Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The activation trajectory of plasmacytoid dendritic cells in vivo during a viral infection

Abstract

Plasmacytoid dendritic cells (pDCs) are a major source of type I interferon (IFN-I). What other functions pDCs exert in vivo during viral infections is controversial, and more studies are needed to understand their orchestration. In the present study, we characterize in depth and link pDC activation states in animals infected by mouse cytomegalovirus by combining Ifnb1 reporter mice with flow cytometry, single-cell RNA sequencing, confocal microscopy and a cognate CD4 T cell activation assay. We show that IFN-I production and T cell activation were performed by the same pDC, but these occurred sequentially in time and in different micro-anatomical locations. In addition, we show that pDC commitment to IFN-I production was marked early on by their downregulation of leukemia inhibitory factor receptor and was promoted by cell-intrinsic tumor necrosis factor signaling. We propose a new model for how individual pDCs are endowed to exert different functions in vivo during a viral infection, in a manner tightly orchestrated in time and space.

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: Bulk transcriptional profiling suggests the induction of distinct pDC activation states in vivo during MCMV infection.
Fig. 2: ScRNA-seq analysis of pDCs from 36-h MCMV-infected mice confirms their heterogeneity and pinpoints LIFR downregulation as a selective marker of IFN-I-producing pDCs.
Fig. 3: ScRNA-seq analysis identifies seven different pDC activation states in vivo during MCMV infection.
Fig. 4: Inference of the pDC activation trajectory during MCMV infection.
Fig. 5: Transcriptional convergence of pDCs toward tDCs over pseudo-time.
Fig. 6: Identification of annotated gene modules regulated along pseudo-time during pDC activation.
Fig. 7: Cell-intrinsic TNF signaling promotes pDC IFN-I production.
Fig. 8: Characterization of the phenotype, function and micro-anatomical location of pDC activation states.

Similar content being viewed by others

Data availability

Microarray and scRNA-seq data have been deposited in the Gene Expression Omnibus (GEO) repository under accession codes GSE150664 and GSE151248, respectively. All the other data that support the findings of the present study are available from the corresponding authors upon reasonable request.

References

  1. Tomasello, E., Pollet, E., Vu Manh, T. P., Uze, G. & Dalod, M. Harnessing mechanistic knowledge on beneficial versus deleterious IFN-I effects to design innovative immunotherapies targeting cytokine activity to specific cell types. Front. Immunol. 5, 526 (2014).

    PubMed  PubMed Central  Google Scholar 

  2. Asselin-Paturel, C. et al. Mouse type I IFN-producing cells are immature APCs with plasmacytoid morphology. Nat. Immunol. 2, 1144–1150 (2001).

    CAS  PubMed  Google Scholar 

  3. Dalod, M. et al. Interferon α/β and interleukin 12 responses to viral infections: pathways regulating dendritic cell cytokine expression in vivo. J. Exp. Med. 195, 517–528 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Kadowaki, N., Antonenko, S., Lau, J. Y. & Liu, Y. J. Natural interferon α/β–producing cells link innate and adaptive immunity. J. Exp. Med. 192, 219–226 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Grouard, G. et al. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J. Exp. Med. 185, 1101–1111 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Dalod, M. et al. Dendritic cell responses to early murine cytomegalovirus infection: subset functional specialization and differential regulation by interferon α/β. J. Exp. Med. 197, 885–898 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. O’Keeffe, M. et al. Mouse plasmacytoid cells: long-lived cells, heterogeneous in surface phenotype and function, that differentiate into CD8+ dendritic cells only after microbial stimulus. J. Exp. Med. 196, 1307–1319 (2002).

    PubMed  PubMed Central  Google Scholar 

  8. Segura, E., Wong, J. & Villadangos, J. A. Cutting edge: B220+CCR9 dendritic cells are not plasmacytoid dendritic cells but are precursors of conventional dendritic cells. J. Immunol. 183, 1514–1517 (2009).

    CAS  PubMed  Google Scholar 

  9. See, P. et al. Mapping the human DC lineage through the integration of high-dimensional techniques. Science 356, eaag3009 (2017).

    PubMed  PubMed Central  Google Scholar 

  10. Dress, R. J. et al. Plasmacytoid dendritic cells develop from Ly6D+ lymphoid progenitors distinct from the myeloid lineage. Nat. Immunol. 20, 852–864 (2019).

