Abstract
Plasmacytoid dendritic cells (pDCs) mount powerful antiviral type I interferon (IFN-I) responses, yet only a fraction of pDCs produces high levels of IFN-I. Here we report that peripheral pDCs in naive mice comprise three subsets (termed A, B and C) that represent progressive differentiation stages. This heterogeneity was generated by tonic IFN-I signaling elicited in part by the cGAS/STING and TLR9 DNA-sensing pathways. A small ‘IFN-I-naive’ subset (pDC-A) could give rise to other subsets; it was expanded in STING deficiency or after the IFN-I receptor blockade, but was abolished by exogenous IFN-I. In response to RNA viruses, pDC-A showed increased Bcl2-dependent survival and superior IFN-I responses, but was susceptible to virus infection. Conversely, the majority of pDCs comprised the ‘IFN-I-primed’ subsets (pDC-B/C) that showed lower IFN-I responses and poor survival, but did not support virus replication. Thus, tonic IFN-I signaling decreases the cytokine-producing capacity and survival of pDCs but increases their virus resistance, facilitating optimal antiviral responses.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout








Data availability
All sequencing data have been deposited in the NCBI Gene Expression Omnibus database under accession number GSE252191. All other data are presented in the paper or are available from the corresponding authors upon request. Source data are provided with this paper.
References
Mesev, E. V., LeDesma, R. A. & Ploss, A. Decoding type I and III interferon signalling during viral infection. Nat. Microbiol. 4, 914–924 (2019).
Motwani, M., Pesiridis, S. & Fitzgerald, K. A. DNA sensing by the cGAS-STING pathway in health and disease. Nat. Rev. Genet. 20, 657–674 (2019).
Adams, N. M., Das, A., Yun, T. J. & Reizis, B. Ontogeny and function of plasmacytoid dendritic cells. Annu. Rev. Immunol. 42, 347–373 (2024).
Swiecki, M., Gilfillan, S., Vermi, W., Wang, Y. & Colonna, M. Plasmacytoid dendritic cell ablation impacts early interferon responses and antiviral NK and CD8+ T cell accrual. Immunity 33, 955–966 (2010).
Cervantes-Barragan, L. et al. Control of coronavirus infection through plasmacytoid dendritic-cell-derived type I interferon. Blood 109, 1131–1137 (2007).
Cervantes-Barragan, L. et al. Plasmacytoid dendritic cells control T-cell response to chronic viral infection. Proc. Natl Acad. Sci. USA 109, 3012–3017 (2012).
Cisse, B. et al. Transcription factor E2-2 is an essential and specific regulator of plasmacytoid dendritic cell development. Cell 135, 37–48 (2008).
Valente, M. et al. Novel mouse models based on intersectional genetics to identify and characterize plasmacytoid dendritic cells. Nat. Immunol. 24, 714–728 (2023).
Leylek, R. et al. Integrated cross-species analysis identifies a conserved transitional dendritic cell population. Cell Rep. 29, 3736–3750 (2019).
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).
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).
Brouiller, F. et al. Single-cell RNA-seq analysis reveals dual sensing of HIV-1 in blood Axl+ dendritic cells. iScience 26, 106019 (2023).
Krug, A. et al. Identification of CpG oligonucleotide sequences with high induction of IFN-ɑ/β in plasmacytoid dendritic cells. Eur. J. Immunol. 31, 2154–2163 (2001).
Yun, T. J. et al. Human plasmacytoid dendritic cells mount a distinct antiviral response to virus-infected cells. Sci. Immunol. 6, eabc7302 (2021).
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).
Kumagai, Y. et al. Alveolar macrophages are the primary interferon-ɑ producer in pulmonary infection with RNA viruses. Immunity 27, 240–252 (2007).
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).
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).
Tomasello, E. et al. Molecular dissection of plasmacytoid dendritic cell activation in vivo during a viral infection. EMBO J. 37, e98836 (2018).
Abbas, A. et al. The activation trajectory of plasmacytoid dendritic cells in vivo during a viral infection. Nat. Immunol. 21, 983–997 (2020).
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).
Bjorck, P., Leong, H. X. & Engleman, E. G. Plasmacytoid dendritic cell dichotomy: identification of IFN-ɑ producing cells as a phenotypically and functionally distinct subset. J. Immunol. 186, 1477–1485 (2011).
