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:

Gremlin 1+ fibroblastic niche maintains dendritic cell homeostasis in lymphoid tissues

Abstract

Fibroblastic reticular cells (FRCs) are specialized stromal cells that define tissue architecture and regulate lymphocyte compartmentalization, homeostasis, and innate and adaptive immunity in secondary lymphoid organs (SLOs). In the present study, we used single-cell RNA sequencing (scRNA-seq) of human and mouse lymph nodes (LNs) to identify a subset of T cell–zone FRCs defined by the expression of Gremlin1 (Grem1) in both species. Grem1-CreERT2 knock-in mice enabled localization, multi-omics characterization and genetic depletion of Grem1+ FRCs. Grem1+ FRCs primarily localize at T–B cell junctions of SLOs, neighboring pre-dendritic cells and conventional dendritic cells (cDCs). As such, their depletion resulted in preferential loss and decreased homeostatic proliferation and survival of resident cDCs and compromised T cell immunity. Trajectory analysis of human LN scRNA-seq data revealed expression similarities to murine FRCs, with GREM1+ cells marking the endpoint of both trajectories. These findings illuminate a new Grem1+ fibroblastic niche in LNs that functions to maintain the homeostasis of lymphoid tissue-resident cDCs.

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: ScRNA-seq identifies Grem1 as a marker for T cell zone FRCs in LNs.
Fig. 2: Grem1-expressing FRCs are localized at the interface of B and T cell zones in SLOs.
Fig. 3: Multi-omics analyses define immune hallmarks of Grem1+ FRCs.
Fig. 4: Selective ablation of Grem1+ FRCs has minimal impact on LN architecture.
Fig. 5: Grem1+ FRC ablation leads to a loss of lymphoid tissue-resident cDCs.
Fig. 6: Grem1+ FRCs cohabitate with pre-cDCs and cDCs in the parafollicular region of LNs and regulate cDC homeostasis.
Fig. 7: Characterization of Grem1+ FRC paracrine signaling events with DCs.
Fig. 8: Decreased T cell response following depletion of Grem1+ FRCs.

Similar content being viewed by others

Data availability

ScRNA-seq and bulk RNA-seq data are available in the ArrayExpress database under accession nos. E-MTAB-10197 (single-cell mouse FRCs), E-MTAB-10196 (single-cell mouse DCs), E-MTAB-10205 (bulk mouse FRCs) and E-MTAB-10206 (single-cell human DCs + FRCs). Source data are provided with this paper.

References

  1. Fletcher, A. L., Acton, S. E. & Knoblich, K. Lymph node fibroblastic reticular cells in health and disease. Nat. Rev. Immunol. 15, 350–361 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Malhotra, D., Fletcher, A. L. & Turley, S. J. Stromal and hematopoietic cells in secondary lymphoid organs: partners in immunity. Immunol. Rev. 251, 160–176 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Rodda, L. B., Bannard, O., Ludewig, B., Nagasawa, T. & Cyster, J. G. Phenotypic and morphological properties of germinal center dark zone Cxcl12-expressing reticular cells. J. Immunol. 195, 4781–4791 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Cremasco, V. et al. B cell homeostasis and follicle confines are governed by fibroblastic reticular cells. Nat. Immunol. 15, 973–981 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Link, A. et al. Fibroblastic reticular cells in lymph nodes regulate the homeostasis of naive T cells. Nat. Immunol. 8, 1255–1265 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Rodda, L. B. et al. Single-cell RNA sequencing of lymph node stromal cells reveals niche-associated heterogeneity. Immunity 48, 1014–1028.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Denton, A. E., Roberts, E. W., Linterman, M. A. & Fearon, D. T. Fibroblastic reticular cells of the lymph node are required for retention of resting but not activated CD8+ T cells. Proc. Natl Acad. Sci. USA 111, 12139–12144 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Merad, M., Sathe, P., Helft, J., Miller, J. & Mortha, A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev. Immunol. 31, 563–604 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Mildner, A. & Jung, S. Development and function of dendritic cell subsets. Immunity 40, 642–656 (2014).

