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:

Single-cell analyses reveal the clonal and molecular aetiology of Flt3L-induced emergency dendritic cell development

Abstract

Regulation of haematopoietic stem and progenitor cell (HSPC) fate is crucial during homeostasis and under stress conditions. Here we examine the aetiology of the Flt3 ligand (Flt3L)-mediated increase of type 1 conventional dendritic cells (cDC1s). Using cellular barcoding we demonstrate this occurs through selective clonal expansion of HSPCs that are primed to produce cDC1s and not through activation of cDC1 fate by other HSPCs. In particular, multi/oligo-potent clones selectively amplify their cDC1 output, without compromising the production of other lineages, via a process we term tuning. We then develop Divi-Seq to simultaneously profile the division history, surface phenotype and transcriptome of individual HSPCs. We discover that Flt3L-responsive HSPCs maintain a proliferative ‘early progenitor’-like state, leading to the selective expansion of multiple transitional cDC1-primed progenitor stages that are marked by Irf8 expression. These findings define the mechanistic action of Flt3L through clonal tuning, which has important implications for other models of ‘emergency’ haematopoiesis.

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: Supra-physiological levels of Flt3L drive the expansion of BM HSPC populations and promote emergency cDC1 development.
Fig. 2: Cellular barcoding reveals minimal contribution of clonal cDC1 fate activation to emergency cDC1 generation.
Fig. 3: Selective clonal expansion is the major driver of emergency cDC1 generation.
Fig. 4: Divi-Seq reveals increased cell division in HSPCs during the early Flt3L response.
Fig. 5: Maintenance of a progenitor-like state in Flt3L-treated cells.
Fig. 6: Emergence of a cDC1 precursor after short-term exposure to Flt3L.
Fig. 7: Flt3L treatment alters the expression of cDC1 genes.
Fig. 8: Flt3L stimulation selectively expands Irf8-expression cDC1-primed progenitors.

Similar content being viewed by others

Data availability

Divi-Seq data that support the findings of this study have been deposited in the Gene Expression Omnibus under the accession code GSE147977. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

All of the R packages that were used are available online, as described in the Methods. Customized code used to analyse Divi-Seq and barcoding data are available on GitHub https://github.com/DawnSLin/Emergency_DC_Development.

References

  1. Boettcher, S. et al. Cutting edge: LPS-induced emergency myelopoiesis depends on TLR4-expressing nonhematopoietic cells. J. Immunol. 188, 5824–5828 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Guermonprez, P. et al. Inflammatory Flt3l is essential to mobilize dendritic cells and for T cell responses during Plasmodium infection. Nat. Med. 19, 730–738 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Manz, M. G. & Boettcher, S. Emergency granulopoiesis. Nat. Rev. Immunol. 14, 302–314 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. King, K. Y. & Goodell, M. A. Inflammatory modulation of HSCs: viewing the HSC as a foundation for the immune response. Nat. Rev. Immunol. 11, 685–692 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Schürch, C. M., Riether, C. & Ochsenbein, A. F. Cytotoxic CD8+ T cells stimulate hematopoietic progenitors by promoting cytokine release from bone marrow mesenchymal stromal cells. Cell Stem Cell 14, 460–472 (2014).

    Article  PubMed  Google Scholar 

  6. Zhao, J. L. et al. Conversion of danger signals into cytokine signals by hematopoietic stem and progenitor cells for regulation of stress-induced hematopoiesis. Cell Stem Cell 14, 445–459 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Takizawa, H., Boettcher, S. & Manz, M. G. Demand-adapted regulation of early hematopoiesis in infection and inflammation. Blood 119, 2991–3002 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Naik, S. H. et al. Diverse and heritable lineage imprinting of early haematopoietic progenitors. Nature 496, 229–232 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Notta, F. et al. Distinct routes of lineage development reshape the human blood hierarchy across ontogeny. Science 351, aab2116 (2016).

