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:

Erythropoietin receptor on cDC1s dictates immune tolerance

Abstract

Type 1 conventional dendritic cells (cDC1s) are unique in their efferocytosis1 and cross-presenting abilities2, resulting in antigen-specific T cell immunity3 or tolerance4,5,6,7,8. However, the mechanisms that underlie cDC1 tolerogenic function remain largely unknown. Here we show that the erythropoietin receptor (EPOR) acts as a critical switch that determines the tolerogenic function of cDC1s and the threshold of antigen-specific T cell responses. In total lymphoid irradiation-induced allograft tolerance9,10, cDC1s upregulate EPOR expression, and conditional knockout of EPOR in cDC1s diminishes antigen-specific induction and expansion of FOXP3+ regulatory T (Treg) cells, resulting in allograft rejection. Mechanistically, EPOR promotes efferocytosis-induced tolerogenic maturation7,11 of splenic cDC1s towards late-stage CCR7+ cDC1s characterized by increased expression of the integrin β8 gene12 (Itgb8), and conditional knockout of Itgb8 in cDC1s impairs tolerance induced by total lymphoid irradiation plus anti-thymocyte serum. Migratory cDC1s in peripheral lymph nodes preferentially express EPOR, and their FOXP3+ Treg cell-inducing capacity is enhanced by erythropoietin. Reciprocally, loss of EPOR enables immunogenic maturation of peripheral lymph node migratory and splenic CCR7+ cDC1s by upregulating genes involved in MHC class II- and class I-mediated antigen presentation, cross-presentation and costimulation. EPOR deficiency in cDC1s reduces tumour growth by enhancing anti-tumour T cell immunity, particularly increasing the generation of precursor exhausted tumour antigen-specific CD8+ T cells13 in tumour-draining lymph nodes and supporting their maintenance within tumours, while concurrently reducing intratumoural Treg cells. Targeting EPOR on cDC1s to induce or inhibit T cell immune tolerance could have potential for treating a variety of diseases.

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

Access options

Buy this article

USD 39.95

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

Fig. 1: Efferocytotic tolerance-inducing cDC1s upregulate Epor following TLI.
Fig. 2: Absence of EPOR on cDC1s abrogates Treg cell-mediated allo-antigen-specific tolerance following TLI/ATS, resulting in allograft rejection.
Fig. 3: scRNA-seq analysis reveals that TLI/ATS promotes EPOR-dependent, efferocytosis-triggered tolerogenic maturation of splenic cDC1s.
Fig. 4: EPO-activated cDC1 EPOR supports antigen-specific FOXP3+ Treg cell induction in PLN and restrains the immunogenic maturation of CCR7+ cDC1s.
Fig. 5: EPOR expression on cDC1s hinders antigen-specific anti-tumour T cell immunity and the loss of EPOR in cDC1s leads to tumour reduction.

Similar content being viewed by others

Data availability

All transcriptional data generated in the current study were deposited at the NCBI Gene Expression Omnibus (GEO) and are publicly available through the following accession numbers: GSE253056 (bulk RNA-seq) and GSE284080 (scRNA-seq), respectively. Source data are provided with this paper.

Code availability

The scripts for replicating the RNA-seq analyses presented are accessible on GitHub (https://github.com/chansigit/Epor-cDC1-bulkRNAseq). Scripts for reproducing all scRNA-seq analyses presented are accessible on GitHub (https://github.com/chris-mcginnis-ucsf/epor_dc_tolerance) and associated processed data objects are available on Synapse (https://synapse.org/Synapse:syn64330568).

References

  1. Roquilly, A., Mintern, J. D. & Villadangos, J. A. Spatiotemporal adaptations of macrophage and dendritic cell development and function. Annu. Rev. Immunol. 40, 525–557 (2022).

    Article  CAS  PubMed  Google Scholar 

  2. Ohara, R. A. & Murphy, K. M. The evolving biology of cross-presentation. Semin. Immunol. 66, 101711 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Anderson, D. A. 3rd & Murphy, K. M. Models of dendritic cell development correlate ontogeny with function. Adv. Immunol. 143, 99–119 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Idoyaga, J. et al. Specialized role of migratory dendritic cells in peripheral tolerance induction. J. Clin. Invest. 123, 844–854 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. 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  CAS  PubMed  Google Scholar 

  6. Wohn, C. et al. Absence of MHC class II on cDC1 dendritic cells triggers fatal autoimmunity to a cross-presented self-antigen. Sci. Immunol. 5, eaba1896 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  8. Iberg, C. A., Jones, A. & Hawiger, D. Dendritic cells as inducers of peripheral tolerance. Trends Immunol. 38, 793–804 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Scandling, J. D., Busque, S., Shizuru, J. A., Engleman, E. G. & Strober, S. Induced immune tolerance for kidney transplantation. N. Engl. J. Med. 365, 1359–1360 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Scandling, J. D. et al. Tolerance and chimerism after renal and hematopoietic-cell transplantation. N. Engl. J. Med. 358, 362–368 (2008).

    Article  CAS  PubMed  Google Scholar 

  11. Bosteels, V. & Janssens, S. Striking a balance: new perspectives on homeostatic dendritic cell maturation. Nat. Rev. Immunol. 25, 125–140 (2024).

    Article  PubMed  Google Scholar 

  12. Travis, M. A. et al. Loss of integrin αvβ8 on dendritic cells causes autoimmunity and colitis in mice. Nature 449, 361–365 (2007).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Billingham, R. E., Brent, L. & Medawar, P. B. Actively acquired tolerance of foreign cells. Nature 172, 603–606 (1953).

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Bluestone, J. A. & Anderson, M. Tolerance in the age of immunotherapy. N. Engl. J. Med. 383, 1156–1166 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mehrotra, P. & Ravichandran, K. S. Drugging the efferocytosis process: concepts and opportunities. Nat. Rev. Drug Discov. 21, 601–620 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Anderson, D. A. 3rd, Dutertre, C. A., Ginhoux, F. & Murphy, K. M. Genetic models of human and mouse dendritic cell development and function. Nat. Rev. Immunol. 21, 101–115 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Ferris, S. T. et al. cDC1 prime and are licensed by CD4+ T cells to induce anti-tumour immunity. Nature 584, 624–629 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Schulz, O. & Reis e Sousa, C. Cross-presentation of cell-associated antigens by CD8alpha+ dendritic cells is attributable to their ability to internalize dead cells. Immunology 107, 183–189 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Theisen, D. & Murphy, K. The role of cDC1s in vivo: CD8 T cell priming through cross-presentation. F1000Res. 6, 98 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Mellman, I., Chen, D. S., Powles, T. & Turley, S. J. The cancer-immunity cycle: Indication, genotype, and immunotype. Immunity 56, 2188–2205 (2023).