    Article  CAS  PubMed  Google Scholar 

  10. Girard, J.-P., Moussion, C. & Förster, R. HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nat. Rev. Immunol. 12, 762–773 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. 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 

  12. Kumar, V. et al. A dendritic-cell–stromal axis maintains immune responses in lymph nodes. Immunity 42, 719–730 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Perez-Shibayama, C. et al. Type I interferon signaling in fibroblastic reticular cells prevents exhaustive activation of antiviral CD8+ T cells. Sci. Immunol. 5, eabb7066 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Bénazet, J.-D. et al. A self-regulatory system of interlinked signaling feedback loops controls mouse limb patterning. Science 323, 1050–1053 (2009).

    Article  PubMed  CAS  Google Scholar 

  15. Pezoldt, J. et al. Neonatally imprinted stromal cell subsets induce tolerogenic dendritic cells in mesenteric lymph nodes. Nat. Commun. 9, 3903 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Martinez, V. G. et al. Fibroblastic reticular cells control conduit matrix deposition during lymph node expansion. Cell Rep. 29, 2810–2822.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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 

  18. Cheng, H.-W. et al. Origin and differentiation trajectories of fibroblastic reticular cells in the splenic white pulp. Nat. Commun. 10, 1739 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Worthley, D. L. et al. Gremlin 1 identifies a skeletal stem cell with bone, cartilage, and reticular stromal potential. Cell 160, 269–284 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. McCarthy, N. et al. Distinct mesenchymal cell populations generate the essential intestinal BMP signaling gradient. Cell Stem Cell 26, 391–402.e5 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Brazil, D. P., Church, R. H., Surae, S., Godson, C. & Martin, F. BMP signalling: agony and antagony in the family. Trends Cell Biol. 25, 249–264 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Cyster, J. G., Dang, E. V., Reboldi, A. & Yi, T. 25-Hydroxycholesterols in innate and adaptive immunity. Nat. Rev. Immunol. 14, 731–743 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Li, J., Lu, E., Yi, T. & Cyster, J. G. EBI2 augments Tfh cell fate by promoting interaction with IL-2-quenching dendritic cells. Nature 533, 110–114 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lu, E., Dang, E. V., McDonald, J. G. & Cyster, J. G. Distinct oxysterol requirements for positioning naïve and activated dendritic cells in the spleen. Sci. Immunol. 2, eaal5237 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Baptista, A. P. et al. The chemoattractant receptor Ebi2 drives intranodal naive CD4+ T cell peripheralization to promote effective adaptive. Immunity 50, 1188–1201.e6 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. Gatto, D. et al. The chemotactic receptor EBI2 regulates the homeostasis, localization and immunological function of splenic dendritic cells. Nat. Immunol. 14, 446–453 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Yi, T. & Cyster, J. G. EBI2-mediated bridging channel positioning supports splenic dendritic cell homeostasis and particulate antigen capture. Elife 2, e00757 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Ghaleb, A. M. & Yang, V. W. Krüppel-like factor 4 (KLF4): what we currently know. Gene 611, 27–37 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hagos, E. G., Ghaleb, A. M., Kumar, A., Neish, A. S. & Yang, V. W. Expression profiling and pathway analysis of Krüppel-like factor 4 in mouse embryonic fibroblasts. Am. J. Cancer Res. 1, 85–97 (2011).

    CAS  PubMed  Google Scholar 

  30. Dieu, M.-C. et al. Selective recruitment of immature and mature dendritic cells by distinct chemokines expressed in different anatomic sites. J. Exp. Med. 188, 373–386 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sallusto, F. et al. Rapid and coordinated switch in chemokine receptor expression during dendritic cell maturation. Eur. J. Immunol. 28, 2760–2769 (1998).