    Article  PubMed  Google Scholar 

  10. Nestorowa, S. et al. A single-cell resolution map of mouse hematopoietic stem and progenitor cell differentiation. Blood 128, e20–e31 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Velten, L. et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat. Cell Biol. 19, 271–281 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lee, J. et al. Lineage specification of human dendritic cells is marked by IRF8 expression in hematopoietic stem cells and multipotent progenitors. Nat. Immunol. 18, 877–888 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lin, D. S. et al. DiSNE movie visualization and assessment of clonal kinetics reveal multiple trajectories of dendritic cell development. Cell Rep. 22, 2557–2566 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Laurenti, E. & Gottgens, B. From haematopoietic stem cells to complex differentiation landscapes. Nature 553, 418–426 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. 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  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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 

  18. Salmon, H. et al. Expansion and activation of CD103+ dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity 44, 924–938 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Spranger, S., Bao, R. & Gajewski, T. F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523, 231–235 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. McKenna, H. J. et al. Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells. Blood 95, 3489–3497 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Ginhoux, F. et al. The origin and development of nonlymphoid tissue CD103+ DCs. J. Exp. Med. 206, 3115–3130 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Durai, V. et al. Altered compensatory cytokine signaling underlies the discrepancy between Flt3−/− and Flt3l−/− mice. J. Exp. Med. 215, 1417–1435 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kingston, D. et al. The concerted action of GM-CSF and Flt3-ligand on in vivo dendritic cell homeostasis. Blood 114, 835–843 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Maraskovsky, E. et al. Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified. J. Exp. Med. 184, 1953–1962 (1996).

    Article  CAS  PubMed  Google Scholar 

  27. O’Keeffe, M. et al. Effects of administration of progenipoietin 1, Flt-3 ligand, granulocyte colony-stimulating factor, and pegylated granulocyte-macrophage colony-stimulating factor on dendritic cell subsets in mice. Blood 99, 2122–2130 (2002).

    Article  PubMed  Google Scholar 

  28. Sanchez-Paulete, A. R. et al. Cancer immunotherapy with immunomodulatory anti-CD137 and anti-PD-1 monoclonal antibodies requires BATF3-Dependent dendritic cells. Cancer Disco. 6, 71–79 (2016).

    Article  CAS  Google Scholar 

  29. Curran, M. A. & Allison, J. P. Tumor vaccines expressing Flt3 ligand synergize with CTLA-4 blockade to reject preimplanted tumors. Cancer Res. 69, 7747–7755 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Dupont, C. D. et al. Flt3 ligand is essential for survival and protective immune responses during toxoplasmosis. J. Immunol. 195, 4369–4377 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Gregory, S. H., Sagnimeni, A. J., Zurowski, N. B. & Thomson, A. W. Flt3 ligand pretreatment promotes protective immunity to Listeria monocytogenes. Cytokine 13, 202–208 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Reeves, R. K., Wei, Q., Stallworth, J. & Fultz, P. N. Systemic dendritic cell mobilization associated with administration of FLT3 ligand to SIV- and SHIV-infected macaques. AIDS Res Hum. Retroviruses 25, 1313–1328 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Shortman, K. Dendritic cell development: a personal historical perspective. Mol. Immunol. 119, 64–68 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Hudak, S. et al. FLT3/FLK2 ligand promotes the growth of murine stem cells and the expansion of colony-forming cells and spleen colony-forming units. Blood 85, 2747–2755 (1995).

    Article  CAS  PubMed  Google Scholar 

  35. Jacobsen, S. E., Okkenhaug, C., Myklebust, J., Veiby, O. P. & Lyman, S. D. The FLT3 ligand potently and directly stimulates the growth and expansion of primitive murine bone marrow progenitor cells in vitro: synergistic interactions with interleukin (IL) 11, IL-12, and other hematopoietic growth factors. J. Exp. Med. 181, 1357–1363 (1995).

    Article  CAS  PubMed  Google Scholar 

  36. Sudo, Y. et al. Synergistic effect of FLT-3 ligand on the granulocyte colony-stimulating factor-induced mobilization of hematopoietic stem cells and progenitor cells into blood in mice. Blood 89, 3186–3191 (1997).

    Article  CAS  PubMed  Google Scholar 

  37. Neipp, M., Zorina, T., Domenick, M. A., Exner, B. G. & Ildstad, S. T. Effect of FLT3 ligand and granulocyte colony-stimulating factor on expansion and mobilization of facilitating cells and hematopoietic stem cells in mice: kinetics and repopulating potential. Blood 92, 3177–3188 (1998).