    CAS  PubMed  Google Scholar 

  11. Rodrigues, P. F. et al. Distinct progenitor lineages contribute to the heterogeneity of plasmacytoid dendritic cells. Nat. Immunol. 19, 711–722 (2018).

    CAS  PubMed  Google Scholar 

  12. Bar-On, L. et al. CX3CR1+ CD8α+ dendritic cells are a steady-state population related to plasmacytoid dendritic cells. Proc. Natl Acad. Sci. USA 107, 14745–14750 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Lau, C. M. et al. Leukemia-associated activating mutation of Flt3 expands dendritic cells and alters T cell responses. J. Exp. Med. 213, 415–431 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Villani, A. C. et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356, eaah4573 (2017).

  15. Leylek, R. et al. Integrated cross-species analysis identifies a conserved transitional dendritic cell population. Cell Rep. 29, 3736–3750.e8 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Alcantara-Hernandez, M. et al. High-dimensional phenotypic mapping of human dendritic cells reveals interindividual variation and tissue specialization. Immunity 47, 1037–1050.e6 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Alculumbre, S. G. et al. Diversification of human plasmacytoid predendritic cells in response to a single stimulus. Nat. Immunol. 19, 63–75 (2018).

    CAS  PubMed  Google Scholar 

  18. Swiecki, M. & Colonna, M. The multifaceted biology of plasmacytoid dendritic cells. Nat. Rev. Immunol. 15, 471–485 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Reizis, B. Plasmacytoid dendritic cells: development, regulation, and function. Immunity 50, 37–50 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Leylek, R. & Idoyaga, J. The versatile plasmacytoid dendritic cell: function, heterogeneity, and plasticity. Int Rev. Cell Mol. Biol. 349, 177–211 (2019).

    PubMed  Google Scholar 

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

    PubMed  Google Scholar 

  22. Tomasello, E. et al. Molecular dissection of plasmacytoid dendritic cell activation in vivo during a viral infection. EMBO J. 37, e98836 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Assil, S. et al. Plasmacytoid Dendritic Cells and Infected Cells Form an Interferogenic Synapse Required for Antiviral Responses. Cell Host Microbe 25, 730–745.e6 (2019).

    CAS  PubMed  Google Scholar 

  24. Frenz, T. et al. Independent of plasmacytoid dendritic cell (pDC) infection, pDC triggered by virus-infected cells mount enhanced type I IFN responses of different composition as opposed to pDC stimulated with free virus. J. Immunol. 193, 2496–2503 (2014).

    CAS  PubMed  Google Scholar 

  25. Scheu, S., Dresing, P. & Locksley, R. M. Visualization of IFNβ production by plasmacytoid versus conventional dendritic cells under specific stimulation conditions in vivo. Proc. Natl Acad. Sci. USA 105, 20416–20421 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. O’Keeffe, M. et al. Nonplasmacytoid, high IFN-α–producing, bone marrow dendritic cells. J. Immunol. 188, 3774–3783 (2012).

    PubMed  Google Scholar 

  27. Zucchini, N. et al. Individual plasmacytoid dendritic cells are major contributors to the production of multiple innate cytokines in an organ-specific manner during viral infection. Int. Immunol. 20, 45–56 (2008).

    CAS  PubMed  Google Scholar 

  28. Hentschel, E. et al. Destabilized eYFP variants for dynamic gene expression studies in Corynebacterium glutamicum. Micro. Biotechnol. 6, 196–201 (2013).

    Google Scholar 

  29. Tombolini, R., Unge, A., Davey, M. E., deBruijn, F. J. & Jansson, J. K. Flow cytometric and microscopic analysis of GFP-tagged Pseudomonas fluorescens bacteria. FEMS Microbiol. Ecol. 22, 17–28 (1997).

    CAS  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    CAS  PubMed  Google Scholar 

  32. Ito, T., Kanzler, H., Duramad, O., Cao, W. & Liu, Y. J. Specialization, kinetics, and repertoire of type 1 interferon responses by human plasmacytoid predendritic cells. Blood 107, 2423–2431 (2006).

    CAS  PubMed  Google Scholar 

  33. Ciancanelli, M. J. et al. Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency. Science 348, 448–453 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Honda, K. et al. IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature 434, 772–777 (2005).

    CAS  PubMed  Google Scholar 

  35. Levy, D. E. Whence interferon? Variety in the production of interferon in response to viral infection. J. Exp. Med. 195, F15–F18 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Robbins, S. H. et al. Novel insights into the relationships between dendritic cell subsets in human and mouse revealed by genome-wide expression profiling. Genome Biol. 9, R17 (2008).

