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:

Optimization of the Irf8 +32-kb enhancer disrupts dendritic cell lineage segregation

Abstract

Autoactivation of lineage-determining transcription factors mediates bistable expression, generating distinct cell phenotypes essential for complex body plans. Classical type 1 dendritic cell (cDC1) and type 2 dendritic cell (cDC2) subsets provide nonredundant functions for defense against distinct immune challenges. Interferon regulatory factor 8 (IRF8), the cDC1 lineage-determining transcription factor, undergoes autoactivation in cDC1 progenitors to establish cDC1 identity, yet its expression is downregulated during cDC2 differentiation by an unknown mechanism. This study reveals that the Irf8 +32-kb enhancer, responsible for IRF8 autoactivation, is naturally suboptimized with low-affinity IRF8 binding sites. Introducing multiple high-affinity IRF8 sites into the Irf8 +32-kb enhancer causes a gain-of-function effect, leading to erroneous IRF8 autoactivation in specified cDC2 progenitors, redirecting them toward cDC1 and a novel hybrid DC subset with mixed-lineage phenotypes. Further, this also causes a loss-of-function effect, reducing Irf8 expression in cDC1s. These developmental alterations critically impair both cDC1-dependent and cDC2-dependent arms of immunity. Collectively, our findings underscore the significance of enhancer suboptimization in the developmental segregation of cDCs required for normal immune function.

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

Access options

Buy this article

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

Fig. 1: The Irf8 +32-kb enhancer is supported by low-affinity AICEs.
Fig. 2: An optimized Irf8 +32-kb enhancer induces a hybrid DC lineage.
Fig. 3: An optimized Irf8 +32-kb enhancer redirects pre-cDC2 to cDC1 and hybrid DC lineages.
Fig. 4: High-affinity AICEs paradoxically reduce IRF8 expression in terminally differentiated cDC1s and compromise cDC1 lineage commitment.
Fig. 5: scRNA-seq profiling of WT and Irf8 +32H/H spleens.
Fig. 6: An optimized Irf8 +32-kb enhancer reduces cDC2 development and impairs TH2 responses.
Fig. 7: An optimized Irf8 +32-kb enhancer reduces IRF8 expression in cDC1 and impair antitumor immunity.

Similar content being viewed by others

Data availability

The bulk RNA-seq data generated in this study on splenic DC populations are available in the Gene Expression Omnibus under accession number GSE241341. The scRNA-seq data generated in this study are available under accession number GSE270060. The following datasets were previously published and reanalyzed in this study: microarrays on DC progenitors and splenic DCs (GSE66565)22, and ChIP–seq for BATF3 and IRF8 in cDC1 (GSE66899)22. The following datasets were reanalyzed previously19 and used in the current study: ChIP–seq for p300, H3K4me1 and H3K27ac in DC progenitors and DCs (GSE66899)22, the assay for transposase-accessible chromatin with sequencing (ATAC-seq) of DC progenitors (GSE132240)23, and Immunological Genome Project (ImmGen) ATAC-seq on splenic DCs (GSE100738)68. Mouse genome (mm10 assembly) Bowtie 2 indexes were downloaded from the Bowtie 2 website (https://genome-idx.s3.amazonaws.com/bt/mm10.zip). Source data are provided with this paper. All other data are available in the paper or Supplementary Information.

References

  1. Kuziora, M. A. & McGinnis, W. Autoregulation of a Drosophila homeotic selector gene. Cell 55, 477–485 (1988).

    Article  CAS  PubMed  Google Scholar 

  2. Thayer, M. J. et al. Positive autoregulation of the myogenic determination gene MyoD1. Cell 58, 241–248 (1989).

    Article  CAS  PubMed  Google Scholar 

  3. Ouyang, W. et al. Stat6-independent GATA-3 autoactivation directs IL-4-independent Th2 development and commitment. Immunity 12, 27–37 (2000).

    Article  CAS  PubMed  Google Scholar 

  4. Liu, K. & Nussenzweig, M. C. Origin and development of dendritic cells. Immunol. Rev. 234, 45–54 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Murphy, T. L. et al. Transcriptional control of dendritic cell development. Annu. Rev. Immunol. 34, 93–119 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Anderson, D. A. III, 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 

  7. den Haan, J. M., Lehar, S. M. & Bevan, M. J. CD8+ but not CD8 dendritic cells cross-prime cytotoxic T cells in vivo. J. Exp. Med. 192, 1685–1696 (2000).