    Article  CAS  PubMed  Google Scholar 

  38. Tsapogas, P. et al. In vivo evidence for an instructive role of fms-like tyrosine kinase-3 (FLT3) ligand in hematopoietic development. Haematologica 99, 638–646 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Buza-Vidas, N. et al. FLT3 receptor and ligand are dispensable for maintenance and posttransplantation expansion of mouse hematopoietic stem cells. Blood 113, 3453–3460 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Waskow, C. et al. The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissues. Nat. Immunol. 9, 676–683 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Pietras, E. M. et al. Functionally distinct subsets of lineage-biased multipotent progenitors control blood production in normal and regenerative conditions. Cell Stem Cell 17, 35–46 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. D’Amico, A. & Wu, L. The early progenitors of mouse dendritic cells and plasmacytoid predendritic cells are within the bone marrow hemopoietic precursors expressing Flt3. J. Exp. Med. 198, 293–303 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Karsunky, H., Merad, M., Cozzio, A., Weissman, I. L. & Manz, M. G. Flt3 ligand regulates dendritic cell development from Flt3+ lymphoid and myeloid-committed progenitors to Flt3+ dendritic cells in vivo. J. Exp. Med. 198, 305–313 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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 

  45. Schlitzer, A. et al. Identification of cDC1- and cDC2-committed DC progenitors reveals early lineage priming at the common DC progenitor stage in the bone marrow. Nat. Immunol. 16, 718–728 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Jackson, J. T. et al. Id2 expression delineates differential checkpoints in the genetic program of CD8α+ and CD103+ dendritic cell lineages. EMBO J. 30, 2690–2704 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Bagadia, P. et al. An Nfil3Zeb2Id2 pathway imposes Irf8 enhancer switching during cDC1 development. Nat. Immunol. 20, 1174–1185 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kurotaki, D. et al. Epigenetic control of early dendritic cell lineage specification by the transcription factor IRF8 in mice. Blood 133, 1803–1813 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. 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 

  50. 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 

  51. Metcalf, D. Hematopoietic cytokines. Blood 111, 485–491 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Enver, T. & Jacobsen, S. E. W. Developmental biology: instructions writ in blood. Nature 461, 183–184 (2009).

    Article  CAS  PubMed  Google Scholar 

  53. Endele, M., Etzrodt, M. & Schroeder, T. Instruction of hematopoietic lineage choice by cytokine signaling. Exp. Cell. Res. 329, 207–213 (2014).

    Article  CAS  PubMed  Google Scholar 

  54. Jensen, C. T. et al. Permissive roles of hematopoietin and cytokine tyrosine kinase receptors in early T-cell development. Blood 111, 2083–2090 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Muenchow von, L. et al. Permissive roles of cytokines interleukin-7 and Flt3 ligand in mouse B-cell lineage commitment. Proc. Natl Acad. Sci. USA 113, E8122–E8130 (2016).

    Article  Google Scholar 

  56. Rieger, M. A., Hoppe, P. S., Smejkal, B. M., Eitelhuber, A. C. & Schroeder, T. Hematopoietic cytokines can instruct lineage choice. Science 325, 217–218 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Mossadegh-Keller, N. et al. M-CSF instructs myeloid lineage fate in single haematopoietic stem cells. Nature 497, 239–243 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Etzrodt, M. et al. Inflammatory signals directly instruct PU.1 in HSCs via TNF. Blood 133, 816–819 (2019).

    Article  CAS  PubMed  Google Scholar 

  59. Onai, N., Obata-Onai, A., Tussiwand, R., Lanzavecchia, A. & Manz, M. G. Activation of the Flt3 signal transduction cascade rescues and enhances type I interferon-producing and dendritic cell development. J. Exp. Med. 203, 227–238 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Buza-Vidas, N. et al. FLT3 expression initiates in fully multipotent mouse hematopoietic progenitor cells. Blood 118, 1544–1548 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Sproull, M., Kramp, T., Tandle, A., Shankavaram, U. & Camphausen, K. Multivariate analysis of radiation responsive proteins to predict radiation exposure in total-body irradiation and partial-body irradiation models. Radiat. Res. 187, 251–258 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. 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 

  63. Dai, Z. et al. edgeR: a versatile tool for the analysis of shRNA-seq and CRISPR–Cas9 genetic screens. F1000Res 3, 95 (2014).

    PubMed  PubMed Central  Google Scholar 

  64. Hashimshony, T. et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 17, 77 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Liao, Y., Smyth, G. K. & Shi, W. The Subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 41, e108 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Tian, L. et al. scPipe: a flexible R/Bioconductor preprocessing pipeline for single-cell RNA-sequencing data. PLoS Comput. Biol. 14, e1006361 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Haghverdi, L., Lun, A. T. L., Morgan, M. D. & Marioni, J. C. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nat. Biotechnol. 36, 421–427 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Walter and Eliza Hall Bioservices facility, FACS laboratory and S. Wilcox for technical support. We thank S. Heinzel, F. Vaillant and J. Visvader for providing critical reagents as well as technical and intellectual advice. We thank K. Shortman for insightful discussions and critical feedback on the paper. This work was supported by grants from the National Health and Medical Research Council (NHMRC), Australia (grant nos GNT1062820, GNT1100033, GNT1101378, GNT1124812 and GNT1145184), the Australia Research Council’s special initiative Stem Cells Australia, and through a funded research agreement with Gilead Inc. P.D.H is supported by an NHMRC fellowship. J.R. is supported by a Victorian Cancer Agency (VCA) grant (grant no. ECSG18020).