    PubMed  PubMed Central  Google Scholar 

  37. Shi, W. et al. Transcriptional profiling of mouse B cell terminal differentiation defines a signature for antibody-secreting plasma cells. Nat. Immunol. 16, 663–673 (2015).

    CAS  PubMed  Google Scholar 

  38. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    PubMed  PubMed Central  Google Scholar 

  39. Zhang, J. et al. A newly identified microRNA, mmu-miR-7578, functions as a negative regulator on inflammatory cytokines tumor necrosis factor-α and interleukin-6 via targeting Egr1 in vivo. J. Biol. Chem. 288, 4310–4320 (2013).

    CAS  PubMed  Google Scholar 

  40. Guha, M. et al. Lipopolysaccharide activation of the MEK-ERK1/2 pathway in human monocytic cells mediates tissue factor and tumor necrosis factor α expression by inducing Elk-1 phosphorylation and Egr-1 expression. Blood 98, 1429–1439 (2001).

    CAS  PubMed  Google Scholar 

  41. Attaf, N. et al. FB5P-seq: FACS-based 5-prime end single-cell RNA-seq for integrative analysis of transcriptome and antigen receptor repertoire in B and T cells. Front. Immunol. 11, 216 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Spinelli, L., Carpentier, S., Montanana Sanchis, F., Dalod, M. & Vu Manh, T. P. BubbleGUM: automatic extraction of phenotype molecular signatures and comprehensive visualization of multiple Gene Set Enrichment Analyses. BMC Genomics 16, 814 (2015).

    PubMed  PubMed Central  Google Scholar 

  43. Palucka, A. K., Blanck, J. P., Bennett, L., Pascual, V. & Banchereau, J. Cross-regulation of TNF and IFN-α in autoimmune diseases. Proc. Natl Acad. Sci. USA 102, 3372–3377 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Jaehn, P. S., Zaenker, K. S., Schmitz, J. & Dzionek, A. Functional dichotomy of plasmacytoid dendritic cells: antigen-specific activation of T cells versus production of type I interferon. Eur. J. Immunol. 38, 1822–1832 (2008).

    CAS  PubMed  Google Scholar 

  45. Reizis, B., Bunin, A., Ghosh, H. S., Lewis, K. L. & Sisirak, V. Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev. Immunol. 29, 163–183 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Wimmers, F. et al. Single-cell analysis reveals that stochasticity and paracrine signaling control interferon-alpha production by plasmacytoid dendritic cells. Nat. Commun. 9, 3317 (2018).

    PubMed  PubMed Central  Google Scholar 

  47. Bauer, J. et al. Cutting edge: IFN-β expression in the spleen is restricted to a subpopulation of plasmacytoid dendritic cells exhibiting a specific immune modulatory transcriptome signature. J. Immunol. 196, 4447–4451 (2016).

    CAS  PubMed  Google Scholar 

  48. Sesti-Costa, R. et al. Leukemia inhibitory factor inhibits plasmacytoid dendritic cell function and development. J. Immunol. 204, ji1900604 (2020).

  49. Swiecki, M. et al. Type I interferon negatively controls plasmacytoid dendritic cell numbers in vivo. J. Exp. Med. 208, 2367–2374 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Ghosh, H. S., Cisse, B., Bunin, A., Lewis, K. L. & Reizis, B. Continuous expression of the transcription factor E2-2 maintains the cell fate of mature plasmacytoid dendritic cells. Immunity 33, 905–916 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. 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).

    CAS  PubMed  Google Scholar 

  52. Baranek, T. et al. Differential responses of immune cells to type I interferon contribute to host resistance to viral infection. Cell Host Microbe 12, 571–584 (2012).