Niederquell, M. et al. Sca-1 expression defines developmental stages of mouse pDCs that show functional heterogeneity in the endosomal but not lysosomal TLR9 response. Eur. J. Immunol. 43, 2993–3005 (2013).
Swiecki, M. et al. Microbiota induces tonic CCL2 systemic levels that control pDC trafficking in steady state. Mucosal Immunol. 10, 936–945 (2017).
Matsui, T. et al. CD2 distinguishes two subsets of human plasmacytoid dendritic cells with distinct phenotype and functions. J. Immunol. 182, 6815–6823 (2009).
Ghanem, M. H. et al. Proteomic and single-cell transcriptomic dissection of human plasmacytoid dendritic cell response to influenza virus. Front. Immunol. 13, 814627 (2022).
Du, Y. et al. Altered X-chromosome inactivation of the TLR7/8 locus and heterogeneity of pDCs in systemic sclerosis. J. Exp. Med. 222, e20231809 (2025).
Silvin, A. et al. Constitutive resistance to viral infection in human CD141+ dendritic cells. Sci. Immunol. 2, eaai8071 (2017).
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).
Rodrigues, P. F. et al. Distinct progenitor lineages contribute to the heterogeneity of plasmacytoid dendritic cells. Nat. Immunol. 19, 711–722 (2018).
Dress, R. J. et al. Plasmacytoid dendritic cells develop from Ly6D+ lymphoid progenitors distinct from the myeloid lineage. Nat. Immunol. 20, 852–864 (2019).
Feng, J. et al. Clonal lineage tracing reveals shared origin of conventional and plasmacytoid dendritic cells. Immunity 55, 405–422 (2022).
Parekh, N. J., Winship, D., Van Dis, E. & Stetson, D. B. Regulation and dynamics of IFN-β expression revealed with a knockin reporter mouse. J. Immunol. 213, 1858–1868 (2024).
Lind, N. A., Rael, V. E., Pestal, K., Liu, B. & Barton, G. M. Regulation of the nucleic acid-sensing Toll-like receptors. Nat. Rev. Immunol. 22, 224–235 (2022).
Schaupp, L. et al. Microbiota-induced type I interferons instruct a poised basal state of dendritic cells. Cell 181, 1080–1096 (2020).
Stefan, K. L., Kim, M. V., Iwasaki, A. & Kasper, D. L. Commensal microbiota modulation of natural resistance to virus infection. Cell 183, 1312–1324 (2020).
Erttmann, S. F. et al. The gut microbiota prime systemic antiviral immunity via the cGAS-STING-IFN-I axis. Immunity 55, 847–861 (2022).
Asselin-Paturel, C. et al. Type I interferon dependence of plasmacytoid dendritic cell activation and migration. J. Exp. Med. 201, 1157–1167 (2005).
Ganal, S. C. et al. Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota. Immunity 37, 171–186 (2012).
Winkler, E. S. et al. The intestinal microbiome restricts alphavirus infection and dissemination through a bile acid-type I IFN signaling axis. Cell 182, 901–918 (2020).
Barchet, W. et al. Virus-induced interferon ɑ production by a dendritic cell subset in the absence of feedback signaling in vivo. J. Exp. Med. 195, 507–516 (2002).
Kindler, E. et al. Early endonuclease-mediated evasion of RNA sensing ensures efficient coronavirus replication. PLoS Pathog. 13, e1006195 (2017).
Carrington, E. M. et al. Prosurvival Bcl-2 family members reveal a distinct apoptotic identity between conventional and plasmacytoid dendritic cells. Proc. Natl Acad. Sci. USA 112, 4044–4049 (2015).
Marsman, C. et al. Plasmacytoid dendritic cell heterogeneity is defined by CXCL10 expression following TLR7 stimulation. Immunol. Cell Biol. 96, 1083–1094 (2018).
Alculumbre, S. G. et al. Diversification of human plasmacytoid predendritic cells in response to a single stimulus. Nat. Immunol. 19, 63–75 (2018).
Weng, C. et al. Deciphering cell states and genealogies of human haematopoiesis. Nature 627, 389–398 (2024).
Yu, P. et al. Nucleic acid-sensing Toll-like receptors are essential for the control of endogenous retrovirus viremia and ERV-induced tumors. Immunity 37, 867–879 (2012).
Swiecki, M. et al. Type I interferon negatively controls plasmacytoid dendritic cell numbers in vivo. J. Exp. Med. 208, 2367–2374 (2011).