Author information

Authors and Affiliations

Authors

Contributions

D.S.L. designed and performed most experiments, did the analysis and wrote the manuscript. S. Tomei, D.A-Z., T.M.B., J.S., O.J.S., J.R. and A.P.N. assisted with experiments. L.T. and T.S.W. assisted with data analysis. N.D.H., A.P.N., S.L.N., S. Taoudi, M.E.R. and P.D.H. provided critical input into the experimental design and analysis. S.H.N. conceptualized and supervised the study and wrote the manuscript.

Corresponding author

Correspondence to Shalin H. Naik.

Ethics declarations

Competing interests

J.R. and N.D.H. are founders and shareholders of oNKo-Innate Pty. Ltd. This project was partly funded through a research agreement between Gilead Sciences, Inc. and S.H.N. The other authors declare no competing interests.

Additional information

Peer review information Nature Cell Biology thanks Simon Haas, Caetano Reis e Sousa, Samantha A. Morris and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Extended data

Extended Data Fig. 1 High levels of Flt3L promote cell division and DC generation in vitro.

a-d, CD11bcKit+Sca1+ HSPCs were labelled with CTV and were cultured (~ 1 × 103 cells per well) with RPMI supplemented with either high (2 μg/ml) or low (2 ng/ml) concentrations of Flt3L in vitro. Cells were serially sampled at each time point from each well (50% of contents) and analysed by flow cytometry daily from days 1-7. a. Changes in CTV intensity of total live cells over time. b. Stacked histogram showing percentage of cells in each division peak from a. n=3 wells per condition. Data shown are representative of two independent experiments. c. Flow cytometry plots comparing upregulation of CD11c or MHCII to CTV profiles on day 4. Numbers depict % of cells within the parent gate. d. Total inferred number of live cells (left) or DCs (right) in both conditions over time. FACS plots in (a, c) show representative plots of one well per condition. Histograms (b) show mean ± s.e.m. Scatter plot (d) show individual replicates where lines connect output from the same well serially sampled over time and numbers extrapolated from the number of prior sampling events; n=3 wells per condition. Data shown are representative of two independent experiments.

Extended Data Fig. 2 Flow cytometric plots of BM HSPC populations.

a. Gating strategy to isolate different HSPC populations from BM. Numbers depict percentage of cells from parent gate. b. Flow cytometry analysis of BM HSPC populations from mice treated with PBS or Flt3L for 5 days. This gating strategy is similar to a, except the exclusion of Flt3 antibody staining, as Flt3 is poorly detected in cells treated with Flt3L. Numbers shown in all FACS plots represent percentage of cells from parent gate. Cell numbers from each population are shown in Fig. 1f.

Extended Data Fig. 3 Gating strategy to isolate mature progeny populations from the spleen.

a. General workflow to fractionate total splenocytes into DC, myeloid, lymphoid/erythroid fractions. Note: red cell lysis was excluded if erythroid lineage was included in analysis. b. General pre-gating strategy to define counting beads and live cells from each fraction. Beads were added to all fractions after enrichment (before final staining) and gated as FSCloSSChi to allow estimation of cell numbers. c. Gating strategy to isolate cDC1s, cDC2s and pDCs from CD11c and Siglec-H - enriched fraction. d. Gating strategy to isolate eosinophils (eos), monocytes (mon) and neutrophils (neu) from CD11b-enrich fraction. e. Gating strategy to isolate B cells and erythroid blast (EryB) from final flow through fraction. f. Example plot showing gating strategy to identify donor-derived cells from each defined population. Numbers shown in all FACS plots represent percentage of cells from parent gate.

Extended Data Fig. 4 Clonal emergency DC development in different barcoding experimental settings.