    Article  CAS  PubMed  Google Scholar 

  32. Alvarez, D., Vollmann, E. H. & von Andrian, U. H. Mechanisms and consequences of dendritic cell migration. Immunity 29, 325–342 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Buch, T. et al. A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin administration. Nat. Methods 2, 419–426 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Cordeiro, O. G. et al. Integrin-alpha IIb identifies murine lymph node lymphatic endothelial cells responsive to RANKL. PLoS ONE 11, e0151848 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Gerner, M. Y., Kastenmuller, W., Ifrim, I., Kabat, J. & Germain, R. N. Histo-cytometry: a method for highly multiplex quantitative tissue imaging analysis applied to dendritic cell subset microanatomy in lymph nodes. Immunity 37, 364–376 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Liu, K. et al. In vivo analysis of dendritic cell development and homeostasis. Science 324, 392–397 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334.e10 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Miller, J. C. et al. Deciphering the transcriptional network of the dendritic cell lineage. Nat. Immunol. 13, 888–899 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Malhotra, D. et al. Transcriptional profiling of stroma from inflamed and resting lymph nodes defines immunological hallmarks. Nat. Immunol. 13, 499–510 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Severino, P. et al. Human lymph node-derived fibroblastic and double-negative reticular cells alter their chemokines and cytokines expression profile following inflammatory stimuli. Front. Immunol. 8, 141 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Bonifaz, L. et al. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance. J. Exp. Med. 196, 1627–1638 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Laurila, R., Parkkila, S., Isola, J., Kallioniemi, A. & Alarmo, E.-L. The expression patterns of gremlin 1 and noggin in normal adult and tumor tissues. Int J. Clin. Exp. Pathol. 6, 1400–1408 (2013).

    PubMed  PubMed Central  Google Scholar 

  44. Braun, A. et al. Afferent lymph-derived T cells and DCs use different chemokine receptor CCR7-dependent routes for entry into the lymph node and intranodal migration. Nat. Immunol. 12, 879–887 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Förster, R. et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell 99, 23–33 (1999).

    Article  PubMed  Google Scholar 

  46. Ulvmar, M. H. et al. The atypical chemokine receptor CCRL1 shapes functional CCL21 gradients in lymph nodes. Nat. Immunol. 15, 623–630 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Vaahtomeri, K. et al. Locally triggered release of the chemokine CCL21 promotes dendritic cell transmigration across lymphatic endothelia. Cell Rep. 19, 902–909 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Moussion, C. & Girard, J.-P. Dendritic cells control lymphocyte entry to lymph nodes through high endothelial venules. Nature 479, 542–546 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Mionnet, C. et al. Identification of a new stromal cell type involved in the regulation of inflamed B cell follicles. PLoS Biol. 11, e1001672 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Chai, Q. et al. Maturation of lymph node fibroblastic reticular cells from myofibroblastic precursors is critical for antiviral immunity. Immunity 38, 1013–1024 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Smithies, O., Gregg, R. G., Boggs, S. S., Koralewski, M. A. & Kucherlapati, R. S. Insertion of DNA sequences into the human chromosomal β-globin locus by homologous recombination. Nature 317, 230–234 (1985).

    Article  CAS  PubMed  Google Scholar 

  52. Thomas, K. R., Folger, K. R. & Capecchi, M. R. High frequency targeting of genes to specific sites in the mammalian genome. Cell 44, 419–428 (1986).

    Article  CAS  PubMed  Google Scholar 

  53. Newman, R. J., Roose-Girma, M. & Warming, S. Efficient conditional knockout targeting vector construction using co-selection BAC recombineering (CoSBR). Nucleic Acids Res. 43, e124 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Gertsenstein, M. et al. Efficient generation of germ line transmitting chimeras from C57BL/6N ES cells by aggregation with outbred host embryos. PLoS ONE 5, e11260 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Hughes, E. D. & Saunders, T. L. Gene targeting in embryonic stem cells. In Advanced Protocols for Animal Transgenesis, An ISTT Manual (eds Pease, S. and Saunders, T. L.) 291–325 (Springer-Verlag, 2011); https://doi.org/10.1007/978-3-642-20792-1_14

  56. Fletcher, A. L. et al. Reproducible isolation of lymph node stromal cells reveals site-dependent differences in fibroblastic reticular cells. Front. Immunol. 2, 35 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genom. 19, 477 (2018).