Motwani, M. et al. cGAS-STING pathway does not promote autoimmunity in murine models of SLE. Front. Immunol. 12, 605930 (2021).
Psarras, A. et al. Functionally impaired plasmacytoid dendritic cells and non-haematopoietic sources of type I interferon characterize human autoimmunity. Nat. Commun. 11, 6149 (2020).
Zuniga, E. I., Liou, L. Y., Mack, L., Mendoza, M. & Oldstone, M. B. Persistent virus infection inhibits type I interferon production by plasmacytoid dendritic cells to facilitate opportunistic infections. Cell Host Microbe 4, 374–386 (2008).
Lee, L. N., Burke, S., Montoya, M. & Borrow, P. Multiple mechanisms contribute to impairment of type 1 interferon production during chronic lymphocytic choriomeningitis virus infection of mice. J. Immunol. 182, 7178–7189 (2009).
Sisirak, V. et al. Impaired IFN-ɑ production by plasmacytoid dendritic cells favors regulatory T-cell expansion that may contribute to breast cancer progression. Cancer Res. 72, 5188–5197 (2012).
Schmitt, H. et al. Siglec-H protects from virus-triggered severe systemic autoimmunity. J. Exp. Med. 213, 1627–1644 (2016).
Bunin, A. et al. Protein tyrosine phosphatase PTPRS is an inhibitory receptor on human and murine plasmacytoid dendritic cells. Immunity 43, 277–288 (2015).
Dalod, M. et al. Dendritic cell responses to early murine cytomegalovirus infection: subset functional specialization and differential regulation by interferon ɑ/β. J. Exp. Med. 197, 885–898 (2003).
Saitoh, S. I. et al. TLR7 mediated viral recognition results in focal type I interferon secretion by dendritic cells. Nat. Commun. 8, 1592 (2017).
Kim, S. et al. Self-priming determines high type I IFN production by plasmacytoid dendritic cells. Eur. J. Immunol. 44, 807–818 (2014).
Uccellini, M. B. & Garcia-Sastre, A. ISRE-reporter mouse reveals high basal and induced type I IFN responses in inflammatory monocytes. Cell Rep. 25, 2784–2796 (2018).
Jung, S. et al. Analysis of fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol. Cell. Biol. 20, 4106–4114 (2000).
Soni, C. et al. Plasmacytoid dendritic cells and type I interferon promote extrafollicular B cell responses to extracellular self-DNA. Immunity 52, 1022–1038 (2020).
Mathian, A., Weinberg, A., Gallegos, M., Banchereau, J. & Koutouzov, S. IFN-ɑ induces early lethal lupus in preautoimmune (New Zealand Black x New Zealand White)F1 but not in BALB/c mice. J. Immunol. 174, 2499–2506 (2005).
Mach, N. et al. Differences in dendritic cells stimulated in vivo by tumors engineered to secrete granulocyte-macrophage colony-stimulating factor or Flt3-ligand. Cancer Res. 60, 3239–3246 (2000).
Turner, M. J., Jellison, E. R., Lingenheld, E. G., Puddington, L. & Lefrancois, L. Avidity maturation of memory CD8 T cells is limited by self-antigen expression. J. Exp. Med. 205, 1859–1868 (2008).
Chua, M. M., MacNamara, K. C., San Mateo, L., Shen, H. & Weiss, S. R. Effects of an epitope-specific CD8+ T-cell response on murine coronavirus central nervous system disease: protection from virus replication and antigen spread and selection of epitope escape mutants. J. Virol. 78, 1150–1159 (2004).
Becht, E. et al. High-throughput single-cell quantification of hundreds of proteins using conventional flow cytometry and machine learning. Sci. Adv. 7, eabg0505 (2021).
Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).
van Buggenum, J. A. et al. A covalent and cleavable antibody-DNA conjugation strategy for sensitive protein detection via immuno-PCR. Sci. Rep. 6, 22675 (2016).
Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).
Stoeckius, M. et al. Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).
Khodadadi-Jamayran, A. et al. iCellR: combined coverage correction and principal component alignment for batch alignment in single-cell sequencing analysis. Preprint at bioRxiv https://doi.org/10.1101/2020.03.31.019109 (2020).
McInnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at https://arxiv.org/abs/1802.03426 (2018).
Heng, T. S. et al. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).
La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).
Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).