    Article  Google Scholar 

  8. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Durai, V. & Murphy, K. M. Functions of murine dendritic cells. Immunity 45, 719–736 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lewis, K. L. et al. Notch2 receptor signaling controls functional differentiation of dendritic cells in the spleen and intestine. Immunity 35, 780–791 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Satpathy, A. T. et al. Notch2-dependent classical dendritic cells orchestrate intestinal immunity to attaching-and-effacing bacterial pathogens. Nat. Immunol. 14, 937–948 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mayer, J. U. et al. Homeostatic IL-13 in healthy skin directs dendritic cell differentiation to promote TH2 and inhibit TH17 cell polarization. Nat. Immunol. 22, 1538–1550 (2021).

    Article  CAS  PubMed  Google Scholar 

  13. Kumamoto, Y. et al. CD301b+ dermal dendritic cells drive T helper 2 cell-mediated immunity. Immunity 39, 733–743 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Tussiwand, R. et al. Klf4 expression in conventional dendritic cells is required for T helper 2 cell responses. Immunity 42, 916–928 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Liu, T.-T. et al. Ablation of cDC2 development by triple mutations within the Zeb2 enhancer. Nature 607, 142–148 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Schiavoni, G. et al. ICSBP is essential for the development of mouse type I interferon-producing cells and for the generation and activation of CD8α+ dendritic cells. J. Exp. Med. 196, 1415–1425 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Tussiwand, R. et al. Compensatory dendritic cell development mediated by BATF–IRF interactions. Nature 490, 502–507 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Glasmacher, E. et al. A genomic regulatory element that directs assembly and function of immune-specific AP-1–IRF complexes. Science 338, 975–980 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kim, S. et al. High amount of transcription factor IRF8 engages AP1–IRF composite elements in enhancers to direct type 1 conventional dendritic cell identity. Immunity 53, 759–774 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Onai, N. et al. Identification of clonogenic common Flt3+ M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow. Nat. Immunol. 8, 1207–1216 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Naik, S. H. et al. Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat. Immunol. 8, 1217–1226 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Farley, E. K. et al. Suboptimization of developmental enhancers. Science 350, 325–328 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Farley, E. K., Olson, K. M., Zhang, W., Rokhsar, D. S. & Levine, M. S. Syntax compensates for poor binding sites to encode tissue specificity of developmental enhancers. Proc. Natl Acad. Sci. USA 113, 6508–6513 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jindal, G. A. & Farley, E. K. Enhancer grammar in development, evolution, and disease: dependencies and interplay. Dev. Cell 56, 575–587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hentsch, B., Mouzaki, A., Pfeuffer, I., Rungger, D. & Serfling, E. The weak, fine-tuned binding of ubiquitous transcription factors to the Il-2 enhancer contributes to its T cell-restricted activity. Nucleic Acids Res. 20, 2657–2665 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jiang, J. & Levine, M. Binding affinities and cooperative interactions with bHLH activators delimit threshold responses to the dorsal gradient morphogen. Cell 72, 741–752 (1993).

    Article  CAS  PubMed  Google Scholar 

  29. Gaudet, J. & Mango, S. E. Regulation of organogenesis by the Caenorhabditis elegans FoxA protein PHA-4. Science 295, 821–825 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Scardigli, R., Bäumer, N., Gruss, P., Guillemot, F. & Le Roux, I. Direct and concentration-dependent regulation of the proneural gene Neurogenin2 by Pax6. Development 130, 3269–3281 (2003).

    Article  CAS  PubMed  Google Scholar 

  31. Rowan, S. et al. Precise temporal control of the eye regulatory gene Pax6 via enhancer-binding site affinity. Genes Dev. 24, 980–985 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Swanson, C. I., Schwimmer, D. B. & Barolo, S. Rapid evolutionary rewiring of a structurally constrained eye enhancer. Curr. Biol. 21, 1186–1196 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Peterson, K. A. et al. Neural-specific Sox2 input and differential Gli-binding affinity provide context and positional information in Shh-directed neural patterning. Genes Dev. 26, 2802–2816 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Crocker, J. et al. Low affinity binding site clusters confer hox specificity and regulatory robustness. Cell 160, 191–203 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Jindal, G. A. et al. Single-nucleotide variants within heart enhancers increase binding affinity and disrupt heart development. Dev. Cell 58, 2206–2216 (2023).