    CAS  PubMed  Google Scholar 

  53. Villani, A. C. & Shekhar, K. Single-cell RNA sequencing of human T cells. Methods Mol. Biol. 1514, 203–239 (2017).

    CAS  PubMed  Google Scholar 

  54. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  55. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    CAS  PubMed  Google Scholar 

  56. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Subramanian, A., Kuehn, H., Gould, J., Tamayo, P. & Mesirov, J. P. GSEA-P: a desktop application for Gene Set Enrichment Analysis. Bioinformatics 23, 3251–3253 (2007).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all the staff of the CIML and CIPHE mouse houses for their assistance, as well as the staff of the CIML flow cytometry and imaging (ImagImm) core facilities. We thank L. Spinelli for fruitful discussions on single-cell data analysis and the CIML genomics and bioinformatics platform for their technical and methodological help. Microarray experiments and sequencing for the SS2 datasets were performed by V. Alunni or B. Jost in the GenomEast platform (Strasbourg, France), a member of the France Génomique consortium (ANR-10-INBS-0009), managed by C. Thibault-Carpentier. FB5P-seq library sequencing was performed by HalioDx testing laboratory, Marseille, France. We thank S. Henri (CIML) for the generous gift of OT-II mice and H. Luche for advice on cell and tissue fixation to preserve the YFP signal. This research was funded by grants from the European Research Council under the European Community’s Seventh Framework Programme (FP7/2007-2013, grant no. 281225, SystemsDendritic, to M.D.), the Agence Nationale de la Recherche (ANR) (SCAPIN, grant no. ANR-15-CE15-0006-01, to M.D.), and the Fondation pour la Recherche Médicale (grant no. DEQ20180339172, Equipe Labellisée, to M.D.). We also thank the DCBIOL Labex (ANR-11-LABEX-0043, grant no. ANR-10-IDEX-0001-02 PSL*), the A*MIDEX project (grant no. ANR-11-IDEX-0001-02) funded by the French government’s Investissements d’Avenir program managed by the ANR, and institutional support from CNRS, INSERM, Aix-Marseille Université and Marseille Immunopole. This work was supported by the French National Research Agency through the Investments for the Future program (France-BioImaging, ANR-10-INBS-04). A.A. and R.C. were supported by the DCBIOL Labex. G. Brelurut’s apprenticeship was supported by INSERM.

Author information

Authors and Affiliations

Authors

Contributions

Studies were designed by E.T., M.D., A.A. and T.-P.V.M., with help from A.-C.V and P.M. Experiments were performed by A.A., E.T., M.V., N.C. and N.A., with help from K.N. and G. Bessou. Data were analyzed by A.A., T.-P.V.M., M.V., N.C., M.D. and E.T., with help from C.D., R.C., G. Brelurut, I.C.-M., M.T.-C. and D.T. Critical reagents and advice were provided by B.R. and J.-L.D. The manuscript was written by A.A., T.-P.V.-M., M.D. and E.T. All authors contributed to discussions and editing of the manuscript.

Corresponding authors

Correspondence to Marc Dalod or Elena Tomasello.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Jamie D. K. Wilson and Ioana Visan were the primary editors on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Validation of the Ifnb1Eyfp reporter mice to track IFN-I-producing pDCs during MCMV infection.

a, IFN-α/β production by pDCs (blue), pDC-like cells (red) and cDCs (black) at 36 h after MCMV infection of Zbtb46GFP reporter mice. Overlaid histograms (bottom right) show CD11c and CD11b expression for the three cell types. The data shown are from one mouse representative of 3 animals from 2 independent experiments. b, YFP expression in pDCs is stable during the biological process examined. Ifnb1EYFP CD45.2+ mice were infected by MCMV. 36 h later, LIFRlo EYFP- or LIFRlo EYFP+ pDCs were sorted by flow cytometry and cultured in vitro for 8 h in CD45.1+ feeder FLT3-L bone marrow cultures. YFP expression was monitored by flow cytometry before and after the culture as indicated. The data shown are from one experiment representative of two independent ones. c–g Around 80% of splenic YFP+ cells are bona fide pDCs at all time points examined during MCMV infection. c, cDCs and pDCs were identified following the gating strategy shown. The analysis was performed after selection of live cells and exclusion of Lin+ cells. d, YFP expression was analyzed in indicated splenic DC populations, isolated from 44 h MCMV-infected Ifnb1Eyfp mice. e, Ccr9 expression was analyzed in splenic cDC1s (red), cDC2s (green) and pDCs (blue) isolated from 36 h (left), 44 h (middle) or 48 h (right) MCMV-infected Ifnb1Eyfp mice. f, Autofluorescence-YFP+ cells were gated in live splenocytes isolated from 44 h MCMV-infected Ifnb1Eyfp mice. The proportions of DCs vs non-DCs (others, grey) in YFP+ cells were analyzed according to the gating strategy shown in (c). g, Summary of the results obtained following the strategy shown in (e) at 36 h (left), 44 h (middle) and 48 h (right) post-infection. For each time point, data (mean ± s.e.m.) are shown for 5 mice from one experiment.