Granot, T. et al. Dendritic cells display subset and tissue-specific maturation dynamics over human life. Immunity 46, 504–515 (2017).
Carpenter, D. J. et al. Human immunology studies using organ donors: impact of clinical variations on immune parameters in tissues and circulation. Am. J. Transpl. 18, 74–88 (2018).
Goltsev, Y. et al. Deep profiling of mouse splenic architecture with CODEX multiplexed imaging. Cell 174, 968–981 (2018).
Acknowledgements
We thank S. Hao (New York Genome Center) for help with CITE-seq, K. Khanna for VSV-GFP, I. Aifantis and D. Papaioannou for Venetoclax, D. Littman and T. Najar for OT-II mice and S. Weiss (University of Pennsylvania) for M-CoV-GFP. This study was supported by the NIH grants no. HL145997 and no. CA009161 (J.N.P.), no. AI072571 (B.R.), no. AI158808 (J.I.) and no. AI128949 (B.R., S.P.W., D.L.F.), and the German Research Foundation grant no. EI1185/1-1 (A.E.). We acknowledge the use of resources provided by NYU Genome Technology Center (GTC; RRID:SCR_017929), Applied Bioinformatics Laboratories (RRID:SCR_019178), High Performance Computing Facility (HPCF), Cytometry and Cell Sorting Laboratory (CSL; RRID:SCR_019179) and the Experimental Pathology Research Laboratory (RRID:SCR_017928), and shared resources partially supported by NIH grant no. P30CA016087 at the Laura and Isaac Perlmutter Cancer Center.
Author information
Authors and Affiliations
Contributions
J.N.P., R.A.M.-A., H.N., F.B.S., A.E., O.A.P., A.D. and J.F. performed experiments and analyzed results. J.N.P., E.E., A.C.R., A.K.-J. and I.D. developed analytical methods and analyzed results. J.Z. advised on the statistical analysis of results. M.S. and P.S. developed experimental technology. E.I., S.S., K.C., S.B.K. D.B.S., S.P.W. and D.L.F. provided research materials. C.S. provided conceptual input. B.R. and J.I. supervised the project and analyzed the results. J.N.P. and B.R. conceived the project and wrote the paper with input from all authors.
Corresponding authors
Ethics declarations
Competing interests
M.S. and P.S. are inventors on a patent related to the CITE-seq method and are or have been employed by 10x Genomics, Inc., but were not affiliated with it or any other commercial entity at the time of the work described in this paper. B.R. is an advisor for Related Sciences and a co-founder of Danger Bio. J.I. serves on the advisory board of Immunitas Therapeutics. None of these affiliations are related to the present work. The other authors declare no competing interests.
Peer review
Peer review information
Nature Immunology thanks Michael Chopin, Marc Dalod, Tsuneyasu Kaisho and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey, 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 Initial analysis of the single-cell transcriptome of splenic pDCs.
Sorted pDCs from pooled spleens of naive mice were subjected to multiome analysis by single-nuclei RNA-seq and ATAC-seq (Fig. 1), and the former was analyzed. (a) UMAP plot of cells based on mRNA transcript reads, indicating the original 9 clusters. (b) Heatmap of top 10 marker genes of each cluster (rows) across individual cells (columns). (c) Correlation heatmap of the original 9 clusters. (d) Correlation heatmap of the reduced clusters (Mt clusters in Fig. 1a).
Extended Data Fig. 2 Initial analysis of the single-cell chromatin of splenic pDCs.
Sorted pDCs from pooled spleens of naive mice were subjected to multiome analysis by single-nuclei RNA-seq and ATAC-seq (Fig. 1), and the latter was analyzed. (a) UMAP plot of cells based on ATAC-Seq reads, indicating the original 9 clusters. (b) Correlation heatmap of the original 9 clusters and after cluster reduction, with the corresponding Leiden clustering resolutions indicated. The heatmap on the right corresponds to the four Ma clusters in Fig. 1e. (c) The projection of Ma clusters onto the UMAP plot based on RNA-Seq from Fig. 1a. Shown are combined projections of all clusters (left panel) and feature plots of individual Mt clusters (right panel). (d) The fraction of Ma clusters within the total dataset and within each Mt cluster.
Extended Data Fig. 3 Additional analysis of CITE-Seq of splenic pDCs.