    Article  CAS  Google Scholar 

  60. Gao, X., Hu, D., Gogol, M. & Li, H. ClusterMap: compare multiple single cell RNA-Seq datasets across different experimental conditions. Bioinformatics 35, 3038–3045 (2019).

    Article  CAS  PubMed  Google Scholar 

  61. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Cabello-Aguilar, S. et al. SingleCellSignalR: inference of intercellular networks from single-cell transcriptomics. Nucleic Acids Res. 48, e55 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Shao, X. et al. CellTalkDB: a manually curated database of ligand–receptor interactions in humans and mice. Brief Bioinform. https://doi.org/10.1093/bib/bbaa269 (2020).

  64. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kwon, A. T., Arenillas, D. J., Hunt, R. W. & Wasserman, W. W. oPOSSUM-3: advanced analysis of regulatory motif over-representation across genes or ChIP-seq datasets. G3 Genes Genomes Genet. 2, 987–1002 (2012).

    CAS  Google Scholar 

  68. Denton, A. E., Roberts, E. W., Linterman, M. A. & Fearon, D. T. Fibroblastic reticular cells of the lymph node are required for retention of resting but not activated CD8+ T cells. Proc. Natl Acad. Sci. USA 111, 12139–12144 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Brown, M. et al. Lymph node blood vessels provide exit routes for metastatic tumor cell dissemination in mice. Science 359, 1408–1411 (2018).

    Article  CAS  PubMed  Google Scholar 

  70. Bonifaz, L. et al. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance. J. Exp. Med. 196, 1627–1638 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank G. Ortiz-Munoz at Genentech for technical assistance with in vivo studies. We thank S. Carlisle at Genentech for assistance with bulk RNA-seq data analysis. We thank members of the Turley lab for helpful discussions. This work was supported by Genentech.

Author information

Authors and Affiliations

Authors

Contributions

V.N.K. designed and performed experiments, analyzed and interpreted the data, and wrote the manuscript. S.M. analyzed and interpreted the bioinformatics data and wrote the manuscript. C.C. performed imaging analysis. M.B., S.K. and E.E.S. performed experiments and analyzed the data. Y.S. and A.W.W. analyzed bioinformatics data. C.B.C. and R.L. analyzed imaging data. A.T.K, C.X.D, J.L.A., A.C., M.N., X.W., J.D., M.Z.C., Z.M. and Y.A.Y. performed a subset of experiments. M.R.G. and L.T. generated a mouse model. V.C., W.P.L., R.B., W.S., A.S.S., F.J.D.S. and I.M. provided input for the experimental design. C.M. designed and performed experiments and interpreted the data. S.J.T. oversaw the project, generated the mouse model, interpreted the results and wrote the manuscript.

Corresponding author

Correspondence to Shannon J. Turley.

Ethics declarations

Competing interests

All of the authors on the manuscript are employees at Genentech and are stock holders of Genentech/Roche. V.C. worked on this project at Dana–Farber Cancer Institute and declares no competing interest.

Additional information

Peer review information Nature Immunology thanks Antonio P. Baptista, Alison Simmons and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. L. A. Dempsey was the primary editor 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 scRNA-seq analysis of CD45 cells in lymph nodes.