    Article  CAS  PubMed  Google Scholar 

  36. Lim, F. et al. Affinity-optimizing enhancer variants disrupt development. Nature 626, 151–159 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Li, P. et al. BATF–JUN is critical for IRF4-mediated transcription in T cells. Nature 490, 543–546 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Iwata, A. et al. Quality of TCR signaling determined by differential affinities of enhancers for the composite BATF–IRF4 transcription factor complex. Nat. Immunol. 18, 563–572 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Pigni, M., Ashok, D., Stevanin, M. & Acha-Orbea, H. Establishment and characterization of a functionally competent type 2 conventional dendritic cell line. Front. Immunol. 9, 1912 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Vremec, D. et al. Maintaining dendritic cell viability in culture. Mol. Immunol. 63, 264–267 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Sulczewski, F. B. et al. Transitional dendritic cells are distinct from conventional DC2 precursors and mediate proinflammatory antiviral responses. Nat. Immunol. 24, 1265–1280 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Rodrigues, P. F. et al. pDC-like cells are pre-DC2 and require KLF4 to control homeostatic CD4 T cells. Sci. Immunol. 8, eadd4132 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Rodrigues, P. F. et al. Progenitors of distinct lineages shape the diversity of mature type 2 conventional dendritic cells. Immunity 57, 1567–1585 (2024).

    Article  CAS  PubMed  Google Scholar 

  44. Bagadia, P. et al. An Nfil3–Zeb2–Id2 pathway imposes Irf8 enhancer switching during cDC1 development. Nat. Immunol. 20, 1174–1185 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kashiwada, M., Pham, N. L., Pewe, L. L., Harty, J. T. & Rothman, P. B. NFIL3/E4BP4 is a key transcription factor for CD8{alpha}+ dendritic cell development. Blood 117, 6193–6197 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Liu, T.-T. et al. Cis interactions in the Irf8 locus regulate stage-dependent enhancer activation. Genes Dev. 37, 291–302 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Liu, Z. et al. Dendritic cell type 3 arises from Ly6C+ monocyte-dendritic cell progenitors. Immunity 56, 1761–1777 (2023).

    Article  CAS  PubMed  Google Scholar 

  48. 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  CAS  PubMed  PubMed Central  Google Scholar 

  49. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lun, Y., Sawadogo, M. & Perry, M. Autoactivation of Xenopus MyoD transcription and its inhibition by USF. Cell Growth Differ. 8, 275–282 (1997).

    CAS  PubMed  Google Scholar 

  52. Datta, R. R. et al. A feed-forward relay integrates the regulatory activities of Bicoid and Orthodenticle via sequential binding to suboptimal sites. Genes Dev. 32, 723–736 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Stewart-Ornstein, J., Nelson, C., DeRisi, J., Weissman, J. S. & El-Samad, H. Msn2 coordinates a stoichiometric gene expression program. Curr. Biol. 23, 2336–2345 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Driever, W., Thoma, G. & Nüsslein-Volhard, C. Determination of spatial domains of zygotic gene expression in the Drosophila embryo by the affinity of binding sites for the bicoid morphogen. Nature 340, 363–367 (1989).

    Article  CAS  PubMed  Google Scholar 

  55. Parker, D. S., White, M. A., Ramos, A. I., Cohen, B. A. & Barolo, S. The cis-regulatory logic of Hedgehog gradient responses: key roles for gli binding affinity, competition, and cooperativity. Sci. Signal. 4, ra38 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Ramos, A. I. & Barolo, S. Low-affinity transcription factor binding sites shape morphogen responses and enhancer evolution. Philos. Trans. R. Soc. Lond. B Biol. Sci. 368, 20130018 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Swanson, C. I., Evans, N. C. & Barolo, S. Structural rules and complex regulatory circuitry constrain expression of a Notch- and EGFR-regulated eye enhancer. Dev. Cell 18, 359–370 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Fukaya, T., Lim, B. & Levine, M. Enhancer control of transcriptional bursting. Cell 166, 358–368 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Heist, T., Fukaya, T. & Levine, M. Large distances separate coregulated genes in living Drosophila embryos. Proc. Natl Acad. Sci. USA 116, 15062–15067 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Du, M. et al. Direct observation of a condensate effect on super-enhancer controlled gene bursting. Cell 187, 331–344.e17 (2024).