    Article  CAS  PubMed  Google Scholar 

  23. Schenkel, J. M. et al. Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1+ CD8+ T cells in tumor-draining lymph nodes. Immunity 54, 2338–2353.e6 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Spranger, S., Dai, D., Horton, B. & Gajewski, T. F. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 31, 711–723.e4 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zagorulya, M. & Spranger, S. Once upon a prime: DCs shape cancer immunity. Trends Cancer 9, 172–184 (2023).

    Article  PubMed  Google Scholar 

  26. Murphy, T. L. & Murphy, K. M. Dendritic cells in cancer immunology. Cell. Mol. Immunol. 19, 3–13 (2022).

    Article  CAS  PubMed  Google Scholar 

  27. Meiser, P. et al. A distinct stimulatory cDC1 subpopulation amplifies CD8+ T cell responses in tumors for protective anti-cancer immunity. Cancer Cell 41, 1498–1515.e10 (2023).

    Article  CAS  PubMed  Google Scholar 

  28. Bottcher, J. P. & Reis e Sousa, C. The role of type 1 conventional dendritic cells in cancer immunity. Trends Cancer 4, 784–792 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Broz, M. L. et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 26, 938 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Roberts, E. W. et al. Critical Role for CD103+/CD141+ dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell 30, 324–336 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Balan, S., Radford, K. J. & Bhardwaj, N. Unexplored horizons of cDC1 in immunity and tolerance. Adv. Immunol. 148, 49–91 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Silva-Sanchez, A. et al. Activation of regulatory dendritic cells by Mertk coincides with a temporal wave of apoptosis in neonatal lungs. Sci. Immunol. 8, eadc9081 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liu, K. et al. Immune tolerance after delivery of dying cells to dendritic cells in situ. J. Exp. Med. 196, 1091–1097 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Canesso, M. C. C Identification of antigen-presenting cell–T cell interactions driving immune responses to food. Science 387, eado5088 (2024).

    Article  Google Scholar 

  35. Rudnitsky, A. et al. A coordinated cellular network regulates tolerance to food. Nature 644, 231–240 (2025).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gargaro, M. et al. Indoleamine 2,3-dioxygenase 1 activation in mature cDC1 promotes tolerogenic education of inflammatory cDC2 via metabolic communication. Immunity 55, 1032–1050.e1014 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Blanco, T. et al. Conventional type I migratory CD103+ dendritic cells are required for corneal allograft survival. Mucosal Immunol. 16, 711–726 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Hongo, D., Tang, X., Zhang, X., Engleman, E. G. & Strober, S. Tolerogenic interactions between CD8+ dendritic cells and NKT cells prevent rejection of bone marrow and organ grafts. Blood 129, 1718–1728 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Slavin, S., Strober, S., Fuks, Z. & Kaplan, H. S. Long-term survival of skin allografts in mice treated with fractionated total lymphoid irradiation. Science 193, 1252–1254 (1976).

    Article  ADS  CAS  PubMed  Google Scholar 

  40. Crozat, K. et al. The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8α+ dendritic cells. J. Exp. Med. 207, 1283–1292 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Grajales-Reyes, G. E. et al. Batf3 maintains autoactivation of Irf8 for commitment of a CD8α+ conventional DC clonogenic progenitor. Nat. Immunol. 16, 708–717 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhang, H. et al. EpoR-tdTomato-Cre mice enable identification of EpoR expression in subsets of tissue macrophages and hematopoietic cells. Blood 138, 1986–1997 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kuhrt, D. & Wojchowski, D. M. Emerging EPO and EPO receptor regulators and signal transducers. Blood 125, 3536–3541 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Alaluf, E. et al. Heme oxygenase-1 orchestrates the immunosuppressive program of tumor-associated macrophages. JCI insight 5, e133929 (2020).

    PubMed  PubMed Central  Google Scholar 

  48. Consonni, F. M. et al. Heme catabolism by tumor-associated macrophages controls metastasis formation. Nat. Immunol. 22, 595–606 (2021).

    Article  CAS  PubMed  Google Scholar 

  49. Doran, A. C., Yurdagul, A. Jr & Tabas, I. Efferocytosis in health and disease. Nat. Rev. Immunol. 20, 254–267 (2020).

    Article  CAS  PubMed  Google Scholar 

  50. Luo, B. et al. Erythropoeitin signaling in macrophages promotes dying cell clearance and immune tolerance. Immunity 44, 287–302 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Dikiy, S. & Rudensky, A. Y. Principles of regulatory T cell function. Immunity 56, 240–255 (2023).

    Article  CAS  PubMed  Google Scholar 

  52. Scandling, J. D. et al. Macrochimerism and clinical transplant tolerance. Hum. Immunol. 79, 266–271 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ehst, B. D., Ingulli, E. & Jenkins, M. K. Development of a novel transgenic mouse for the study of interactions between CD4 and CD8 T cells during graft rejection. Am. J. Transplant. 3, 1355–1362 (2003).

    Article  CAS  PubMed  Google Scholar 

  54. 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  CAS  PubMed  Google Scholar 

  55. Strober, S. Use of hematopoietic cell transplants to achieve tolerance in patients with solid organ transplants. Blood 127, 1539–1543 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Moon, J. J. et al. Naive CD4+ T cell frequency varies for different epitopes and predicts repertoire diversity and response magnitude. Immunity 27, 203–213 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Shao, T. Y. et al. Reproductive outcomes after pregnancy-induced displacement of preexisting microchimeric cells. Science 381, 1324–1330 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  58. Liu, F. T. & Stowell, S. R. The role of galectins in immunity and infection. Nat. Rev. Immunol. 23, 479–494 (2023).

    Article  CAS  PubMed  Google Scholar 

  59. Gonzales, G. A. et al. The pore-forming apolipoprotein APOL7C drives phagosomal rupture and antigen cross-presentation by dendritic cells. Sci. Immunol. 9, eadn2168 (2024).