a, UMAP plot of 5,470 stromal cells (dots) from lymph nodes of 10 mice colored by cluster (top) and split by replicate (bottom). b, Left: Projection as in a) colored by expression of indicated genes. Right: Heatmap of the 50 most strongly upregulated genes (rows) in cells from each of the 3 main cell populations (columns): Blood endothelial cells [BECs], lymph node endothelial cells [LECs] and fibroblasts. c, Left: Heatmap of the 10 most strongly upregulated genes (rows) for each LEC subpopulation compared to all other LEC cells (columns). Right: Heatmap of the 10 most cluster-specific genes for each BEC subpopulation compared to all other BECs. d, UMAP plot of fibroblastic cells from Fig. 1a colored by the expression of indicated marker genes. e, Heatmap as described in (c), here of the 10 most cluster-specific genes for each Cd34+ fibroblast subpopulation from Fig. 1a compared to all other Cd34+ fibroblasts. f, Heatmap as described in (c), here of the most strongly enriched genes for fibroblast subpopulations from Fig. 1a. For all heatmaps most strongly enriched genes were defined as the genes with largest LogFC compared to all other cells in the dataset; at most 10 genes are shown per cluster, but less if one or more genes were not uniquely enriched in just the cluster under consideration.

Extended Data Fig. 2 Gremlin1 expressing FRCs are localized outside the B-cell follicle in Peyer’s patches.

a, Fraction of SLN Grem1+ cells in FRCs from Fig. 1a for each replicate separately. b, Left: Gating strategy for identification of FDC (follicular dendritic), MRC (marginal reticular) and FRC (fibroblastic reticular) cells within the CD45Ter119CD31PDPN+ gate; Middle: relative proportion; Right: absolute numbers of EYFP+ (Grem1+) in FRC, MRC and FDC subsets from Grem1 Cre.YFP mice (n = 4) post-tamoxifen induction. c, Top Left: UMAP dimensionality reduction of stromal cells isolated from mouse spleen, colored by cluster membership. Right/bottom left: UMAP as on top left, colored by indicated marker gene expression. Bottom right: Fraction of Grem1+ fibroblasts shown on the top left. d, EYFP in CD45 stromal cells in bone marrow and bones of Grem1 Cre.YFP mice (n = 3) post-tamoxifen induction. e, Plots showing in-vitro EYFP expression by lymph node FRCs in control and 4-hydroxytamoxifen treated cultures (4-OHT) from Grem1 Cre.YFP mice. (f, g) Imaging of lymph node sections from Grem1 Cre.YFP mice post-tamoxifen induction. f, Anti-YFP (Grem1+ FRCs), anti-PDPN (stromal cells) staining in SLN. Scale bars, 150μm. g, Anti-YFP (Grem1+ FRCs), anti-PDPN (stromal cells), anti-PNAd (HEV), anti-LYVE-1 (lymphatic, LEC) staining in SLN. Scale bars, 150μm. h, Imaging of spleen sections from Grem1 Cre.YFP mice post-tamoxifen induction; stained for anti-B220 and anti-YFP (left). Scale bars, 150μm; (right) enlargement of sections on the left. Scale bars, 75μm. i, Imaging of Peyer’s patches from Grem1 Cre.YFP mice post-tamoxifen induction, stained for anti-YFP (Grem1+ FRCs), anti-PDPN (stromal cells), anti-B220 (B cells). Scale bars, 150μm; zoomed image on right, scale bar, 75μm. Data representative of two independent experiments (b, d); data pooled from three independent experiments (e) [control n = 3; OHT treated n = 6]; data representative of images from 13 SLN, three independent experiments (f, g); representative image from 5 spleens, two independent experiments (h); representative images from 4 Peyer’s patches, two independent experiments (i). Data shown as mean ± SEM.

Source data

Extended Data Fig. 3 RNA-seq reveals differences in chemokine activity and integrin mediated signaling between Grem1+ and Grem1 FRCs.