    Article  CAS  PubMed  Google Scholar 

  61. Grant, C. E., Bailey, T. L. & Noble, W. S. FIMO: scanning for occurrences of a given motif. Bioinformatics 27, 1017–1018 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kamizono, S. et al. Nfil3/E4bp4 is required for the development and maturation of NK cells in vivo. J. Exp. Med. 206, 2977–2986 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Briseño, C. G. et al. Distinct transcriptional programs control cross-priming in classical and monocyte-derived dendritic cells. Cell Rep. 15, 2462–2474 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Ou, F. et al. Enhanced in vitro type 1 conventional dendritic cell generation via the recruitment of hematopoietic stem cells and early progenitors by Kit ligand. Eur. J. Immunol. 53, e2250201 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhang, Y., Parmigiani, G. & Johnson, W. E. ComBat-seq: batch effect adjustment for RNA-seq count data. NAR Genom. Bioinform 2, lqaa078 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Camberis, M., Le Gros, G. & Urban, J., Jr. Animal model of Nippostrongylus brasiliensis and Heligmosomoides polygyrus. Curr. Protoc. Immunol. 55, 19.12.1–19.12.27 (2003).

  68. Heng, T. S. P. et al. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the US National Institute of Health (NIH) to K.M.M. (R01AI150297, R01CA248919, R21AI164142, R01AI162643 and R21AI163421). S.T.F. is a Cancer Research Institute Irvington Fellow supported by the Cancer Research Institute. We thank J. M. White at the Department of Pathology and Immunology Transgenic Mouse Core at Washington University in St. Louis and the Genetic Editing and iPS Cell Center at Washington University in St. Louis for generating the mouse models. We thank the GTAC@MGI at Washington University School of Medicine for sequencing services. We are grateful to C. Fan and C. A. Miller at Washington University School of Medicine, and Y. Zhou at Stanford University School of Medicine for their valuable advice on genomic analysis and S. Hui at Washington University School of Medicine for help with structural modeling. We acknowledge the NIH Tetramer Core Facility (contract no. 75N93020D00005) for providing SIINFEKL-Kb tetramers. We thank E. K. Farley at the University of California, San Diego, D. L. Stern at HHMI’s Janelia Research Campus and E. V. Rothenberg at the California Institute of Technology for insights about enhancer affinity and syntax.

Author information

Authors and Affiliations

Authors

Contributions

F.O., T.L.M. and K.M.M. designed the study. F.O., T.-T.L., P.D., S.T.F., S.K., H.S., R.A.O., S.J. and J.C. performed experiments with advice from J.L.P., S.D. and M.S.D. F.O. and K.M.M. wrote the manuscript with advice from T.L.M. and T.-T.L.

Corresponding author

Correspondence to Kenneth M. Murphy.

Ethics declarations

Competing interests

M.S.D. is a consultant or advisor for Inbios, Vir Biotechnology, Moderna, Merck, GlaxoSmithKline, IntegerBio and Akagera Medicines. The Diamond laboratory has received unrelated funding support in sponsored research agreements from Moderna, Vir Biotechnology, IntegerBio and Emergent BioSolutions. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: S. Houston, in collaboration with the Nature Immunology team.

Additional information

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

Extended data

Extended Data Fig. 1 The Irf8 + 32 kb comprises four AICE motifs.

(a) ATAC-seq and ChIP-seq tracks for the indicated cell types are shown for the Irf8 locus. The locations of the Irf8 + 32 kb, +41 kb and +56 kb enhancers are highlighted. The tracks are displayed using the UCSC genome browser with vertical auto scale. (b) A zoomed-in view of a 1,168 bp region surrounding the Irf8 + 32 kb enhancer highlighting the locations of AICEs. The previously deleted 149 bp (blue) and 421 bp (red) regions from Irf8 + 32 5′−/− mice and Irf8 + 32−/− mice, respectively23, are indicated relative to the AICEs. (c) Sequence of the 421 bp region deleted previously in Irf8 + 32−/− mice23, with AICEs underlined and highlighted in blue. The AICEs are displayed from left to right in the order of site 1, site 4, site 3, and site 2.