    Article  CAS  PubMed  Google Scholar 

  60. Wild, A. B. et al. CD83 orchestrates immunity toward self and non-self in dendritic cells. JCI Insight 4, e126246 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Sisirak, V. et al. Digestion of chromatin in apoptotic cell microparticles prevents autoimmunity. Cell 166, 88–101 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mucida, D. et al. Retinoic acid can directly promote TGF-β-mediated Foxp3+ Treg cell conversion of naive T cells. Immunity 30, 471–472 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Larange, A. & Cheroutre, H. Retinoic acid and retinoic acid receptors as pleiotropic modulators of the immune system. Annu. Rev. Immunol. 34, 369–394 (2016).

    Article  CAS  PubMed  Google Scholar 

  64. Wu, R. et al. Mechanisms of CD40-dependent cDC1 licensing beyond costimulation. Nat. Immunol. 23, 1536–1550 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Forster, R., Davalos-Misslitz, A. C. & Rot, A. CCR7 and its ligands: balancing immunity and tolerance. Nat. Rev. Immunol. 8, 362–371 (2008).

    Article  PubMed  Google Scholar 

  66. Ohl, L. et al. CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity 21, 279–288 (2004).

    Article  CAS  PubMed  Google Scholar 

  67. Azukizawa, H. et al. Steady state migratory RelB+ langerin+ dermal dendritic cells mediate peripheral induction of antigen-specific CD4+CD25+ Foxp3+ regulatory T cells. Eur. J. Immunol. 41, 1420–1434 (2011).

    Article  CAS  PubMed  Google Scholar 

  68. Brown, H., Komnick, M. R., Brigleb, P. H., Dermody, T. S. & Esterhazy, D. Lymph node sharing between pancreas, gut, and liver leads to immune crosstalk and regulation of pancreatic autoimmunity. Immunity 56, 2070–2085.e11 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Cruz de Casas, P., Knopper, K., Dey Sarkar, R. & Kastenmuller, W. Same yet different — how lymph node heterogeneity affects immune responses. Nat. Rev. Immunol. 24, 358–374 (2023).

    Article  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  71. Dixon, K. O. et al. TIM-3 restrains anti-tumour immunity by regulating inflammasome activation. Nature 595, 101–106 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Roche, P. A. & Furuta, K. The ins and outs of MHC class II-mediated antigen processing and presentation. Nat. Rev. Immunol. 15, 203–216 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Jinushi, M. et al. MFG-E8-mediated uptake of apoptotic cells by APCs links the pro- and antiinflammatory activities of GM-CSF. J. Clin. Invest. 117, 1902–1913 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Lei, X. et al. CD4+ helper T cells endow cDC1 with cancer-impeding functions in the human tumor micro-environment. Nat. Commun. 14, 217 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  76. Bonacina, F. et al. Myeloid apolipoprotein E controls dendritic cell antigen presentation and T cell activation. Nat. Commun. 9, 3083 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  77. Kool, M. et al. The ubiquitin-editing protein A20 prevents dendritic cell activation, recognition of apoptotic cells, and systemic autoimmunity. Immunity 35, 82–96 (2011).

    Article  CAS  PubMed  Google Scholar 

  78. Reith, W., LeibundGut-Landmann, S. & Waldburger, J. M. Regulation of MHC class II gene expression by the class II transactivator. Nat. Rev. Immunol. 5, 793–806 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  80. Mortier, E. et al. Macrophage- and dendritic-cell-derived interleukin-15 receptor alpha supports homeostasis of distinct CD8+ T cell subsets. Immunity 31, 811–822 (2009).

    Article  CAS  PubMed  Google Scholar 

  81. Pittet, M. J., Di Pilato, M., Garris, C. & Mempel, T. R. Dendritic cells as shepherds of T cell immunity in cancer. Immunity 56, 2218–2230 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Prokhnevska, N. et al. CD8+ T cell activation in cancer comprises an initial activation phase in lymph nodes followed by effector differentiation within the tumor. Immunity 56, 107–124.e105 (2023).

    Article  CAS  PubMed  Google Scholar 

  83. Huang, Q. et al. The primordial differentiation of tumor-specific memory CD8(+ ) T cells as bona fide responders to PD-1/PD-L1 blockade in draining lymph nodes. Cell 185, 4049–4066.e4025 (2022).

    Article  CAS  PubMed  Google Scholar 

  84. Jansen, C. S. et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 576, 465–470 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  85. Siddiqui, I. et al. Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211.e110 (2019).

    Article  CAS  PubMed  Google Scholar 

  86. Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Rahim, M. K. et al. Dynamic CD8+ T cell responses to cancer immunotherapy in human regional lymph nodes are disrupted in metastatic lymph nodes. Cell 186, 1127–1143.e18 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  89. Zagorulya, M. et al. Tissue-specific abundance of interferon-gamma drives regulatory T cells to restrain DC1-mediated priming of cytotoxic T cells against lung cancer. Immunity 56, 386–405.e10 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Ramirez, D. E. & Turk, M. J. Th1-like Treg cells are dressed to suppress anti-tumor immunity. Immunity 56, 1437–1439 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Moreno Ayala, M. A. et al. CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8+ T cell antitumor immunity. Immunity 56, 1613–1630.e5 (2023).

    Article  CAS  PubMed  Google Scholar 

  92. Wei, X. et al. Erythropoietin protects against murine cerebral malaria through actions on host cellular immunity. Infect. Immun. 82, 165–173 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  93. Zhang, Q. et al. Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell 179, 829–845.e20 (2019).

    Article  CAS  PubMed  Google Scholar 

  94. 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).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  95. Mair, F. et al. Extricating human tumour immune alterations from tissue inflammation. Nature 605, 728–735 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  96. Wu, R. & Murphy, K. M. DCs at the center of help: origins and evolution of the three-cell-type hypothesis. J. Exp. Med. 219, e20211519 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Kim, J. M., Rasmussen, J. P. & Rudensky, A. Y. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 8, 191–197 (2007).

    Article  CAS  PubMed  Google Scholar 

  98. Nakawesi, J. et al. alphavbeta8 integrin-expression by BATF3-dependent dendritic cells facilitates early IgA responses to Rotavirus. Mucosal Immunol. 14, 53–67 (2021).