a, Top: UMAP as in Fig. 1a), here colored by Cd200 expression. Bottom: Violin plot showing the expression of Cd200 in Grem1+ clusters 1 and 3 from Fig. 1a. b, Representative plots showing CD200 expression on EYFP+ (Grem1+) FRCs (FRC: CD45Ter119CD31Madcam1CD21/35 PDPN+) in skin draining lymph nodes (SLN) of Grem1-Cre-ERT2 Rosa26 EYFP mice (n = 4) post-tamoxifen induction c, Left: Representative plots showing CD90 and CD26 expression on EYFP (Grem1) CD34+ FRCs in SLN of Grem1-Cre-ERT2 Rosa26 EYFP mice (n = 4) at day 25 post-tamoxifen induction. Right: UMAP as in Fig. 1a, here colored by Cd34, Dpp4, and Thy1 expression. d, Relative average expression of 25 of the most up – and downregulated genes in bulk RNA-seq samples of Grem1+ and Grem1 FRCs. e, Heatmap of expression of marker genes (columns) determined via scRNA-seq [MAST, adj. p-val <0.05 in both replicates comparing Grem1+ FRCs to all other fibroblastic cells] in bulk RNA-seq samples of Grem1+ and Grem1 fibroblasts (rows). f, Gene set enrichment analysis comparing the distribution of the log2FC of Grem1+ vs Grem1 cells across all genes (blue) to the distribution of genes in indicated categories (orange); fry with p-value adjustment. g, Heatmap of genes (rows) from indicated gene ontology (GO) categories with significant (voom/limma, adj. p-val <0.05) differences in gene expression between Grem1+ and Grem1 bulk RNA-seq libraries (columns). h, Enrichment of transcription factor binding sites in genes with significant (MAST, adj. p-val <0.05) upregulation in Grem1+ FRCs compared to Grem1 FRCs in scRNA-seq. i, Volcano plot visualizing the fold-change (x-axis, log2 transformed) and adjusted p-value (y-axis, Spectronaut adjusted p-val <0.001) for proteins (dots) between Grem1+ and Grem1 cells. j, Comparison of indicated proteins from proteomic analysis between Grem1+ and Grem1 FRCs. Data are representative of two independent experiments (b, c); data are compilation of proteomic analysis from three independent sorts (j). ***P < 0.001, **P < 0.01, *P < 0.05 (two-tailed unpaired Student’s t-test). Exact P-values in extended source data. Data are shown as mean ± SEM.

Source data

Extended Data Fig. 4 Normal lymph node HEV morphology and numbers upon Grem1+ FRC ablation.

a, Representative plots with relative proportion (top) and absolute numbers (bottom) of CD45 EYFP+ stromal cells in Grem1-Cre-ERT2 Rosa26 EYFP (n = 5) and Rosa26 EYFP/iDTR (n = 5) mice upon DTxn mediated ablation. Plots are gated on CD45 stromal cells. b, Spleen weight (left) and cellularity (right) in Grem1-Cre-ERT2 Rosa26 EYFP (n = 12) and Rosa26 EYFP/iDTR (n = 14) mice upon DTxn mediated ablation. c, Immuno-fluorescence imaging of lymph node sections from Grem1-Cre-ERT2 Rosa26 EYFP (top) and Rosa26 EYFP/iDTR mice (bottom) upon DTxn mediated ablation, sections were stained for anti-LYVE-1(lymphatics, LECs) and anti-PNAd (HEVs). Scale bars, 150μm. d, Number of HEVs per lymph node as identified by PNAd staining as in (c), here comparing Grem1-Cre-ERT2 Rosa26 EYFP and Rosa26 EYFP/iDTR mice upon DTxn mediated ablation. e, Images of lymph node sections from Grem1-Cre-ERT2 Rosa26 EYFP (left) and Rosa26 eYFP/iDTR mice (right) upon DTxn mediated ablation, sections were stained for anti-PDPN (stromal cells) and anti-PNAd (HEVs). Scale bars, 25μm. f, Immuno-fluorescence imaging of lymph node sections from Grem1-Cre-ERT2 Rosa26 EYFP (left) and Rosa26 EYFP/iDTR mice (right) upon DTxn mediated ablation, sections were stained for anti-PDPN (stromal cells). Scale bars, 75μm. g, Relative proportion of ITGA2b + LEC in Grem1-Cre-ERT2 Rosa26 EYFP (n = 10) and Rosa26 EYFP/iDTR (n = 9) mice upon DTxn mediated ablation. Plots are gated on LEC: CD45 Ter119 CD31+ PDPN+. Data are representative of three independent experiments (a); pooled data from three independent experiments (b); Representative image and quantitation of ten lymph nodes from Grem1-Cre-ERT2 Rosa26 EYFP and fourteen lymph nodes from Rosa26 EYFP/iDTR mice from three independent experiments (c, d, f); representative image of seven lymph nodes from three independent experiments (e); pooled data from two independent experiments (g). ****P < 0.0001, **P < 0.01 (two-tailed unpaired Student’s t-test). Exact P-values in extended source data. Data are shown as mean ± SEM.