Sorted pDCs from spleens of three individual naive mice were hashtagged and analyzed by CITE-Seq for single-cell transcriptome and phenotype (antibody-derived tags, ADT). (a) UMAP plot and clustering of the dataset prior to the tDC cluster removal (clusters are numbered and colored randomly). (b-c) Violin plots showing transcript levels of key pDC (b) and tDC (c) identity genes. (d) Heatmap of top 10 marker gene expression based on mRNA transcripts or ADT reads for the clusters in panel A. (e) Heatmap of ADT for markers expressed in pDCs across the three Ct clusters defined in Fig. 2b. (f) RNA velocity analysis. Shown are velocity vectors projected onto the KnetL and UMAP plots from Fig. 2b, with the three Ct clusters indicated.
Extended Data Fig. 4 Verification of transcriptional heterogeneity in murine splenic pDCs.
A previously reported scRNA-seq dataset of splenic pDCs from naive mice (GSE114313) was reanalyzed to assess steady-state heterogeneity. (a) UMAP plot, with clusters from the KNetL clustering in the next panel indicated by colors. (b) KNetL plot and clustering. (c) Violin plots showing transcript levels of key pDC identity genes by KNetL clusters. (d) Heatmap of marker gene expression for KNetL clusters. (e) Feature plots of key marker gene transcripts on the KNetL plot from panel B; cluster with prominent ISG expression (cluster 2) is highlighted. (f) The expression of MHC cl II transcript shown on the feature plot (with the MHC cl. II-low cluster 5 highlighted) and violin plot.
Extended Data Fig. 5 The analysis of transcriptional heterogeneity in human pDCs.
A previously reported scRNA-seq dataset of pDCs from peripheral blood of three healthy human donors (GSE189120) was reanalyzed to assess steady-state heterogeneity. (a) KNetL and UMAP plots, with KNetL-based human transcriptome (Ht) clusters indicated. (b) Violin plots showing transcript levels of key pDC identity genes by KNetL clusters. (c) KNetL plots of Ht clusters, split up by individual donor. (d) Heatmap of marker gene expression for KNetL clusters.
Extended Data Fig. 6 Prospective identification and isolation of murine pDCs.
(a-b) Gating strategy for the analysis of pDC subsets in the spleen (panel a) and bone marrow (panel b). (c) Gating strategy for the recovery of pDC subsets in the hosts of adoptive transfer experiments in Fig. 3k-m. (d) Gating strategy for the sorting (top panel) and post-sort purity (bottom panel) of pDC subsets isolated for functional experiments in Figs. 7–8.
Extended Data Fig. 7 Modulation of pDC heterogeneity in bone marrow-derived pDCs by IFN-I blockade.
(a) Total BM cells were harvested from naïve wild-type mice and cultured in the presence of Flt3L for 12 days to induce DC differentiation. Anti-IFNAR1 mAb was added throughout the culture (Blockade), during the first 6 days (D6 washoff) or during the last 6 days (D6 blockade). (b) The dynamics of Sca1 expression on pDCs between days 6 and 12 of culture, as determined by flow cytometry. Cultures from Ifnar1−/− mice were used as controls (dashed line). Data represent averages of biological replicates (cultures from individual mice) from one experiment. (c) The fraction of Sca1+ pDCs on days 6 and 9 of culture. Bars represent means ± SD, symbols represent biological replicates (cultures from individual mice) from one experiment. Statistical significance was analyzed using one-way ANOVA followed by Tukey’s test. * p < 0.05; ** p < 0.005; *** p < 0.0005; **** p < 0.00005. Illustration in a created using BioRender.com.
Supplementary information
Supplementary Table 1
Marker genes of cell clusters in the single-nucleus RNA-seq of mouse pDCs.
Supplementary Table 2
Marker peaks of cell clusters in the single-nucleus ATAC-seq of mouse pDCs.
Supplementary Table 3
Marker genes of cell clusters in the CITE-seq of mouse pDCs.
Supplementary Table 4
Gene expression in prospectively isolated subsets of mouse pDCs.
Source data
Source Data for Figs. 1–8 and Extended Data Figs. 1–7
Statistical source data.
Source Data Fig. 2
Raw image files (JPEG) for Fig. 2g,h.
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.
About this article
Cite this article
Pucella, J.N., Maqueda-Alfaro, R.A., Ni, H. et al. Tonic type I interferon signaling optimizes the antiviral function of plasmacytoid dendritic cells. Nat Immunol (2025). https://doi.org/10.1038/s41590-025-02279-4
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41590-025-02279-4