Extended Data Fig. 2 Design and quality control of the SS2 dataset#2.

a, Flow cytometry gating and overall strategy for index sorting of pDCs from one uninfected (UN, top panel) and one 36 h MCMV-infected (bottom panel) Ifnb1Eyfp mice, using LIFR and BST2 expression levels to enrich IFN-IEyfp+ pDCs, for scRNAseq. Numbers in parentheses indicate the total number of cells sorted in each gate. b, t-SNE and cell clustering analysis for the 323 cells that passed quality controls. c, Sorting phenotype projection on the t-SNE space. d, LIFR expression projection on the t-SNE space. Data are expressed as inverse hyperbolic arcsine (asinh) of fluorescence intensity. e, BubbleMap illustrating GSEA results for 8 selected gene sets (columns) in pairwise comparisons between the cell clusters (rows) identified in (b). ND, not determined. f, Heatmap (left) showing mRNA expression levels of representative genes (rows) across the cell clusters (columns) identified in (b). Most of the genes shown were selected due to their contribution to the GSEA results from (e), as informed by the grid on the right of the heatmap where filled cells mean belonging of the gene (row) to a gene set (column). The gene set order and color code on the top of the grid is the same as in panel e. The far right column of the grid (*) corresponds to genes selectively expressed to high levels in plasmocytes. g, Normalized expression of Ccl3 vs the IFN-I meta-gene along pseudo-time. h, Projection on the UMAP space of the predicted induction (red) vs termination (blue) of Ccl3 transcription.

Extended Data Fig. 3 Kinetics of IL-12 production by pDCs during MCMV infection.

a, Frequency (mean ± s.e.m.) of YFP+, IFN+ and IL-12+ pDCs isolated from Ifnb1Eyfp mice at indicated time points after MCMV infection. b, Data from individual animals for the frequencies of IL-12+ and YFP+ cells in pDCs isolated from Ifnb1Eyfp mice at indicated time points, with overlay of mean ± s.e.m. c, Flow cytometry dot plots showing IL-12 vs YFP expression in pDCs isolated from one representative Ifnb1Eyfp mouse for each time point. The data from all panels were analyzed from the same experiments, with 5 mice at 0 h, 7 at 33 h, 10 at 36 h, 5 at 40 h, 3 at 44 h and 3 at 48 h, from one experiment for 44 h and 48 h, or pooled from 2 (resp. 3) independent experiments for 33 h and 44 h (resp. 36 h).

Extended Data Fig. 4 LIFR downregulation enables enrichment from WT C57BL/6 mice of the pDCs engaged in IFN-I.

pDCs were sorted from 36 h MCMV-infected WT C57BL/6 mice, with a protocol including an enrichment of LIFRlo cells to increase the capture efficiency for pDCs engaged in IFN-I production. a, Monocle pseudo-temporal analysis showing bifurcation of the inferred pDC activation trajectory in two major branches, Y53 and Y50. b, Expression of the IFN-I meta-gene along pseudo-time for the Y53 (top) and Y50 (bottom) branches of the pDC activation trajectory. c, LIFR expression along pseudo-time on the pDCs from the common root (empty thin orange circles), Y53 branch (filled orange triangles) and Y50 branch (empty thick dark red circles) of the pDC activation trajectory. d, Expression of individual genes along pseudo-time for the cells from the common root (empty thin orange circles), Y53 branch (filled orange triangles) and Y50 branch (empty thick dark red circles) of the pDC activation trajectory. A polynomial curve was fit to the data for each of the three segments of the trajectory.

Extended Data Fig. 5 Design, quality controls and RNA velocity analysis of the FB5P kinetics dataset.

a, Experimental design. For each time point, splenocytes were isolated from one Ifnb1Eyfp mouse, depleted of Lin+ cells by magnetic sorting and used for index sorting of pDCs using three sorting gates: i) total (bulk) pDCs, ii) LIFRlo pDCs irrespective of their YFP expression, and iii) YFP+ pDCs. FB5P-seq scRNAseq libraries were then prepared. b, Table indicating the total numbers of cells sorted for each time point and sorting gate, the numbers of cells that passed quality control upon data analysis, and the number of bona fide pDC ultimately kept after identification and removal of contaminating cell types. c,d, Identification and removal of contaminating B and pDC-like cells. c, UMAP and clustering analysis. The analysis of the genes differentially expressed across clusters combined with their mining for expression across immune cell types by using the MyGeneSet tool of Immgen enabled identification of contaminating B cells (cluster 7, highlighted in orange). A GSEA analysis performed by using BubbleGUM (not shown) enabled identification of contaminating pDC-like cells (cluster 11, highlighted in red). d, Projection on the UMAP space of the expression of two B cell-specific genes, Jchain and Iglv1, two pDC-like cell-specific genes, Ms4a6b and Vim, and 2 genes selectively expressed at high levels in pDCs, Klk1 and Ly6d. e, Projections of the velocity vector of each pDC in the UMAP space obtained after contaminant removal.