Source data

Extended Data Fig. 5 Comparison of lymphoid cellularity upon Grem1+ FRC ablation.

a, Relative proportion (top row) and absolute numbers (bottom row) of indicated cell subsets in lymph nodes of Grem1-Cre-ERT2 Rosa26 EYFP (n = 12) and Rosa26 EYFP/iDTR (n = 14) mice upon DTxn mediated ablation. b, Representative plot with relative proportion of EYFP (Grem1) expression on CD11chi MHCIIinter cDCs in lymph nodes of Grem1-Cre-ERT2 Rosa26 EYFP (n = 5) mice. c, Relative proportion (top row) and absolute numbers (bottom row) of CD8a+ (left column) or CD11b+ CD4+ (right column) cDCs in spleens of Grem1-Cre-ERT2 Rosa26 EYFP (n = 12) and Rosa26 EYFP/iDTR mice (n = 14) upon DTxn mediated ablation d, Total cell numbers (top, left) and relative proportions of B cells (top, right), CD4+ T (bottom, left) and CD8+ T (bottom, right) in lymph nodes of Grem1-Cre-ERT2 Rosa26 EYFP (n = 7) and Rosa26 EYFP/iDTR (n = 9) mice at day 5 post-DTxn mediated ablation. Data are pooled from three independent experiments (a, c); representative of three independent experiments (b); data are pooled from two independent experiments (d). ****P < 0.0001, ***P < 0.001, *P < 0.05 (two-tailed unpaired Student’s t-test). Exact P-values in extended source data. Data are shown as mean ± SEM.

Source data

Extended Data Fig. 6 T cell and pre-cDC trafficking to secondary lymphoid organs is not affected upon Grem1+ FRC ablation.

a, High resolution image of lymph nodes from Grem1-Cre-ERT2 Rosa26 EYFP mice, panels were stained for anti-YFP, anti-CD11c and anti-IgD to detect Grem1+ FRC, CD11c+ and B cells respectively at the B cell follicle boundary. Scale bars, 25μm. b, Confocal imaging of lymph nodes from Grem1-Cre-ERT2 Rosa26 EYFP (left) and Rosa26 EYFP/iDTR (right) mice upon DTxn mediated ablation, panels were stained for anti-YFP, anti-DEC205 and anti-CD11b to detect Grem1+ FRC, cDC1 and cDC2 cells respectively. Scale bars, 125μm. c, Top: schematic of T cell transfer and trafficking experiment; relative proportion of donor T cells two hours after adoptive transfer in lymph nodes (bottom, left) and spleen (bottom, right) of Grem1-Cre-ERT2 Rosa26 EYFP (n = 9) and Rosa26 EYFP/iDTR mice (n = 10 for LN, n = 9 for SPL). d, Top: schematic of pre-cDC transfer and trafficking experiment; relative proportion of donor pre-cDCs twenty hours after adoptive transfer in lymph nodes (bottom, left) and spleen (bottom, right) of Grem1-Cre-ERT2 Rosa26 EYFP (n = 8) and Rosa26 EYFP/iDTR mice (n = 8). e, Relative proportion of BrdU incorporation (19 hours pulse) by CD8a + cDC1s (top) and CD4 + CD11b + cDC2s (bottom) in lymph nodes of Grem1-Cre-ERT2 Rosa26 EYFP (n = 12) and Rosa26 EYFP/iDTR (n = 13) at day 7 after DTxn mediated ablation. Data are representative of images of six lymph nodes of Grem1-Cre-ERT2 Rosa26 EYFP mice from two independent experiments (a); representative of image from thirteen lymph nodes from Grem1-Cre-ERT2 Rosa26 EYFP and eight lymph nodes from Rosa26 EYFP/iDTR mice from three independent experiments (b); data are pooled from two independent experiments (c, d); data are pooled from three independent experiments (e). ****P < 0.0001, *P < 0.05 (two-tailed unpaired Student’s t-test). Exact P-values in extended source data. Data are shown as mean ± SEM.