    Article  CAS  PubMed  Google Scholar 

  99. Weckel, A. et al. Long-term tolerance to skin commensals is established neonatally through a specialized dendritic cell subgroup. Immunity 56, 1239–1254.e7 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  103. Gu, Z. Complex heatmap visualization. iMeta 1, e43 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation 2, 100141 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Liberzon, A. et al. The Molecular Signatures Database Hallmark Gene Set Collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Schurch, C. M. et al. Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front. Cell 182, 1341–1359.e19 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. McGinnis, C. S. et al. MULTI-seq: sample multiplexing for single-cell RNA sequencing using lipid-tagged indices. Nat. Methods 16, 619–626 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Hao, Y. et al. Dictionary learning for integrative, multimodal and scalable single-cell analysis. Nat. Biotechnol. 42, 293–304 (2024).

    Article  CAS  PubMed  Google Scholar 

  109. Zhu, Q., Conrad, D. N. & Gartner, Z. J. deMULTIplex2: robust sample demultiplexing for scRNA-seq. Genome Biol. 25, 37 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Alquicira-Hernandez, J. & Powell, J. E. Nebulosa recovers single-cell gene expression signals by kernel density estimation. Bioinformatics 37, 2485–2487 (2021).

    Article  CAS  PubMed  Google Scholar 

  111. Phipson, B. et al. propeller: testing for differences in cell type proportions in single cell data. Bioinformatics 38, 4720–4726 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Idoyaga for providing Xcr1cre-mTFP1 mice; E. B. Rankin for providing Eporflox/flox mice; V. K. Kuchroo for providing MC38-OVA and B16F10-OVA tumour cell lines; P. Giang for preparing CD45.1+ Foxp3DTR/DTR spleens; NIH Tetramer Core Facility (NIH Contract 75N93020D00005 and RRID:SCR_026557) for providing I-Ab|mouse 2W1S|EAWGALANWAVDSA|PE-labelled tetramer; C. Zhu for processing the scRNA-seq FASTQ data and assistance with data analysis; C. Brown for assistance with resource acquisition; T. L. Roth for valuable discussion; L. L. Tolentino, K. Nguyen, C. Barclay and J. N. Delos Reyes for expert technical support in flow cytometry and fluorescence-activated cell sorting; the DIMC Core of the Stanford Diabetes Research Center; and Breakthrough T1D Center of Excellence for support. This work was supported by the following grants: U54 CA274511, CA251174, CA244114 (E.G.E.) and P01HL149626 (X.A.).

Author information

Authors and Affiliations

Authors

Contributions

X.Z. and E.G.E. conceived the study. X.Z. designed and performed the experiments, analysed data, interpreted the results and wrote the manuscript with E.G.E. C.S.M. and S.C. conducted the scRNA-seq analyses and wrote the scRNA-seq results and methods sections together with X.Z. and E.G.E. K.J.H.-G. and W.Y. prepared the scRNA-seq libraries. S.C., P.Z., N.E.R.-F. and X.Z. carried out the RNA-seq analyses. C.M.S. and J.W.H. performed the CODEX experiments. G.Y., W.G. and J.Q. assisted with flow cytometry staining and cell sorting, in vitro cell culture, T cell adoptive transfers, tumour growth studies and data recording. A.M. contributed to the flow cytometry analysis of cDC1 Epor-tdT expression in the brain and assisted with tissue preparation for in vivo studies. I.L.L. aided in the in vivo tumour studies. H.Y. and T.H. performed heart transplantation. V.M.T., W.Q. and D.B.-V. assisted with Aldh1a2 and Itgb8 animal models. B.Y. made DEC205-OVA. A.T.S. supervised the scRNA-seq analyses. K.J.H.-G., X.A., Y.X., H.P., T.C.S., M.A., D.S., H.C., A.T.S., S.S.W., B.M. and S.S. provided critical intellectual insights. E.G.E. supervised the study. All authors provided feedback on the manuscript draft.

Corresponding authors

Correspondence to Xiangyue Zhang or Edgar G. Engleman.

Ethics declarations

Competing interests

X.Z. is a cofounder and shareholder of ImmunEdge Inc. E.G.E. is a founder, shareholder and board member of ImmunEdge Inc. B.Y. is a shareholder of ImmunEdge Inc. X.Z and E.G.E. are Stanford-affiliated inventors of PCT/US2023/063997, entitled ‘Epo Receptor Agonists and Antagonists’. C.S.M. holds patents related to MULTI-seq. C.M.S. is a cofounder and scientific advisor of Vicinity Bio GmbH and is on the scientific advisory board of and has received research funding from Enable Medicine Inc., all outside the current work. T.C.S. is a scientific advisory board member for Concerto Biosciences. M.A. is a consultant, board member, and shareholder in Ionpath Inc. D.S. is a founder of Pliant Therapeutics and Glial Biosciences and is on the Genentech Scientific Review Board and the Amgen Inflammation Scientific Review Board, and an advisor to Lila Biologics, Arda Therapeutics and TCGFB Inc. H.C. is a consultant for Kumquat Biosciences and TCura Bioscience. A.T.S. is a founder of Immunai, Cartography Biosciences and Prox Biosciences, an advisor to Zafrens and Wing Venture Capital, and receives research funding from Merck Research Laboratories. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 XCR1+CD8α+ cDC1s in the spleen following TLI/ATS are bona fide cDC1s.

a, Total splenocyte frequency per spleen (UNT n=5; TLI/ATS n=10). b, Gating of CD11chighMHCIIhigh cDCs from the splenocytes. Summary graph of the frequency of cDCs in total splenocytes (UNT, n = 6; TLI/ATS, n = 7). c, Gating of CD8α+CD11b cDC1s and CD8αCD11b+ cDC2s; frequencies in cDCs (UNT, n = 6; TLI/ATS, n = 6). d, XCR1+ and e, Ki67+ cDC1 frequencies (UNT, n = 5; TLI/ATS, n = 5). f, qPCR of Batf3 and Irf8 in XCR1+CD8α+ cDC1s (n = 5/group). g, IRF8 and h, Zbtb46-GFP expression (UNT, n = 6; TLI/ATS, n = 5). i-k, MFI of MafB-mCherry expression and % of MafB-mCherry+ cells in XCR1+CD8α+ cDC1s (i), cDC2s (j) and red pulp macrophages (RPMΦs) (k). Day 0 (UNT), n = 4; TLI/ATS Day 5, n = 5; TLI/ATS Day 9, n = 5 (i,j,k). Data are shown from one experiment, representative of at least two independent experiments with similar results (a-k). Statistical analysis was performed using unpaired two-tailed Student’s t-test (a,b,c,d,e,h), or one-way ANOVA with Tukey’s multiple-comparison test (f,g,i,j,k). Data are mean ± s.e.m. (a-k).

