1Cre/+ CD40fl/fl).

Source data

Extended Data Fig. 9 OT-II CD4+ T cells fail to localize to the tumour in Xcr1Cre/+ MHCIIfl/fl mice.

a, Graph of per cent accumulation of transferred OT-I in tumours of Xcr1+/+ β2mfl/fl, Xcr1Cre/+ β2mfl/fl injected with 106 1956-EV or 1956-mOVA on day 0. OT-I cells were transferred intravenously on day 2 and assessed as a percentage of total CD45+ cells on (left) day 5 and (right) day 7. Data represent pooled biologically independent samples from two independent experiments (n = 1 for Xcr1+/+ β2mfl/fl tdLN and tumour 1956-EV day 5, n = 4 for Xcr1+/+ β2mfl/fl 1956-mOVA tdLN and tumour Day 7 n = 2 for all other groups). b, Graph of per cent accumulation of transferred OT-I in tumours of Xcr1+/+ MHCIIfl/fl, Xcr1Cre/+ MHCIIfl/fl injected with 106 1956-EV or 1956-mOVA on day 0. OT-I cells were transferred intravenously on day 2 and assessed as a percentage of total CD45+ cells on (left) day 5 and (right) day 7. Data represent pooled biologically independent samples from two independent experiments (n = 4 for Xcr1+/+ MHCIIfl/fl tdLN and tumour 1956-mOVA day 7, n = 3 for Xcr1+/+ MHCIIfl/fl tdLN and tumour 1956-mOVA day 5 and for Xcr1+/+ MHCIIfl/fl tdLN and tumour 1956-EV Day 7, n = 2 for all other samples). c, Graph of per cent accumulation of transferred OT-II in tumours of Xcr1+/+ MHCIIfl/fl, Xcr1Cre/+ MHCIIfl/fl injected with 106 1956-EV or 1956-mOVA on day 0. OT-II cells were transferred intravenously on day 2 and assessed as a percentage of total CD45+ cells on (left) day 5 and (right) day 7. Data represent pooled biologically independent samples from two independent experiments (n = 6 for Xcr1+/+ MHCIIfl/fl tdLN and tumour 1956-mOVA day 7, n = 4 for Xcr1+/+ MHCIIfl/fl tdLN and tumour 1956-EV Day 7, n = 3 for Xcr1+/+ MHCIIfl/fl tdLN and tumour 1956-mOVA day 5, n = 2 for all other samples).

Source data

Supplementary information

Supplementary Figure

This file contains full scan southern blots for Extended Figures 3b-c.

Reporting Summary

Supplementary Table

This file contains Supplementary Table 1.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ferris, S.T., Durai, V., Wu, R. et al. cDC1 prime and are licensed by CD4+ T cells to induce anti-tumour immunity. Nature 584, 624–629 (2020). https://doi.org/10.1038/s41586-020-2611-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2611-3

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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