Source data

Extended Data Fig. 7 Interactions between Grem1+ FRCs and DCs.

a, Left: Uniform Manifold Approximation and Projection (UMAP) plot visualization of 12,620 murine dendritic cell sorts (dots) colored by cluster identity. Right: UMAP as on the left, here colored by expression of indicated marker genes. b, Average relative expression of the 10 most strongly upregulated genes (by LogFC, rows) across clusters from (a). Two representative genes per cluster are highlighted. c, Fold change in expression of Flt3l mRNA in indicated cell types from skin draining lymph nodes of Grem1-Cre-ERT2 Rosa26 EYFP/iDTR mice with respect to Grem1-Cre-ERT2 Rosa26 EYFP mice upon DTxn mediated ablation, results are normalized to those of the gene encoding Rpl19 (ribosomal protein L19). d, Quantification of FLT3L by ELISA of total lymph node protein lysates from skin draining lymph nodes of Grem1-Cre-ERT2 Rosa26 EYFP and Rosa26 EYFP/iDTR mice upon DTxn mediated ablation. e, Left: UMAP plot visualizing 16,226 human CD11c + cells form lymph nodes of three patients colored by cluster identity. Middle: UMAP as on the left, here colored by expression of marker genes. Right: Average relative expression of the 10 most strongly upregulated genes (by LogFC, rows) in clusters from the left. f, Percentage of cells from each patient in each of the clusters from Fig. 7f. g, Enrichment analysis of ligands expressed by GREM1+ FRCs predicted to be involved in paracrine signaling events with DC1. (c) data are pooled from three independent experiments; (d) data are pooled from three independent experiments (YFP n = 14; YFP/iDTR n = 13). **P < 0.01 (two-tailed unpaired Student’s t-test). Exact P-values in extended source data. Data are shown as mean ± SEM.

Source data

Extended Data Fig. 8 Decreased CD4/CD8 T cell responses upon Grem1+ FRC depletion.

a, schematic of anti-DEC205OVA immunization protocol. b, relative proportion (top row) and absolute numbers (bottom row) of IFNg+ OT-I T cells in lymph nodes (left column) and spleen (right column) of Grem1-Cre-ERT2 Rosa26 EYFP (n = 5) and Rosa26 EYFP/iDTR (n = 5) at day 8 after DTxn mediated ablation. c, schematic of CFA/OVA immunization protocol. d, relative proportion (top) and absolute numbers (bottom) of transferred Thy1.1 OT-I T cells (left) or transferred labeled OT-II T cells (right) in spleen of Grem1-Cre-ERT2 Rosa26 EYFP (n = 5) and Rosa26 EYFP/iDTR (n = 5) at day 7 after DTxn mediated ablation. Data are representative of two independent experiments (b, d). **P < 0.01, *P < 0.05 (two-tailed unpaired Student’s t-test). Exact P-values in extended source data. Data are shown as mean ± SEM.

Source data

Supplementary information

Supplementary data

Gating scheme.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–11.

Supplementary Video

Supplementary Video 1.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

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. 2

Statistical source data.

Source Data Extended Data Fig. 3

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. 7

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

Kapoor, V.N., Müller, S., Keerthivasan, S. et al. Gremlin 1+ fibroblastic niche maintains dendritic cell homeostasis in lymphoid tissues. Nat Immunol 22, 571–585 (2021). https://doi.org/10.1038/s41590-021-00920-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00920-6

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