Source data

Extended Data Fig. 2 TLI/ATS leads to widespread apoptosis and extramedullary erythropoiesis in the spleen and a marked rise in serum EPO.

a, TUNEL staining of spleen sections, UNT vs. TLI/ATS (scale bar = 200 µm). b, Spleen cell composition after ATS, TLI, or TLI/ATS; pie chart shows mean frequencies of indicated populations (n = 3). T cells (TCRβ+CD19NK1.1), B cells (CD19+TCRβNK1.1), erythroid progenitors (CD11cTER119+CD71+), cDCs (CD3εB220SiglecHPDCA-1CD11chighMHCIIhigh), other myeloid cells are subdivided into CD11b+Ly6CLy6G+, CD11b+Ly6G Ly6C+ and CD11bintF4/80+. c, Serum EPO levels over time after TLI/ATS (ELISA, n = 8). d, CD71+TER119+ erythroid progenitors in spleen (day 6) (upper) and in spleen/BM (lower) over time after TLI/ATS (n = 3). e, Co-detection by indexing (CODEX) imaging of WT C57BL/6 spleen (UNT vs. 1 day after TLI; scale bar = 500 µm). f, Scheme of EPO treatment in Epor-tdTomato-Cre mice (i.p. × 5 days). g,h, Flow cytometry showing splenic cDC1 frequency among cDCs (g) and Epor-tdT+ cDC1 frequency/MFI (h) (+PBS, n = 5; +EPO, n = 5). i-j, Frequencies of Epor-tdT+ and Epor-tdT TER119+ erythroid cells (i) and Epor-tdT+CD11bintF4/80+Epor-tdT+ RPMΦs (j), (+PBS, n = 5; +EPO, n = 5). k, CCR7 vs. Epor-tdT expression in XCR1+CD8α+ cDC1s that were gated as live-dead aquaCD3εCD19B220 SiglecHPDCA-1CD11chighMHCIIhigh; histogram overlay for CCR7+ cDC1s (UNT, n = 5; TLI/ATS, n = 5). Data are shown from one experiment, representative of at least two independent experiments with similar results (a,b,c,d,g,h,i,j,k) or from one experiment (e). Statistical analysis was performed using unpaired two-tailed Student’s t-test (g,h,i,j,k). Data are mean ± s.e.m. (d,g-k). The diagram in f was created in BioRender. Zhang, X. (2025) https://BioRender.com/cx0n3vn.

Source data

Extended Data Fig. 3 EPO-EPOR downstream signaling is activated in cDC1s following TLI/ATS.

a-c, Gene Set Enrichment Analysis (GSEA) of transcriptional profiles using the Hallmark gene set of MSigDB. NES, normalized enrichment score; FDR, false discovery rate. Red: upregulated; Blue: downregulated. TLI/ATS vs. UNT. b, Upregulated gene sets. c, Downregulated gene sets. d-e, Intracellular phospho-flow cytometric analysis of EPO-EPOR downstream signaling in live-dead blueLinSiglecHPDCA-1CD11chighMHCIIhigh. Spleens were harvested on the next day following the last dose of TLI or TLI/ATS. UNT (n = 4) vs. TLI (n = 4) vs. TLI/ATS (n = 4). d, XCR1+CD8α+ cDC1s and e, XCR1CD8α cDC2s. f,g, Histograms and MFI of the indicated EPO-EPOR downstream signaling molecules with fluorescence minus one (FMO) as controls by intracellular phospho-flow staining on the next day following the last dose of TLI/ATS treatment. Eporflox/flox (n = 4) vs. EporΔXcr1 (n = 5). cDC1s (f) and cDC2s (g). h, Ex vivo analysis of EPO-EPOR downstream signaling in splenic cDC1s. Splenic cDCs were MACS-purified with a pan-DC isolation kit and cultured at 5 × 106 cells/ml, then rested overnight. Cells were isolated from UNT or TLI/ATS-treated Eporflox/flox (n = 4; n = 4) and EporΔXcr1 (n = 4; n = 4) mice. cDCs from TLI/ATS-treated mice were stimulated ex vivo with EPO (10 IU/200 μl) or PBS (control) overnight. Phosphorylation of downstream signaling molecules was assessed by flow cytometry, after gating on XCR1+SIRPα splenic cDC1s. Data are shown from one experiment, representative of at least two independent experiments with similar results (d-h). Statistical analysis was performed using unpaired two-tailed Student’s t-test (f,g), or one-way ANOVA Tukey’s multiple-comparison test (d, e and h left), or paired two-tailed Student’s t-test (h right). Data are mean ± s.e.m. (d-h).

Source data

Extended Data Fig. 4 FOXP3+ Tregs play an indispensable role in TLI/ATS-induced cDC1 EPOR-dependent immune tolerance.

a, Representative pseudocolor plots showing FOXP3+ Treg depletion efficiency in recipient mice on day 6 after DT treatment (DT injections on days 0, 2, and 4). b,c, Representative pseudocolor plots of C57BL/6, Batf3−/−, or EporΔXcr1 recipient conventional CD4+ T cell percentages and FOXP3+ Treg percentages in CD4+ T cells. d,e, Absolute cell number of indicated cell populations. b,d, Day 0 (UNT, n = 5; n = 5; n = 5 and TLI/ATS, n = 6; n = 5; n = 4) and c,e, Day 14 of UNT or TLI/ATS-treated groups post allo-BM infusion (UNT, n = 11; n = 6; n = 9 and TLI/ATS, n = 3; n = 11; n = 10). f, MHCII expression on cDC1s and cDC2s from MHCIIflox/flox and MHCIIΔXcr1 spleens. g,h,i, MHCIIflox/flox (n = 6) and MHCIIΔXcr1 (n = 6) recipients were given TLI/ATS and i.v. infused with BALB/c donor BM cells. 14 days post BM infusion, the percentages of donor type (H2Kd+) cells among leukocyte populations were determined in the peripheral blood of hosts. g,i, Recipient MHCI (H-2Kb)+TCRβ+CD4+ T cell frequency among total live cells and FOXP3+ frequency among CD4+ T cells were analyzed on day 14. j, CD45.2+FOXP3WTEporflox/flox (+ PBS/without DT, n = 8; +DT, n = 8) or EporΔXcr1 (+ PBS/ without DT, n = 8; +DT, n = 8) mice were injected with 30 million CD45.1+FOXP3DTR CD4+ T cells isolated by MACS. Two consecutive doses of DT or PBS were given on each of the following 2 days. Subsequently, the mice were treated with TLI/ATS, and 2W1S-BALB/c donor BM cells were infused i.v., and 14 days later, 2W1S-tetramer+CD44+H-2Kb+TCRβ+CD4+ T cells from the spleens were analyzed for FOXP3 expression by flow cytometry. FOXP3 expression in CD45.1+ or CD45.2+2W1S-tetramer+ CD4+ T cells is shown. One experiment (j) or one of two independent experiments with similar results are shown (a-i). Statistical analysis was performed using unpaired two-tailed Student’s t-test (g,i), two-way ANOVA with Tukey’s multiple-comparison test (d,e,j). Data are mean ± s.e.m. (d,e,g,i,j). The diagram in j was created in BioRender. Zhang, X. (2025) https://BioRender.com/cx0n3vn.