(a-c) Standard setting as in Figs. 3 and 4 (day -3; 5 Gy irradiation; i.v. transplantation), with analysis of erythroid lineage. n = 5 mice per treatment, pooled from two independent experiments. (d-f) Intra-BM (not i.v.) transplantation, same irradiation regimen (day -3; 5 Gy), with analysis of erythroid lineage. n = 6 mice per treatment, pooled from two independent experiments. (g-i) Lorw dose irradiation (day -3; 2 Gy; i.v.). n = 5 PBS-treated mice and n = 6 Flt3L-treated mice, pooled from two independent experiments. (a, b, d, e, g & h) Fold change in number of cells (a, d & g) or barcodes (b, e & h) from each cell type produced by Flt3L-treated mice compared to the average of PBS-treated mice from the same experiment; mean ± s.e.m., P-values from two-tailed unpaired t-tests. (c, f & i) Heatmaps of all barcodes from the corresponding experiments. (e, f, h & i) Note that barcode data filtering was not applied due to low read counts and/or low correlation between technical replicates in a number of samples in these experimental settings.

Source data

Extended Data Fig. 5 Distribution of clone size from each fate cluster.

Violin plots showing the distribution of number of cells produced per clone (clone size) from all PBS- or Flt3L-treat clones in each fate cluster, from cluster 5-17. Results for cluster 1-4 is shown in Fig. 4f. Average fold change in clone size (Flt3L/PBS) and P-values from two-tailed unpaired t-tests are shown for each pair. See Source Data Extended Fig. 5 for details.

Source data

Extended Data Fig. 6 Flow-cytometric analysis of cells from Divi-Seq experiments.

a, Gating strategy used during index sorting of single donor-derived cells for Divi-Seq experiments in Fig. 4. As donor cells were CTV-labelled prior to transplantation, CTV+ cells were sorted. b, Gating strategy used to remove potential dead (based on FSC, SSC and PI) and contaminated endogenous cells (CD45.1lowCD45.2+) after sorting. c. Flow-cytometric analysis of HSPC populations from the endogenous or donor-derived compartment, numbers depict percentage of cells from the endogenous or donor-derived compartment, respectively. d. Estimated cell number of each phenotypically defined HSPC population from the donor-derived compartment. e. estimated number of cells in each cluster from BM (tibia, femur and ilium; donor-derived) of each mouse. from the second Divi-Seq experiment. (d-e) n = 3 mice per treatment from the second Divi-Seq experiment; mean ± s.e.m., P-values from two-tailed unpaired t-tests with Holm-Sidak correction for multiple comparison.

Source data

Extended Data Fig. 7 Assessing similarity to known cell types using SingleR.

A heatmap showing similarity scores to known cell types (compared to Immgen database) of each single cell computed by SingleR, with the following annotation per cell: 1) Cluster ID identified by Seurat as in Fig. 4c; 2) Cell type labels identified by SingleR; 3) PBS or Flt3L treatment. Note: Information regarding expression of surface markers and cluster marker genes in Fig. 4c and Supplementary Table 1 is sufficient for accurate cell type annotation of Seurat clusters, independent of SingleR analysis. SingleR analysis is performed and shown to further validate the annotation.

Extended Data Fig. 8 Gene expression of cells from different cluster in Divi-Seq.

Violin plots showing gene expression of cells from different clusters. Each dot represents a cell. Colour depicts PBS or Flt3L treatment. Large black dot indicates mean expression. See Source Data Extended Fig. 8 for details.

Source data

Supplementary information

Reporting Summary

Peer Review Information

Supplementary Tables

Supplementary Table 1. List of marker genes for clusters defined by Seurat. Cluster marker genes were identified using the FindAllMarker function in Seurat via differential expression analysis, which used non-parameteric Wilcoxon’s rank-sum test to compute P values. Adjustments for multiple testing were made and are presented as p_val_adj. Cluster 0, HSC/MPP-like; Cluster 1, lymphoid progenitor-like; Cluster 2, cDC-like; Cluster 3, myeloid progenitor-like; Cluster 4, monocyte-like; Cluster 5, pDC-like and Cluster 6, neutrophil-like. Supplementary Table 2. List of antibodies used.

Source data

Source Data Fig. 1

Statistical source data

Source Data Fig. 2

Statistical source data

Source Data Fig. 3

Statistical source data

Source Data Fig. 4

Statistical source data

Source Data Fig. 6

Statistical source data

Source Data Fig. 7

Statistical source data

Source Data Fig. 8

Statistical source data

Source Data Extended Data Fig. 4

Statistical source data

Source Data Extended Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 8

Statistical source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, D.S., Tian, L., Tomei, S. et al. Single-cell analyses reveal the clonal and molecular aetiology of Flt3L-induced emergency dendritic cell development. Nat Cell Biol 23, 219–231 (2021). https://doi.org/10.1038/s41556-021-00636-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-021-00636-7

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