Extended Data Fig. 6 Ex vivo unidirectional transition of pDCs isolated from MCMV-infected mice from a YFP+CCR7- to a YFP+CCR7+ activation state.

Ifnb1Eyfp CD45.2+ mice were infected by MCMV. 36 h later, LIFRlo YFP+ CCR7- pDCs and LIFRlo YFP+ CCR7+ pDCs were sorted and cultured in vitro for 8 h in CD45.1+ feeder FLT3-L bone marrow cultures. CCR7 expression was monitored by flow cytometry before and after the culture, as depicted in the right panel. The data shown are from one experiment representative of two independent ones.

Extended Data Fig. 7 Micro-anatomical locations of splenic YFP+ cells during MCMV infection of Ifnb1Eyfp mice.

a, b, c, Number of IFN-α/β spots per mm2 (a), of YFP+IFN-α/β+ cells/mm2 (b), and of YFP+ cells per mm2 (c), in whole spleen sections from Ifnb1Eyfp mice at indicated time points after MCMV infection. d, Number of YFP+ cells per mm2 residing in the different spleen zones. MZ, marginal zone; RP, red pulp; WP, white pulp. Fifteen individual data points are shown on each graph for each time point, corresponding to quantitation of 5 different whole spleen sections per mouse, from 3 different mice, with overlay of mean±s.e.m.

Extended Data Fig. 8 Proposed model of the spatiotemporal dynamics of splenic pDC activation and functions during MCMV infection.

MCMV initially targets and replicates in stromal cells and/or metallophilic macrophages in the marginal zone (❶). These infected cells may then upregulate their expression of ICAM-1 and express mTNF, leading to their specific recognition by, and interactions with, quiescent pDCs. This interaction is proposed to downregulate LIFR expression on pDCs, to induce low levels of Tnf and Ccl3 in pDCs. It may also lead to the generation of an interferogenic synapse (❷) promoting local targeted delivery of viral material from the infected cell to pDCs, as illustrated on the upper left detailed drawing enlarged from the corresponding delimited area in the main drawing. This viral material is engulfed in pDCs and routed into dedicated endosomes, allowing TLR9 triggering, with the downstream enforcement of Tnf and Ccl3 expression and the induction of IFN-I genes. At this early activation state, pDCs from Ifnb1Eyfp reporter mice already start to express IFN-I but not yet clearly detectable levels of YFP. Then, pDCs further enhance their expression of IFN-I, leading to their expression of high levels of YFP in Ifnb1Eyfp reporter mice, and they simultaneously start to express IL-12 (❸). After termination of their IFN-I production, pDCs further enhance their IL-12 production, acquire CCR7 expression and migrate from the marginal zone to the white pulp through bridging channels (❹). Ultimately, pDCs relocate to the T cell zone where they harbor clear features of mature DCs, with a transcriptional, morphologic and functional convergence with tDCs, presumably including the acquisition of a dendritic morphology upon expression of Fscn1 and other genes involved into cytoskeleton remodeling, and acquisition of the ability to prime naïve CD4+ T cells (❺). CD169+ MMM, marginal zone metallophilic macrophages; mTNF, plasma membrane-bound TNF; pIRF7, phosphorylated IRF7.

Supplementary information

Supplementary Information

Supplementary Tables 1–3; Supplementary text corresponding to the sequence of the Eyfp gene in FASTA format.

Reporting summary

Supplementary Data 1

Pipeline and parameters used for the computational analysis of scRNA-seq data.

Supplementary Data 2

Source and gene content of the gene sets used for GSEA.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Abbas, A., Vu Manh, TP., Valente, M. et al. The activation trajectory of plasmacytoid dendritic cells in vivo during a viral infection. Nat Immunol 21, 983–997 (2020). https://doi.org/10.1038/s41590-020-0731-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0731-4

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