Source data

Extended Data Fig. 5 Differentially expressed genes (DEGs) in cDC1s in scRNA-seq analysis and ex vivo TGFβ-dependent Ag-specific FOXP3+ Treg induction by CCR7+ cDC1s.

a, UMAP of splenic cDC1 gene expression by sample identity. b, Dot plots of top condition-specific DEGs in Eporflox/flox and EporΔXcr1 mice (TLI/ATS vs. UNT). c, Absolute cDC1 numbers per spleen in UNT vs. TLI/ATS-treated Eporflox/flox (n = 5/condition) and EporΔXcr1 (n = 5/condition) mice. d, UMAPs of cDC1 subtypes in Epor-tdT+ and Epor-tdT cells. e-g, Dot plots of top condition-specific DEGs in Epor-tdT+ and Epor-tdT cDC1s (TLI/ATS) and in Eporflox/flox and EporΔXcr1 mice (TLI/ATS vs. UNT). Dot color = expression, size = % of indicated gene expressed cells (b,e-g). h, Bar charts showing cDC subtype (d) proportions in Epor-tdT+ and Epor-tdT cDC1s following TLI/ATS. i, Role of TGFβ in FOXP3+ Treg induction by CCR7+ cDC1s: 12 h after apoptotic Act-mOVA injection, CCR7+ cDC1s (1×104) were cocultured with CD45.1+ CTV-labeled naïve OT-II cells ± anti-TGFβ; FOXP3 expression was analyzed by flow cytometry (n = 5/group). j,k, Representative flow cytometry analysis and l,m, Absolute cell number of indicated cell populations of Fig. 3j, Itgb8ΔXcr1 vs. littermate controls. j,l, Day 0 and k,m, Day 14 of UNT (n = 5; n = 5) or TLI/ATS-treated (n = 5; n = 5) groups post allo-BM infusion. n,o, Aldh1a2ΔCD11c: Batf3−/− (n = 6) vs. Aldh1a2flox/flox: Batf3−/− (n = 7) BM chimeric recipient mice (CD45.1+) were given TLI/ATS. 1 day after the last dose of TLI/ATS, 2W1S-BALB/c donor BM cells were infused i.v., and 14 days later, the percentages of donor type (H2Kd+) cells among leukocyte populations in the peripheral blood of hosts were determined (n) and 2W1S-tetramer+CD44+H-2Kb+TCRβ+CD4+ T cells from the spleens were analyzed for FOXP3 expression by flow cytometry and FOXP3+ Tregs were counted (o). Data are representative of at least three independent experiments with similar results (c,i) or one experiment (j-o). Statistical analysis was performed using unpaired two-tailed Student’s t-test (c,i,n,o), or two-way ANOVA followed by Tukey’s multiple-comparison test with P values adjusted (l,m), or propeller test, two-sided, no multiple-comparison correction (b), or wilcoxon rank sum test, two-sided, Bonferroni correction (h). Data are mean ± s.e.m. (c,i,l,m,n,o). The diagram in i was created in BioRender. Zhang, X. (2025) https://BioRender.com/rq2yp2e.

Source data

Extended Data Fig. 6 Absence of EPOR on cDC1s gives rise to immunogenic cDC1s that promote both CD8+ T cell cross-priming and CD4+ T cell priming to cell-associated Ags.

a, MFI of indicated molecules on gated cDC1s with fluorescence minus one (FMO) as controls. b, MFI of indicated molecules on gated cDC2s with fluorescence minus one (FMO) as controls. c, Percentages of cDC2s in splenic cDCs. d, Representative flow gating of CCR7+XCR1+SIRPα cDC1s in splenic cDC1s (Upper), and percentages and absolute numbers of CCR7+ cDC1s (Lower). a-d, Eporflox/flox (n = 5) vs. EporΔXcr1 (n = 5) mice. e, MFI of indicated molecules on CCR7+ vs. CCR7 cDC1s. CD40 and PD-L1: Eporflox/flox (n = 5); EporΔXcr1 (n = 5). CD80 and CD86: Eporflox/flox (n = 6); EporΔXcr1 (n = 6). f, Cross-presentation assay: apoptotic Act-mOVA thymocytes injected into Eporflox/flox (n = 5) or EporΔXcr1 (n = 5) mice 1 day after transfer of CTV-labeled naïve CD45.1+ naïve OT-I cells; spleens analyzed on day 4 for OT-I expansion and proliferation. g, Same setup with OT-II cells; percentages and absolute numbers of OT-II cells and proliferating OT-II cells were assessed. Ag-specific CD4+ T cell response: Ag-specific CD4+ T cell immune response following i.v. injection of apoptotic Act-mOVA thymocytes 1 day after i.v. injection of CTV-labeled naïve CD45.1+ naïve OT-II cells. Spleens were analyzed at day 4 for OT-II expansion and proliferation. Eporflox/flox (n = 5) and EporΔXcr1 (n = 5) mice. Data are shown from one experiment, representative of at least three independent experiments with similar results (a-g). Statistical analysis was performed using unpaired two-tailed Student’s t-test (a,b,c,d,f,g) and two-way ANOVA followed by Tukey’s multiple-comparison test (e). Data are mean ± s.e.m. (a-g). The diagrams in f,g were created in BioRender. Zhang, X. (2025) https://BioRender.com/bth22u6.