Extended Data Fig. 2 AICEs in mouse and human IRF8 enhancers show low binding affinities to the BATF3-JUNB-IRF8 complexes.

(a) Competitive EMSA showing binding of 32P-dCTP-labeled Ctla4 probe with the indicated unlabeled competitors at increasing amounts (0, 25, 100, 400, and 1600fold excess). The NEs used were combined from 293FT cells transfected with Junb and Batf3 and 293FT cells transfected with Irf8. Bands corresponding JUNB-BATF3-IRF8 complexes and free probes are indicated by the upper and lower arrows, respectively. Data shown are representative of two similar experiments. (b) EMSA performed with 32P-dCTP-labeled probes on various NEs: NEs from 293FT cells transfected with Junb and Batf3 (B), combined NEs from 293FT cells transfected with Junb and Batf3 and 293FT cells transfected with IRF8 (B + I), and NEs from MutuDCs. Bands corresponding JUNB-BATF3-IRF8 and JUNB-BATF3 complexes are indicated by the upper and lower arrows, respectively. Data shown are representative of three similar experiments. (c) Alignment of a 161 bp region encompassing the four AICEs in the mouse Irf8 + 32 kb enhancer (chr8:120,768,539-120,768,699, mm10) across several mammalian species, colored by conservation (Jalview). The six most probable AICEs (defined using FIMO and the AICE PWM from Fig. 1b) are indicated. (d) Retroviral reporter analysis of the mouse Irf8 + 32 kb enhancer and the human IRF8 + 48 kb enhancer in cDC1s and cDC2s obtained from KitL/Flt3L cultured mouse BM cells. Shown are enhancer-driven GFP expression in transduced cDC1s (pre-gate: B220- CD11c+ MHCII+ CD24+ Sirpa Thy1.1+ cells) and transduced cDC2s (pre-gate: B220 CD11c+ MHCII+ Sirpa+ Thy1.1+ cells). Numbers are GFP geometric MFI. Data shown are representative of two similar experiments. (e) Competitive EMSA performed with 32P-dCTP-labeled Ctla4 probe in the presence of 200-fold excess of various unlabeled competitors. Competitors derived from the human +48 kb enhancer are denoted as ‘Hu’. Bands corresponding JUNB-BATF3-IRF8 complexes and free probes are indicated by the upper and lower arrows, respectively. Data shown are representative of two similar experiments.

Extended Data Fig. 3 Characterization of DC progenitors and splenic DC populations.

(a) Expression levels of selected TF-encoding genes in different stages of DC development assessed by microarray. (b) IRF8 Expression across different stages of DC development in WT mice as determined by intracellular staining. Numbers are geometric MFI. Data shown are representative of two similar experiments. MDP, monocyte-dendritic cell progenitors. (c) Frequencies of the indicated DC progenitors from WT, H/+, and H/H mice as percentages of lin SiglecH BM cells. Data are pooled from three independent experiments (n = 5 for WT, 3 for H/+, and 4 for H/H mice). Statistical significance was determined by one-way ANOVA. (d) Frequencies of splenic pDCs (gated as B220 SiglecH+ splenocytes) from WT, H/+, and H/H mice. Data are pooled from three independent experiments (n = 9 for WT, 5 for H/+, and 7 for H/H mice). Statistical significance was determined by one-way ANOVA. (e) ZBTB46 expression in the indicated cell types from H/H spleens measured by intracellular staining. B cells serve as a negative control for ZBTB46 expression. (f) In vitro cross-presentation assay with different DC populations. Shown are representative flow cytometry plots depicting CellTrace Violet-labeled OT-I CD8 T cells co-cultured with the indicated DC subsets and HKLM-OVA antigen for three days (pre-gate: CD45.2 CD45.1+ CD8a+ CD4 Va2+ cells). Divided OT-I cells indicate successful cross-presentation and T cell activation. (g) Frequencies of divided OT-I cells from in vitro cross-presentation assays. Data are pooled from two independent experiments. (h) Overlaid flow cytometry plots showing splenic cDC1s (cyan), cDC2s (red), and hybrid DCs (purple) before (top panel) and after (middle and bottom panels) culturing in the specified conditions. The data presented are representative of three similar experiments. Data in c and d are presented as mean values +/− SD.