Source data

Extended Data Fig. 7 Phenotypes of T cells in the spleens of Eporflox/flox vs. EporΔXcr1 mice and role of EPOR in cDC1-mediated cell-associated Ag-specific CD4+ T cell priming and proliferation and FOXP3+ Treg induction.

a-e, Percentages and absolute numbers of CD4+ T cells (a), FOXP3+CD25+ Tregs in CD4+ T cells (b), CD44highCD62Llow effector cells and CD44lowCD62Lhigh naïve cells in CD4+ T cells (c), CD8+ T cells (d), and CD44highCD62Llow effector cells and CD44lowCD62Lhigh naïve cells in CD8+ T cells (e) in the spleens of EporΔXcr1 and littermate Eporflox/flox control mice with representative flow cytometric plots. a-e, Eporflox/flox, n = 5; EporΔXcr1, n = 5. f,g, Flow cytometry-based measurement of cell-associated Ag-specific CD4+ T cell immune response in the spleen following i.v. injection of apoptotic Act-mOVA thymocytes into mice of the indicated genotypes 1 day after i.v. injection of CTV-labeled naïve CD45.1+ OT-II cells. f, WT C57BL/6 (n = 5) and Batf3−/− (n = 7). g, FOXP3+ Treg induction in Eporflox/flox and EporΔXcr1 mice. Recombinant EPO or PBS was administered daily, from Day −3 to Day 4. +PBS: Eporflox/flox (n = 5), EporΔXcr1 (n = 5); +EPO: Eporflox/flox (n = 5), EporΔXcr1 (n = 5) mice. Data are shown from one experiment, representative of at least three independent experiments with similar results (a-e), or two independent experiments with similar results (f,g). Statistical analysis was performed using unpaired two-tailed Student’s t-test (a-f), or two-way ANOVA followed by Tukey’s multiple-comparison test (g). Data are mean ± s.e.m. (a-g). The diagrams in f,g were created in BioRender. Zhang, X. (2025) https://BioRender.com/bth22u6.

Source data

Extended Data Fig. 8 Epor-tdT expression on XCR1+ cDC1s in selected organs and tolerogenic phenotype of Epor-tdT+ migratory cDC1s in PLN.

a,b,c CCR7- and Batf3-dependent Epor-tdT expression on migratory cDCs in pLNs. Migratory cDCs were gated as CD11cintMHCIIhigh from live-dead aquaLinSiglecHPDCA-1EpCAM cells, and resident cDCs were gated as CD11chighMHCIIint from live-dead aquaLinSiglecHPDCA-1 cells. pLNs including inguinal, axillary, brachial, and superficial cervical LNs were combined for analysis by flow cytometry (a,b). Ccr7−/−EportdT/+, Batf3−/−EportdT/+ and WT C57BL/6 mice (a). Histogram overlay of Epor-tdT expression on migratory or resident cDCs from individual mouse strains (b). Epor-tdT expression on migratory cDCs from individual pLNs of EportdT/+ mice (c). d, Epor-tdT expression on cDC1s obtained from the indicated organs in Zbtb46GFP/+EportdT/+ mice. cDCs were gated in CD45+ cells as CD11c+Zbtb46-GFP+, in which cDC1s were further gated as XCR1+CD103+. e, Flow cytometric analysis of tolerance associated cell-surface molecules on PLN Epor-tdT+ migratory cDC1s compared with Epor-tdT cDCs and resident cDCs with FMO serving as controls (n = 8). Data are representative of at least two independent experiments with similar results (a-e). Statistical analysis was performed using one-way ANOVA Tukey’s multiple-comparison test (e). Data are mean ± s.e.m. (e). The diagrams in a,e were created in BioRender. Zhang, X. (2025) https://BioRender.com/5sr2iny.

Source data

Extended Data Fig. 9 Induction of Ag-specific CD4+FOXP3+ Tregs by PLN migratory Epor-tdT+ cDC1s.

a, FACS-sorted PLN Epor-tdT+ (n = 4) or Epor-tdT (n = 4) XCR1+ migratory cDC1s from Epor-tdT mice were cocultured for 5 days with CTV-labeled naïve OT-II cells + DEC205-OVA (ratio 1:5); FOXP3 expression in OT-II cells was analyzed by flow cytometry. b, Same setup as (a) but with apoptotic Act-mOVA thymocytes (ratio 1:5:2) ± EPO (20 IU per well per day for 5 days); FOXP3 expression in OT-II cells was measured. Epor-tdT+ (+ PBS or w/o, n = 4; +EPO, n = 4) or Epor-tdT (+ PBS or w/o, n = 4; +EPO, n = 3) c, FACS-sorted PLN Epor-tdT+ or Epor-tdT migratory cDC1s were cocultured for 12 h with apoptotic CD45.1+ thymocytes ± EPO; MFIs of surface markers were analyzed. Epor-tdT+: n = 2. Epor-tdT: n = 2. d, Migratory cDC1s from Eporflox/flox or EporΔXcr1 mice were cocultured with naïve CTV-labeled OT-II cells and Act-mOVA thymocytes (1:5:2) ± EPO (20 IU per well per day for 5 days); FOXP3 induction was assessed. n = 6/group. e, Efferocytosis of PKH67-labeled apoptotic thymocytes by migratory cDC1s and cDC2s in dLNs 12 h post-injection. f, Act-mOVACD45.1/CD45.2 mice were reconstituted with either Eporflox/flox (n = 5) or EporΔXcr1 (n = 6) BM cells after lethal irradiation. 8 weeks post-reconstitution, naïve CTV-labeled OT-II cells were i.v. infused (day 0), and EPO was administered on days −2 to 2. FOXP3 induction in OT-II cells was assessed in inguinal LNs on day 9. Data are shown from one experiment, representative of two independent experiments with similar results (a-e) or one (f) independent experiment. Statistical analysis was performed using unpaired two-tailed Student’s t-test (a,b,f), two-way ANOVA with Tukey’s multiple-comparison test (d). Data are mean ± s.e.m. (a,b,c,d,f). The diagrams in a,b,c,e,f were created in BioRender. Zhang, X. (2025) https://BioRender.com/u560oi2.

Source data

Extended Data Fig. 10 Phenotypes of T cells in the PLNs of Eporflox/flox vs. EporΔXcr1 mice.

a-e, Percentages and absolute numbers of CD4+ T cells (a), FOXP3+CD25+ Tregs in CD4+ T cells (b), CD44highCD62Llow effector cells and CD44lowCD62Lhigh naïve cells in CD4+ T cells (c), CD8+ T cells (d), and CD44highCD62Llow effector cells and CD44lowCD62Lhigh naïve cells in CD8+ T cells (e) in the PLNs of EporΔXcr1 and littermate Eporflox/flox control mice with representative flow cytometry plots. a-e, Eporflox/flox (n = 9) and EporΔXcr1 (n = 9). Data are shown from one experiment, representative of at least three independent experiments with similar results (a-e). Statistical analysis was performed using unpaired two-tailed Student’s t-test (a-e). Data are mean ± s.e.m. (a-e).

Source data

Extended Data Fig. 11 EPOR expression on tumor-infiltrating leukocytes (TILs), tumor Ag-carrying migratory cDC1s in TDLNs and tumors, and the correlation of tumor growth with systemic EPO levels.

a, Zbtb46GFP/+EportdT/+ mice were implanted s.c. with MC38 or B16F10 tumor, or EO771 tumor in the mammary fat pad. On day 12, tumors were harvested for flow cytometric analysis of Epor-tdT on cDCs (live-dead aquaCD45+Zbtb46-GFP+CD11c+); cDC1s were gated as XCR1+ and non-cDC1s as XCR1. b-d, Mice implanted s.c. with MC38-OVAdim or B16F10-OVA; on day 10, tumors were analyzed for Epor-tdT expression in TILs. b, Representative gating strategy of individual live-dead blue TIL populations. c,d, Histogram overlay showing Epor-tdT expression in individual cell populations. e, Zbtb46GFP/+EportdT/+ mice with MC38-OVAdim tumors (day 12) were analyzed for Epor-tdT on tumor-infiltrating cDCs; CCR7+ (population I) and CCR7 (populations II/III by Ly6A) subsets were gated, with XCR1/CD103 staining to define cDC1s and cDC2s. f, Quantification of Epor-tdT expression on individual tumor infiltrating cDC subsets. MC38-OVAdim (n = 4) and B16F10-OVA (n = 4) tumors were harvested on day 12 post-s.c. implantation for flow cytometry. Gating strategy as in Fig. 5a and Extended Data Fig. 11e. g, Flow cytometry analysis of Epor-tdT expression on TDLN migratory cDC1s. Overlay of migratory cDC1s with Lin live cells to show Epor-tdT expression levels. h, Serum EPO levels were measured by ELISA on the indicated days after s.c. implantation of MC38-OVAdim (n = 6) or B16F10-OVA (n = 5) tumors in WT mice. i,j,k, B16F10-OVA-ZsGreen cells were s.c. implanted into EportdT/+ mice, and tdLN and tumor were analyzed on day 9 after inoculation. j, Flow cytometry analysis of Epor-tdT expression on tdLN ZsGreen+ migratory and resident XCR1+ cDC1s. Overlay of migratory ZsGreen+ cDC1s or resident ZsGreen+ cDC1s with Linlive cells to show Epor-tdT expression levels. k, Flow cytometry analysis of Epor-tdT expression on tumor infiltrating ZsGreen+ cDC1s. Data are shown from one experiment, representative of at least two independent experiments with similar results (a-e,f,g,h,j,k). Statistical analysis was performed using one-way ANOVA with Dunnett’s multiple-comparison test (f). Data are mean ± s.e.m. (f,h). The diagrams in a,b,e,g,i were created in BioRender. Zhang, X. (2025) https://BioRender.com/gjjtedh.

Source data

Extended Data Fig. 12 Loss of EPOR in cDC1s limits tumor growth and promotes immunogenic function of tumor Ag-carrying cDC1s in both TDLN and tumor.

a, Growth of MC38-OVAdim tumor cells implanted s.c. into Eporflox/flox (n = 8) and EporΔXcr1 mice (n = 9). b, Growth of B16F10-OVA tumor cells implanted s.c. into Eporflox/flox (n = 6) and EporΔXcr1 mice (n = 7). c, Experimental design for phenotyping tumor-Ag carrying ZsGreen+ cDC1s in tdLN and tumors in Eporflox/flox vs. EporΔXcr1 mice. B16F10-OVA-ZsGreen cells were implanted s.c. into EportdT/+ mice, and TDLNs and tumors were analyzed on day 9 after implantation. d, Flow cytometry analysis of Epor-tdT expression on ZsGreen+ migratory XCR1+SIRPα cDC1s in tdLNs with summary graph of statistical quantification. Eporflox/flox (n = 7) and EporΔXcr1 (n = 8). e, Flow cytometry analysis of CD40, CD80 and CD86 expression on tumor infiltrating ZsGreen+ cDC1s with summary graph of statistical quantification. Eporflox/flox (n = 7) and EporΔXcr1 (n = 8) mice. (f-l) MC38-OVAdim tumors were s.c. implanted into Eporflox/flox (n = 8) and EporΔXcr1 (n = 7) and 10 days later TILs were analyzed. f. Percentages of CD45+ live immune cells and CD8+ or CD4+ T cells in CD45+ TILs. g, Frequency of OVA257-264-dextramer+CD8+ T cells among CD8+ T cells. h, Representative flow plots and quantification of CD8+ T cells expressing TIM-3 and PD-1. i, Representative flow plots and quantification of TCF1+TIM-3CD8+ T cells. j, Representative histograms and quantification of perforin, granzyme-B, IFNγ and TNFα expression in tumor-infiltrating CD8+ T cells. k, Percentage of FOXP3+ Tregs in CD4+ T cells with representative flow plots (Left). Absolute number of Tregs (Right). l, Representative flow plots and percentages of T-bet+CXCR3+ Tregs in CD4+ FOXP3+ Tregs. (f-l). Data are shown from one experiment, representative of at least two independent experiments with similar results (a,b,d,e,f-l). Statistical analysis was performed using two-way ANOVA with Šídák’s multiple comparison test (a,b), or two-tailed unpaired Student’s t-test (d,e,f,g,i,j,k,l), or two-way ANOVA with Tukey’s multiple-comparison test (h). Data are mean ± s.e.m. (a,b,d,e-l). The diagram in c was created in BioRender. Zhang, X. (2025) https://BioRender.com/3jod9q7.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2, which contain the full gene list used to generate the signature and a list of detected differentially expressed genes (related to Fig. 4).

Reporting Summary

Peer Review File

Source data

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

Zhang, X., McGinnis, C.S., Yu, G. et al. Erythropoietin receptor on cDC1s dictates immune tolerance. Nature (2025). https://doi.org/10.1038/s41586-025-09824-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Version of record:

  • DOI: https://doi.org/10.1038/s41586-025-09824-z

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