Extended Data Fig. 4 An optimized Irf8 + 32 kb enhancer maintains IRF8 expression in pre-cDC2, impeding their lineage commitment to cDC2.

(a) IRF8 Expression in the indicated DC progenitors from WT and Irf8 + 32H/H (H/H) mice as determined by intracellular staining. Numbers are geometric MFI. Data shown are representative of two similar experiments. (b) Representative flow cytometry plots depicting intracellular IRF8 in WT and H/H pre-cDC2s after 18 hours of culturing in the presence of Flt3L. (c) Frequencies of IRF8hi pre-cDC2s after 18 h of culturing. Data are pooled from three independent experiments (n = 5 for WT and H/H mice). Statistical significance was determined by unpaired, two-tailed Student’s t test. (d) Representative flow cytometry plots showing splenic cDC populations from WT, Irf4−/−, H/H, and H/H Irf4−/− mice (pre-gate: B220 SiglecH CD11c+ MHCII+ splenocytes). (e) Frequencies of splenic cDC2s and hybrid DCs from the indicated mice. Data are pooled from five independent experiments. Statistical significance was evaluated using one-way ANOVA with Dunnett’s multiple comparisons test. (f) Representative flow cytometry plot depicting cDCs differentiated from the indicated DC progenitors sorted from WT and H/H Irf4−/− BM and cultured in the presence of Flt3L for four days (pre-gate: B220 SiglecH CD11c+ MHCII+ cells). (g) Frequencies of cDC1s differentiated from WT and H/H Irf4−/− pre-cDC2s. Data are pooled from three independent experiments. Statistical significance was determined by unpaired, two-tailed Student’s t test. Data in c, e, and g are presented as mean values +/− SD. P values are indicated above the graphs.

Extended Data Fig. 5 The Irf8 + 32H enhancer drives hybrid DC development independently of the Irf8 + 41 kb enhancer.

(a) Diagram illustrating the deletion of the Irf8 + 41 kb enhancer on the Irf8 + 32H/H background using CRISPR/Cas9. (b) Sequence of a 658 bp region surrounding the Irf8 + 41 kb enhancer. Nucleotides deleted in both the Irf8 + 41−/− line23 and the Irf8 + 32H/H + 41−/− line are highlighted in red. Additional nucleotides deleted only in the Irf8 + 32H/H + 41−/− line highlighted in blue. sgRNAs target sites are underlined. (c) Representative flow cytometry plots of BM cells stained for pre-cDC1 (upper panel), and splenocytes stained for cDCs (lower panel) from WT, Irf8 + 41−/− (+41−/−), Irf8 + 32H/+ + 41−/− (+32H/+ + 41−/−), and Irf8 + 32H/H + 41−/− (+32H/H + 41−/−) mice. Chromatin phasing diagrams are displayed above the plots for context. Pre-gates: lin SiglecH Flt3+ cells for BM, and B220 SiglecH CD11c+ MHCII+ cells for spleen. (d) Frequencies of pre-cDC1s as a percentage of lin SiglecH BM cells across the indicated genotypes. Data are pooled from three independent experiments (n = 4 for WT, 5 for +41−/−, 5 for +32H/+ + 41−/−, and 4 for +32H/H + 41−/− mice). Statistical significance was determined by one-way ANOVA with Dunnett’s multiple comparison test. (e) Frequencies of splenic cDC1s, cDC2s and hybrid DCs across the indicated genotypes. Data are pooled from six independent experiments. Statistical significance was determined by one-way ANOVA with Dunnett’s multiple comparison test. (f) IRF8 expression in cDC2s and hybrid DCs from the indicated genotypes as determined by intracellular staining. Data shown are representative of two similar experiments. Data in d and e are presented as mean values +/− SD. P values are indicated above the graphs.

Extended Data Fig. 6 An optimized Irf8 + 32 kb enhancer drives convergent differentiation of pre-cDC1 and pre-cDC2 toward a hybrid DC phenotype.

(a) Left panel: heatmap visualization of the bulk RNA-seq data as in Fig. 4e. Shown are the top 464 genes differentially expressed between WT cDC1 and cDC2 identified in Fig. 2f. Right panel: zoomed-in views of selected clusters. (b) Schematics illustrating the differentiation of CDP into cDC1, cDC2, and hybrid DC in WT, H/+, and H/H mice. Cell types are color-coded based on IRF8 and BATF3 expression.

Extended Data Fig. 7 Analyses of intermediately optimized Irf8 + 32 kb enhancers.

(a) Diagrams and sequences of the different Irf8 + 32 kb enhancer alleles generated in this study. (b) Retroviral reporter activities of various Irf8 + 32 kb enhancer constructs in cDC1s obtained from Kit/Flt3L-cultured BM. Shown are enhancer-driven GFP expression in transduced cDC1s (pre-gate: B220 CD11c+ MHCII+ CD24+ Sirpa Thy1.1+ cells). Numbers are GFP geometric MFI. Data shown are pooled from two independent experiments (n = 3 for WT mice). Statistical significance was determined by one-way ANOVA with Dunnett’s multiple comparison test. (c) Representative flow cytometry plots of splenocytes stained for cDCs (pre-gate: B220 SiglecH CD11c+ MHCII+ splenocytes). (d) Frequencies of the indicated DC subsets as percentages of B220 SiglecH CD11c+ MHCII+ cDCs. Data are pooled from six independent experiments. Statistical significance was evaluated using one-way ANOVA with Dunnett’s multiple comparisons test. (e) Representative flow cytometry plots of CD11c-enriched splenocytes stained for cDCs (pre-gate: B220 CD11c+ MHCII+ splenocytes). (f) Frequencies of the indicated DC subsets as percentages of B220 CD11c+ MHCII+ cDCs. Data are pooled from two independent experiments (n = 4 for WT and I/I mice). Statistical significance was determined by unpaired, two-tailed Student’s t test. (g) Representative histograms showing intracellular IRF8 levels in DC subsets gated in (e). Numbers are geometric MFI. (h) IRF8 geometric MFI in cDC1s gated in (e). Statistical significance was determined by unpaired, two-tailed Student’s t test. Data shown are pooled from two independent experiments (n = 4 for WT and I/I mice). Data in b, d, f, and h are presented as mean values +/− SD. P values are indicated above the graphs.

Extended Data Fig. 8 scRNA-seq analysis of WT and Irf8 + 32H/H BM and spleen.

(a) Clustering of BM lin Flt3+ Kitint-lo cells from WT and H/H mice projected onto a UMAP space (22,839 total cells, with 11,441 from WT and 11,398 from H/H mice). (b) Distributions of WT (blue) and H/H (red) cells across the UMAP in (a). (c) Dot plot of top 10 differentially expressed genes for each cluster in (a). (d) Clustering of BM lin Flt3+ Kitint-lo cells in (a) split by individual biological replicates. (e) Normalized and scaled Irf8 expression in WT (upper panel) and H/H (lower panel) cells. (f) Violin plot of normalized and scaled Irf8 expression across clusters. WT (blue) and H/H (red) cells are displayed side by side for each cluster. Statistical significance was determined by a two-tailed Wilcoxon rank sum test, applying a log2 fold-change threshold of 0.1. P values are indicated above the graphs for statistically significant comparisons. (g) Clustering of splenic CD11c+ cells from Fig. 5a split by individual biological replicates. (h) Normalized and scaled expressions of selected cluster marker genes in splenic CD11c+ cells.

Supplementary information

Supplementary Information

Supplementary Fig. 1: Gating strategies for flow cytometry.

Reporting Summary

Supplementary Tables 1–4

Supplementary Table 1: EMSA probes and competitors. Supplementary Table 2: Enhancer variants utilized in retroviral reporter assays. Supplementary Table 3: Antibodies used for flow cytometry. Supplementary Table 4: gRNAs, ssODN and genotyping primers related to the new mouse strains.

Source data

Source Data

Gel 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

Ou, F., Liu, TT., Desai, P. et al. Optimization of the Irf8 +32-kb enhancer disrupts dendritic cell lineage segregation. Nat Immunol 25, 2043–2056 (2024). https://doi.org/10.1038/s41590-024-01976-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-024-01976-w

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